1
|
Alcindor T, Tankel J, Fiset PO, Pal S, Opu T, Strasser M, Dehghani M, Bertos N, Zuo D, Mueller C, Cools-Lartigue J, Hickeson M, Marcus V, Camilleri-Broet S, Spatz A, Evaristo G, Farag M, Artho G, Elkrief A, Saleh R, Bailey S, Park M, Huang S, Sangwan V, Ferri L. Phase 2 trial of perioperative chemo-immunotherapy for gastro-esophageal adenocarcinoma: The role of M2 macrophage landscape in predicting response. Cell Rep Med 2025; 6:102045. [PMID: 40239627 PMCID: PMC12047487 DOI: 10.1016/j.xcrm.2025.102045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/28/2024] [Accepted: 03/06/2025] [Indexed: 04/18/2025]
Abstract
We present the clinical results of a phase 2 trial combining neoadjuvant docetaxel, cisplatin, 5 Flourouracil, and the PD-L1 inhibitor avelumab in locally advanced gastro-esophageal adenocarcinoma (GEA). Fifty-one patients receive neoadjuvant therapy with 50 proceeding to surgery. Grade 3-4 adverse events occur in 40%; complete/major pathological response is found in 7/50 (14%) and 9/50 (18%), with 2-year disease-free survival of 67.5%. There is no correlation between tumor regression and PD-L1 or mismatch repair (MMR) status. Multiplex immunohistochemistry and longitudinal single-cell transcriptomic profiling reveal alterations in certain innate immune cell populations, particularly noting an M2-tumor-associated macrophage (M2-TAM) proliferation in non-responding tumors. These findings describe the effective nature of this treatment regimen for GEA and reveal associated features of the inflammatory milieux associated with response to chemo-immunotherapy. The specific character of the inflammatory environment in non-responders may, in the future, help personalize treatment. This study was registered at ClinicalTrials.gov (NCT03288350).
Collapse
Affiliation(s)
- Thierry Alcindor
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada; Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada.
| | - James Tankel
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Pierre-Olivier Fiset
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Sanjima Pal
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Touhid Opu
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | | | - Mehrnoush Dehghani
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Nicholas Bertos
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Dongmei Zuo
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Carmen Mueller
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | | | - Marc Hickeson
- Department of Nuclear Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Victoria Marcus
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Sophie Camilleri-Broet
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Alan Spatz
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Gertruda Evaristo
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Mina Farag
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Giovanni Artho
- Department of Diagnostic Radiology, McGill University Health Centre, Montreal, QC, Canada
| | - Arielle Elkrief
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Ramy Saleh
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada; Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Swneke Bailey
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Morag Park
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA, USA
| | - Veena Sangwan
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Lorenzo Ferri
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
2
|
Karlsen W, Akily L, Mierzejewska M, Teodorczyk J, Bandura A, Zaucha R, Cytawa W. Is 18F-FDG-PET/CT an Optimal Imaging Modality for Detecting Immune-Related Adverse Events after Immune-Checkpoint Inhibitor Therapy? Pros and Cons. Cancers (Basel) 2024; 16:1990. [PMID: 38893111 PMCID: PMC11171385 DOI: 10.3390/cancers16111990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
Immunotherapy with immune checkpoint inhibitors (ICIs) has revolutionized contemporary oncology, presenting efficacy in various solid tumors and lymphomas. However, ICIs may potentially overstimulate the immune system, leading to immune-related adverse events (irAEs). IrAEs may affect multiple organs, such as the colon, stomach, small intestine, kidneys, skin, lungs, joints, liver, lymph nodes, bone marrow, brain, heart, and endocrine glands (e.g., pancreas, thyroid, or adrenal glands), exhibiting autoimmune inflammation. 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) is commonly used in oncology for staging and assessment of therapy responses, but it may also serve as a tool for detecting irAEs. This review aims to present various patterns of metabolic activation associated with irAEs due to ICI treatment, identifiable through 18F-FDG PET/CT. It describes the advantages of early detection of irAEs, but also presents the challenges in differentiating them from tumor progression. It also delves into aspects of molecular response assessment within the context of pseudoprogression and hyperprogression, along with typical imaging findings related to these phenomena. Lastly, it summarizes the role of functional PET imaging in oncological immunotherapy, speculating on its future significance and limitations.
Collapse
Affiliation(s)
- William Karlsen
- Students’ Scientific Circle Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (W.K.); (L.A.)
| | - Lin Akily
- Students’ Scientific Circle Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (W.K.); (L.A.)
| | - Monika Mierzejewska
- Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (M.M.); (J.T.)
| | - Jacek Teodorczyk
- Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (M.M.); (J.T.)
| | - Artur Bandura
- Department of Clinical Oncology and Radiotherapy, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (A.B.); (R.Z.)
| | - Renata Zaucha
- Department of Clinical Oncology and Radiotherapy, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (A.B.); (R.Z.)
| | - Wojciech Cytawa
- Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (M.M.); (J.T.)
| |
Collapse
|
3
|
Jha AK, Mithun S, Sherkhane UB, Dwivedi P, Puts S, Osong B, Traverso A, Purandare N, Wee L, Rangarajan V, Dekker A. Emerging role of quantitative imaging (radiomics) and artificial intelligence in precision oncology. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:569-582. [PMID: 37720353 PMCID: PMC10501896 DOI: 10.37349/etat.2023.00153] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/20/2023] [Indexed: 09/19/2023] Open
Abstract
Cancer is a fatal disease and the second most cause of death worldwide. Treatment of cancer is a complex process and requires a multi-modality-based approach. Cancer detection and treatment starts with screening/diagnosis and continues till the patient is alive. Screening/diagnosis of the disease is the beginning of cancer management and continued with the staging of the disease, planning and delivery of treatment, treatment monitoring, and ongoing monitoring and follow-up. Imaging plays an important role in all stages of cancer management. Conventional oncology practice considers that all patients are similar in a disease type, whereas biomarkers subgroup the patients in a disease type which leads to the development of precision oncology. The utilization of the radiomic process has facilitated the advancement of diverse imaging biomarkers that find application in precision oncology. The role of imaging biomarkers and artificial intelligence (AI) in oncology has been investigated by many researchers in the past. The existing literature is suggestive of the increasing role of imaging biomarkers and AI in oncology. However, the stability of radiomic features has also been questioned. The radiomic community has recognized that the instability of radiomic features poses a danger to the global generalization of radiomic-based prediction models. In order to establish radiomic-based imaging biomarkers in oncology, the robustness of radiomic features needs to be established on a priority basis. This is because radiomic models developed in one institution frequently perform poorly in other institutions, most likely due to radiomic feature instability. To generalize radiomic-based prediction models in oncology, a number of initiatives, including Quantitative Imaging Network (QIN), Quantitative Imaging Biomarkers Alliance (QIBA), and Image Biomarker Standardisation Initiative (IBSI), have been launched to stabilize the radiomic features.
Collapse
Affiliation(s)
- Ashish Kumar Jha
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Sneha Mithun
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Umeshkumar B. Sherkhane
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
| | - Pooj Dwivedi
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
- Department of Nuclear Medicine, Advance Center for Treatment, Research, Education in Cancer, Kharghar, Navi-Mumbai 410210, Maharashtra, India
| | - Senders Puts
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| | - Biche Osong
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| | - Alberto Traverso
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| | - Nilendu Purandare
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine, Tata Memorial Hospital, Mumbai 400012, Maharashtra, India
- Homi Bhabha National Institute, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, 6200 Maastricht, The Netherlands
| |
Collapse
|
4
|
Change in Density Not Size of Esophageal Adenocarcinoma During Neoadjuvant Chemotherapy Is Associated with Improved Survival Outcomes. J Gastrointest Surg 2022; 26:2417-2425. [PMID: 36214951 DOI: 10.1007/s11605-022-05422-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/16/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Changes in the size and density of esophageal malignancy during neoadjuvant chemotherapy (NCT) may be useful in predicting overall survival (OS). The aim of this study was to explore this relationship in patients with adenocarcinoma. METHODS A retrospective single-centre cohort study was performed. Consecutive patients with esophageal adenocarcinoma who received NCT followed by en bloc resection with curative intent were identified. Pre- and post-NCT computed tomography scans were reviewed. The percentage difference between the greatest tumor diameter, esophageal wall thickness and tumor density was calculated. Multivariate Cox regression analysis identified variables independently associated with OS. A ROC analysis was performed on radiological markers to identify optimal cut-off points with Kaplan-Meier plots subsequently created. RESULTS Of the 167 identified, 88 (51.5%) had disease of the gastro-esophageal junction and 149 (89.2%) were clinical T3. In total, 122 (73.1%) had node-positive disease. Increased tumor density (HR 1.01 per % change, 95% CI 1.00-1.02, p = 0.007), lymphovascular invasion (HR 3.23, 95% CI 1.34-7.52, p = 0.006) and perineural invasion (HR 2.51, 95% CI 1.03-6.08, p = 0.048) were independently associated with a decrease in OS. Patients who had a decrease in their tumor density during the time they received NCT of ≥ 20% in Hounsfield units had significantly longer OS than those who did not (75.5 months versus 34.4 months, 95% CI 38.83-105.13/18.63-35.07, p = 0.025). CONCLUSIONS Interval changes in the density, not size, of esophageal adenocarcinoma during the time that NCT are independently associated with OS.
Collapse
|
5
|
Tustumi F, Albenda DG, Perrotta FS, Sallum RAA, Ribeiro Junior U, Buchpiguel CA, Duarte PS. Prognostic Value of Bone Marrow Uptake Using 18F-FDG PET/CT Scans in Solid Neoplasms. J Imaging 2022; 8:297. [PMID: 36354870 PMCID: PMC9692285 DOI: 10.3390/jimaging8110297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/09/2022] [Accepted: 10/27/2022] [Indexed: 08/30/2023] Open
Abstract
BACKGROUND Fluorine-18-fluorodeoxyglucose positron emission tomography/computerized tomography (18F-FDG PET/CT) uptake is known to increase in infective and inflammatory conditions. Systemic inflammation plays a role in oncologic prognosis. Consequently, bone marrow increased uptake in oncology patients could potentially depict the systemic cancer burden. METHODS A single institute cohort analysis and a systematic review were performed, evaluating the prognostic role of 18F-FDG uptake in the bone marrow in solid neoplasms before treatment. The cohort included 113 esophageal cancer patients (adenocarcinoma or squamous cell carcinoma). The systematic review was based on 18 studies evaluating solid neoplasms, including gynecological, lung, pleura, breast, pancreas, head and neck, esophagus, stomach, colorectal, and anus. RESULTS Bone marrow 18F-FDG uptake in esophageal cancer was not correlated with staging, pathological response, and survival. High bone marrow uptake was related to advanced staging in colorectal, head and neck, and breast cancer, but not in lung cancer. Bone marrow 18F-FDG uptake was significantly associated with survival rates for lung, head and neck, breast, gastric, colorectal, pancreatic, and gynecological neoplasms but was not significantly associated with survival in pediatric neuroblastoma and esophageal cancer. CONCLUSION 18F-FDG bone marrow uptake in PET/CT has prognostic value in several solid neoplasms, including lung, gastric, colorectal, head and neck, breast, pancreas, and gynecological cancers. However, future studies are still needed to define the role of bone marrow role in cancer prognostication.
Collapse
Affiliation(s)
- Francisco Tustumi
- Department of Gastroenterology, Digestive Surgery Division, Universidade de São Paulo, Av. Dr. Eneas de Carvalho Aguiar 255, São Paulo 05403-000, SP, Brazil
| | - David Gutiérrez Albenda
- Department of Radiology and Oncology, Nuclear Medicine Division, Universidade de São Paulo, Av. Dr. Eneas de Carvalho Aguiar 255, São Paulo 05403-000, SP, Brazil
| | - Fernando Simionato Perrotta
- Department of Gastroenterology, Digestive Surgery Division, Universidade de São Paulo, Av. Dr. Eneas de Carvalho Aguiar 255, São Paulo 05403-000, SP, Brazil
| | - Rubens Antonio Aissar Sallum
- Department of Gastroenterology, Digestive Surgery Division, Universidade de São Paulo, Av. Dr. Eneas de Carvalho Aguiar 255, São Paulo 05403-000, SP, Brazil
| | - Ulysses Ribeiro Junior
- Department of Gastroenterology, Digestive Surgery Division, Universidade de São Paulo, Av. Dr. Eneas de Carvalho Aguiar 255, São Paulo 05403-000, SP, Brazil
| | - Carlos Alberto Buchpiguel
- Department of Radiology and Oncology, Nuclear Medicine Division, Universidade de São Paulo, Av. Dr. Eneas de Carvalho Aguiar 255, São Paulo 05403-000, SP, Brazil
| | - Paulo Schiavom Duarte
- Department of Radiology and Oncology, Nuclear Medicine Division, Universidade de São Paulo, Av. Dr. Eneas de Carvalho Aguiar 255, São Paulo 05403-000, SP, Brazil
| |
Collapse
|
6
|
A Transcriptomic Liquid Biopsy Assay for Predicting Resistance to Neoadjuvant Therapy in Esophageal Squamous Cell Carcinoma. Ann Surg 2022; 276:101-110. [PMID: 35703443 PMCID: PMC9276630 DOI: 10.1097/sla.0000000000005473] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to establish a liquid-biopsy assay to predict response to neoadjuvant therapy (NAT) in esophageal squamous cell carcinoma (ESCC) patients. SUMMARY BACKGROUND DATA Pretreatment prediction of resistance to NAT is of great significance for the selection of treatment options in ESCC patients. In this study, we comprehensively translated tissue-based microRNA (miRNA) and messenger RNA (mRNA) expression biomarkers into a liquid biopsy assay. METHODS We analyzed 186 clinical ESCC samples, which included 128 formalin-fixed paraffin-embedded and a matched subset of 58 serum samples, from 2 independent institutions. We performed quantitative reverse-transcription polymerase chain reaction, and developed a resistance-prediction model using the logistic regression analyses. RESULTS We first evaluated the potential of 4-miRNAs and 3-mRNAs panel, which robustly predicted resistance to NAT [area under the curve (AUC): 0.85]. Moreover, addition of tumor size to this panel increased predictive potential to establish a combination signature (AUC: 0.92). We successfully validated this signature performance in independent cohort, and our model was more accurate when the signature was combined with clinical predictors (AUC: 0.81) to establish a NAT resistance risk (NATRR) model. Finally, we successfully translated our NATRR model into a liquid biopsy assay (AUC: 0.78), and a multivariate regression analysis revealed this model as an independent predictor for response to NAT (odds ratio: 6.10; P < 0.01). CONCLUSIONS We successfully developed a liquid biopsy-based assay that allows robust prediction of response to NAT in ESCC patients, and our assay provides fundamentals of developing precision-medicine.
Collapse
|
7
|
Han J, Song Q, Guo F, Du R, Fang H, Kang J, Lu Z. Evaluation of response to stereotactic body radiation therapy for nonsmall cell lung cancer: PET response criteria in solid tumors versus response evaluation criteria in solid tumors. Nucl Med Commun 2022; 43:717-724. [PMID: 35354781 DOI: 10.1097/mnm.0000000000001556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Recommendations for surveillance after stereotactic body radiation therapy (SBRT) for early-stage nonsmall cell lung cancer (NSCLC) are not well defined. Recently, PET response criteria in solid tumors (PERCIST) have been proposed as a new standardized method to assess radiotherapeutic response both quantitatively and metabolically. The aim of this study was to evaluate therapeutic response following SBRT in early-stage NSCLC patients by comparing PERCIST with the currently widely used RECIST. MATERIALS AND METHODS Forty-nine patients with early-stage NSCLC who had been prescribed SBRT were studied. Responses of lesion were evaluated using CT and 18F-FDG PET according to the RECIST and PERCIST methods. PET-CT scans were obtained before SBRT and 3-6 months after SBRT. Associations between overall survival (OS) and clinicopathologic results (histology, tumor location, tumor size, lymphatic invasion, clinical stage, and radiotherapeutic responses in RECIST and PERCIST) were statistically analyzed. The median patient follow-up was 30 months. RESULTS Thirteen patients had stage IA, 9 stage IB, 10 stage IIA, and 17 stage IIB biopsy-proven NSCLC. Three-year OS was 79.6%. CT scans indicated three regional recurrences. PET-CT/chest indicated three regional recurrences and distant metastasis. Significant differences were observed in response classification between RECIST and PERCIST (Wilcoxon signed-rank test, P = 0.0041). Univariate analysis showed that clinical stage, RECIST, and PERCIST were significant factors associated with OS, whereas by multivariate analysis PERCIST was the only predictor of OS. SMD, PMD/PMR, and CMR in PERCIST criteria were indicative of a 9.900-fold increase in the risk of OS in early NSCLC patients [risk ratio, 9.900 (95% CI, 1.040-21.591); P = 0.001]. CONCLUSION RECIST based on the anatomic size reduction rate did not demonstrate the correlation between radiotherapeutic response and prognosis in patients with early-stage NSCLC receiving SBRT. However, PERCIST was shown as the strongest independent predictor of outcomes. PERCIST might be considered more suitable for the evaluation of NSCLC tumor response to SBRT than RECIST.
Collapse
Affiliation(s)
- Jixia Han
- Department of Radiological and Environmental Medicine, China Institute for Radiation Protection, Taiyuan
| | - Qi Song
- Senior Department of Obstetrics and Gynecology, The Seventh Medical Center of PLA General Hospital
| | - Feng Guo
- Radiation Oncology and Integrative Oncology, The Sixth Medical Center of the General Hospital of the People's Liberation Army of China, Beijing, P. R. China
| | - Rui Du
- Radiation Oncology and Integrative Oncology, The Sixth Medical Center of the General Hospital of the People's Liberation Army of China, Beijing, P. R. China
| | - Henghu Fang
- Radiation Oncology and Integrative Oncology, The Sixth Medical Center of the General Hospital of the People's Liberation Army of China, Beijing, P. R. China
| | - Jingbo Kang
- Radiation Oncology and Integrative Oncology, The Sixth Medical Center of the General Hospital of the People's Liberation Army of China, Beijing, P. R. China
| | - Zejun Lu
- Radiation Oncology and Integrative Oncology, The Sixth Medical Center of the General Hospital of the People's Liberation Army of China, Beijing, P. R. China
| |
Collapse
|
8
|
An evaluation of the response to neoadjuvant chemotherapy for osteosarcoma of extremities: PERCIST versus RECIST 1.1 criteria after long-term follow-up. Ann Nucl Med 2022; 36:553-561. [PMID: 35380350 DOI: 10.1007/s12149-022-01737-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/13/2022] [Indexed: 11/01/2022]
Abstract
OBJECTIVE The aim of this study was to compare the recent Positron emission tomography (PET) Response Criteria in Solid Tumors (PERCIST) and Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria for evaluating the response of osteosarcoma to neoadjuvant chemotherapy of the extremities. METHODS We retrospectively reviewed patients with osteosarcoma of the extremities who received neoadjuvant chemotherapy and then surgical resection at Peking University People's Hospital. Fluorine-18 fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) and magnetic resonance imaging (MRI) were performed prior to chemotherapy and before surgical resection. Therapeutic response was assessed separately by the PERCIST and RECIST 1.1 criteria. The association between the data acquired by the PERCIST and RECIST 1.1 criteria was then analyzed by Wilcoxon's signed-rank test. The association between the PERCIST criteria and the pathological necrosis rate was analyzed by Fisher's exact test. Finally, the impact of a range of clinicopathological factors on overall survival (OS) and event-free survival (EFS) was analyzed by Cox proportional hazards regression. RESULTS We recruited 68 patients with a median follow-up of 74 months (range 45-102 months). The evaluations resulting from the RECIST 1.1 and PERCIST criteria were significantly different (p = 0.000). Only two responders were identified according to the RECIST 1.1 criteria. However, 34 responders were identified by the PERCIST criteria. Data arising from the PERCIST criteria were in accordance with the pathological necrosis rate. Survival analysis showed that metastasis at diagnosis, poor pathological response, and disease progression (according to the RECIST 1.1 or PERCIST criteria) were all associated with a poor prognosis (p < 0.05). CONCLUSION Our data indicate that the PERCIST criteria are significantly more sensitive than RECIST 1.1 criteria to identify more responders when evaluating the response of osteosarcoma to neoadjuvant chemotherapy.
Collapse
|
9
|
Yang B, Ma H, Bian Y. LINC00261 Inhibits Esophageal Cancer Radioresistance by Down-Regulating microRNA-552-3p and Promoting DIRAS1. Cancer Manag Res 2021; 13:8559-8573. [PMID: 34803403 PMCID: PMC8597985 DOI: 10.2147/cmar.s332640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/14/2021] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Esophageal cancer (EC) represents a life-threatening tumor with an ever-increasing incidence rate. Long intergenic non-protein coding RNAs (LINCs) have also become a topic of interest in EC. In a similar light, the current study aimed to investigate the role of LINC00261 in EC radioresistance. METHODS Firstly, radioresistant EC cell lines TE-1-R and TE-5-R were established using TE-1 and TE-5 cells. Subsequently, LINC00261, microRNA (miR)-552-3p, and DIRAS1 expression patterns in EC tissues and adjacent normal tissues and EC cells were evaluated. In addition, survival fraction (SF), colony formation, apoptosis, and γ-H2AX levels were analyzed, followed by the detection of the binding relation between LINC00261 and miR-552-3p and between miR-552-3p and DIRAS1. Lastly, xenograft transplantation was carried out to confirm the effects of LINC00261 on EC radioresistance in vivo. RESULTS LINC00261 and DIRAS1 were poorly-expressed in EC tissues and cells, but miR-552-3p was over-expressed. In EC cells with X-ray radiation, over-expression of LINC00261 reduced SF and cell viability, strengthened γ-H2AX levels, and promoted apoptosis, while all these trends were counteracted by miR-522-3p over-expression or DIRAS1 silencing. Mechanistic investigation further validated the binding relation between LINC00261 and miR-552-3p, and between miR-552-3p and DIRAS1. Moreover, LINC00261 over-expression suppressed tumor growth and reduced EC radioresistance in vivo. CONCLUSION Altogether, our findings indicated that LINC00261 exerts a suppressive effect on EC radioresistance via the competing endogenous RNA network to sponge miR-552-3p and up-regulate DIRAS1 transcription.
Collapse
Affiliation(s)
- Baolong Yang
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Xi ‘an Jiaotong University, Xi ‘an, Shanxi Province, 710004, People’s Republic of China
| | - Hongbing Ma
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Xi ‘an Jiaotong University, Xi ‘an, Shanxi Province, 710004, People’s Republic of China
| | - Yan Bian
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Xi ‘an Jiaotong University, Xi ‘an, Shanxi Province, 710004, People’s Republic of China
| |
Collapse
|
10
|
Taniyama Y, Murakami K, Yoshida N, Takahashi K, Matsubara H, Baba H, Kamei T. Evaluating the effect of Neoadjuvant chemotherapy for esophageal Cancer using the RECIST system with shorter-axis measurements: a retrospective multicenter study. BMC Cancer 2021; 21:1008. [PMID: 34496769 PMCID: PMC8428108 DOI: 10.1186/s12885-021-08747-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/29/2021] [Indexed: 11/16/2022] Open
Abstract
Background Evaluating the effect on primary lesions is important in determining treatment strategies for esophageal cancer. The Response Evaluation Criteria in Solid Tumors system, which employs the longest diameter for measuring tumors, is commonly used for evaluating treatment effects. However, the usefulness of these criteria in assessing primary esophageal tumors remains controversial. Thus, we evaluated this issue by measuring not only the longest diameter but also the shorter axis of the tumor. Methods We retrospectively reviewed data from 313 patients with esophageal cancer treated with neoadjuvant chemotherapy followed by esophagectomy at three major high-volume centers in Japan. All patients underwent contrast-enhanced computed tomography before and after chemotherapy. The longest and shortest tumor diameters were measured in each case. Treatment effects were adapted to the Response Evaluation Criteria in Solid Tumors system. Correlations between pathological and survival data were also analyzed. Results Inter-observer discrepancies were examined for changes in the longest diameter and shorter axis of the tumor (the intraclass correlation coefficients were 0.550 and 0.624, respectively). The shorter axis was correlated with the pathological response in the multivariate analysis (p < 0.001). The shorter axis was significantly associated with overall survival and disease-free survival (both p < 0.001), whereas this association was not observed for the longest tumor diameter. Conclusions This multicenter study demonstrated that the Response Evaluation Criteria in Solid Tumors system is useful for predicting pathological response and survival by incorporating the shorter axis of the primary esophageal tumor. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08747-y.
Collapse
Affiliation(s)
- Yusuke Taniyama
- Department of Surgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Kentaro Murakami
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kozue Takahashi
- Department of Surgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.,Department of Radiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takashi Kamei
- Department of Surgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
11
|
Xie CY, Pang CL, Chan B, Wong EYY, Dou Q, Vardhanabhuti V. Machine Learning and Radiomics Applications in Esophageal Cancers Using Non-Invasive Imaging Methods-A Critical Review of Literature. Cancers (Basel) 2021; 13:2469. [PMID: 34069367 PMCID: PMC8158761 DOI: 10.3390/cancers13102469] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 11/16/2022] Open
Abstract
Esophageal cancer (EC) is of public health significance as one of the leading causes of cancer death worldwide. Accurate staging, treatment planning and prognostication in EC patients are of vital importance. Recent advances in machine learning (ML) techniques demonstrate their potential to provide novel quantitative imaging markers in medical imaging. Radiomics approaches that could quantify medical images into high-dimensional data have been shown to improve the imaging-based classification system in characterizing the heterogeneity of primary tumors and lymph nodes in EC patients. In this review, we aim to provide a comprehensive summary of the evidence of the most recent developments in ML application in imaging pertinent to EC patient care. According to the published results, ML models evaluating treatment response and lymph node metastasis achieve reliable predictions, ranging from acceptable to outstanding in their validation groups. Patients stratified by ML models in different risk groups have a significant or borderline significant difference in survival outcomes. Prospective large multi-center studies are suggested to improve the generalizability of ML techniques with standardized imaging protocols and harmonization between different centers.
Collapse
Affiliation(s)
- Chen-Yi Xie
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Chun-Lap Pang
- Department of Radiology, The Christies’ Hospital, Manchester M20 4BX, UK;
- Division of Dentistry, School of Medical Sciences, University of Manchester, Manchester M15 6FH, UK
| | - Benjamin Chan
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (B.C.); (E.Y.-Y.W.)
| | - Emily Yuen-Yuen Wong
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (B.C.); (E.Y.-Y.W.)
| | - Qi Dou
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China;
| | - Varut Vardhanabhuti
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| |
Collapse
|
12
|
Kaida H, Kitajima K, Nakajo M, Ishibashi M, Matsunaga T, Minamimoto R, Hirata K, Nakatani K, Hung A, Hattori S, Yasuda T, Ishii K. Predicting tumor response and prognosis to neoadjuvant chemotherapy in esophageal squamous cell carcinoma patients using PERCIST: a multicenter study in Japan. Eur J Nucl Med Mol Imaging 2021; 48:3666-3682. [PMID: 33934168 DOI: 10.1007/s00259-021-05365-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 04/11/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE To investigate the usefulness of the positron emission tomography response criteria in solid tumors 1.0 (PERCIST1.0) for predicting tumor response to neoadjuvant chemotherapy and prognosis and determine whether PERCIST improvements are necessary for esophageal squamous cell carcinoma (ESCC) patients. PATIENTS AND METHODS We analyzed the cases of 177 ESCC patients and examined the association between PERCIST and their pathological responses. Associations of whole-PERCIST with progression-free survival (PFS) and overall survival (OS) were evaluated by a Kaplan-Meier analysis and Cox proportional hazards model. To investigate potential PERCIST improvements, we used the survival tree technique to understand patients' prognoses. RESULTS There were significant correlations between the pathologic response and PERCIST of primary tumor (p < 0.001). The optimal cutoff value of the primary tumors' SULpeak response to classify pathologic responses was -50.0%. The diagnostic accuracy of SULpeak response was 87.3% sensitivity, 54.1% specificity, 68.9% accuracy, positive predictive value 60.5%, and negative predictive value 84.1%. Whole-PERCIST was significantly associated with PFS and OS. The survival tree results indicated that a high reduction of the whole SULpeak response was significantly correlated with the patients' prognoses. The cutoff values for the separation of prognoses were - 52.5 for PFS and - 47.1% for OS. CONCLUSION PERCIST1.0 can help predict tumor responses and prognoses. However, 18F-FDG-PET/CT tends to underestimate residual tumors in histopathological response evaluations. Modified PERCIST, in which the partial metabolic response is further classified by the SULpeak response (-50%), might be more appropriate than PERCIST1.0 for evaluating tumor responses and stratifying high-risk patients for recurrence and poor prognosis.
Collapse
Affiliation(s)
- Hayato Kaida
- Department of Radiology, Kindai University Faculty of Medicine, 377-2, Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan.
| | - Kazuhiro Kitajima
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Masatoyo Nakajo
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-5-31, Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Mana Ishibashi
- Division of Radiology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, 36-1, Nishi-cho, Yonago, Tottori, 683-8504, Japan
| | - Tomoyuki Matsunaga
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University of Faculty of Medicine, 36-1, Nishi-cho, Yonago, Tottori, 683-8504, Japan
| | - Ryogo Minamimoto
- Division of Nuclear Medicine, Department of Radiology, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Kenji Hirata
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, Kita15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Koya Nakatani
- Department of Diagnostic Radiology, Kurashiki Central Hospital, 1-1-1, Miwa, Kurashiki, Okayama, 710-8602, Japan
| | - Ao Hung
- Department of Biomedical Statistics, Osaka University Graduate School of Medicine, 2-2, Yamadagaoka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Hattori
- Department of Biomedical Statistics, Osaka University Graduate School of Medicine, 2-2, Yamadagaoka, Suita, Osaka, 565-0871, Japan.,Institute for Open and Transdisciplinary Research Initiative, Osaka University, 2-2, Yamadagaoka, Suita, Osaka, 565-0871, Japan
| | - Takushi Yasuda
- Department of Surgery, Kindai University Faculty of Medicine, 377-2, Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan
| | - Kazunari Ishii
- Department of Radiology, Kindai University Faculty of Medicine, 377-2, Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan
| |
Collapse
|
13
|
Duclos V, Iep A, Gomez L, Goldfarb L, Besson FL. PET Molecular Imaging: A Holistic Review of Current Practice and Emerging Perspectives for Diagnosis, Therapeutic Evaluation and Prognosis in Clinical Oncology. Int J Mol Sci 2021; 22:4159. [PMID: 33923839 PMCID: PMC8073681 DOI: 10.3390/ijms22084159] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
PET/CT molecular imaging has been imposed in clinical oncological practice over the past 20 years, driven by its two well-grounded foundations: quantification and radiolabeled molecular probe vectorization. From basic visual interpretation to more sophisticated full kinetic modeling, PET technology provides a unique opportunity to characterize various biological processes with different levels of analysis. In clinical practice, many efforts have been made during the last two decades to standardize image analyses at the international level, but advanced metrics are still under use in practice. In parallel, the integration of PET imaging with radionuclide therapy, also known as radiolabeled theranostics, has paved the way towards highly sensitive radionuclide-based precision medicine, with major breakthroughs emerging in neuroendocrine tumors and prostate cancer. PET imaging of tumor immunity and beyond is also emerging, emphasizing the unique capabilities of PET molecular imaging to constantly adapt to emerging oncological challenges. However, these new horizons face the growing complexity of multidimensional data. In the era of precision medicine, statistical and computer sciences are currently revolutionizing image-based decision making, paving the way for more holistic cancer molecular imaging analyses at the whole-body level.
Collapse
Affiliation(s)
- Valentin Duclos
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
| | - Alex Iep
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
| | - Léa Gomez
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
| | - Lucas Goldfarb
- Service Hospitalier Frédéric Joliot-CEA, 91401 Orsay, France;
| | - Florent L. Besson
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
- Université Paris Saclay, CEA, CNRS, Inserm, BioMaps, 91401 Orsay, France
- School of Medicine, Université Paris Saclay, 94720 Le Kremlin-Bicêtre, France
| |
Collapse
|
14
|
Ni C, Qin D, Cheng H, Zhou M, Luo D. Effect Evaluation of Combined Application of Magnetic Resonance Diffusion Tensor Imaging and Brain Function Imaging in Radiation Therapy of Brain Tumours Involving Motor Pathways. JOURNAL OF MEDICAL IMAGING AND HEALTH INFORMATICS 2021. [DOI: 10.1166/jmihi.2021.3329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study is an attempt to find a way for functional imaging information to be applied clinically in radiation therapy. The basal nucleus is a collective term for a group of neural nucleus in the central nervous system that connects the pontine, brainstem, and cerebral cortex, including
the caudate nucleus, the bean-shaped nucleus, the screen-shaped nucleus, and the amygdala. It is difficult to find the exact position of these neural nuclei on the computed tomography (CT) image or the T1 or T2 sequence of magnetic resonance. However, the development of neurosurgery has partially
confirmed that these functional nuclei are involved in advanced cognitive functions such as memory, emotion, and learning. Neurosurgery has tried to avoid damaging these nucleus groups during surgery to improve the quality of life of patients, and there is currently no clear strategy for this
in radiotherapy. Because CT and magnetic resonance spin echo (SE) sequences are difficult to find the anatomical location of the nucleus, it is difficult to have any strategy to protect these functions in radiotherapy planning. This article uses diffusion tensor imaging (DTI) images and fiber
bundle tracking to obtain a more accurate anatomical position of the nerve nucleus on the image, and provides some available strategies for radiotherapy to protect patients’ brain function. The conclusion of this paper is that the combined application of DTI and functional magnetic resonance
imaging (fMRI) can better observe the relationship among tumours, functional areas and white matter fibers, and guide the designation of radiotherapy plans.
Collapse
Affiliation(s)
- Cheng Ni
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, En Shi 445000, China
| | - Daming Qin
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, En Shi 445000, China
| | - Hong Cheng
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, En Shi 445000, China
| | - Meng Zhou
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, En Shi 445000, China
| | - Dandan Luo
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, En Shi 445000, China
| |
Collapse
|
15
|
Assessment of tumor response to definitive chemoradiotherapy and prognosis prediction in patients with esophageal cancer judged by PET response criteria in solid tumors: multicenter study in Japan. Nucl Med Commun 2021; 41:443-451. [PMID: 32187159 DOI: 10.1097/mnm.0000000000001168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The aim of the study was to evaluate PET response criteria in solid tumors (PERCIST) to indicate therapeutic response to definitive chemoradiotherapy, as well as prediction of recurrence and death in patients with esophageal cancer. METHODS Before and after recieving definitive chemoradiotherapy, 181 patients with esophageal cancer underwent fluorodeoxyglucose-PET/computed tomography (FDG-PET/CT). PERCIST, reduction rates of tumor uptake and volume of whole lesions, tumor node metastasis (TNM) staging regarding progression-free survival (PFS), and overall survival (OS) were analyzed using log-rank and Cox methods. RESULTS Complete metabolic response (CMR), partial metabolic response (PMR), stable metabolic disease (SMD), and progressive metabolic disease (PMD) shown by PERCIST were seen in 42 (23.2%), 113 (62.4%), 14 (7.7%), and 12 (6.6%) patients, respectively. Progression developed in 137 (75.7%) patients and 101 (56.1%) patients died (median follow-up 16.9, range 3.2-124.9 months). Those who achieved CMR showed significantly longer PFS and OS as compared with patients who did not (PMR, SMD, and PMD) (both P < 0.0001). In univariate analysis, initial clinical T status (P = 0.0048), N status (P = 0.011), and TNM stage (P = 0.0006), PERCIST (P < 0.0001), and reduction rate of peak lean body mass standardized uptake value (P < 0.0001), of metabolic tumor volume (P < 0.0001), and of total lesion glycolysis (TLG) (P < 0.0001) were associated with significantly increased OS. Multivariate analysis confirmed PERCIST [hazard ratio (HR): 13.15, 95% confidence interval (CI), 4.54-55.8; P < 0.0001], and TLG reduction rate (HR: 2.21, 95% CI, 1.04-4.68; P = 0.040) as independent OS predictors. CONCLUSION PERCIST is useful for evaluating therapeutic response to definitive chemoradiotherapy, and predicting progression and death in patients with esophageal cancer.
Collapse
|
16
|
Cheng W, Shi X, Lin M, Yao Q, Ma J, Li J. LncRNA MAGI2-AS3 Overexpression Sensitizes Esophageal Cancer Cells to Irradiation Through Down-Regulation of HOXB7 via EZH2. Front Cell Dev Biol 2020; 8:552822. [PMID: 33330444 PMCID: PMC7732634 DOI: 10.3389/fcell.2020.552822] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/29/2020] [Indexed: 12/31/2022] Open
Abstract
Background Accumulating evidence has suggested that aberrant expression of long non-coding RNAs (lncRNAs) may contribute to cancer progression in association with radioresistance. The current study aimed to identify the potential role of lncRNA MAGI2-AS3 and the underlying mechanism in its regulation of the radio-sensitivity of esophageal cancer cells. Methods and Results Initially, we detected high expression of HOXB7 from microarray-based gene expression profiling of esophageal cancer. Then, we identified the interactions among MAGI2-AS3, HOXB7, and EZH2 by dual-luciferase reporter gene assay, RNA pull-down assay, RIP assay and ChIP assay. HOXB7 was highly-expressed, while MAGI2-AS3 was poorly-expressed in esophageal cancer tissues and cells. The effect of MAGI2-AS3 and HOXB7 on esophageal cancer cell proliferation and apoptosis as well as tumorigenicity of radioresistant cells was examined by gain- and loss-of-function experiments. Interestingly, MAGI2-AS3 down-regulated HOXB7 through interaction with EZH2, which promoted cell apoptosis and inhibited proliferation and radio-resistance. Besides, down-regulation of MAGI2-AS3 exerted a promoting effect on these malignant phenotypes. Conclusion Taken together, our results reveal the potential role of MAGI2-AS3 over-expression in controlling esophageal cancer resistance to radiotherapy by down-regulating HOXB7, this providing a candidate biomarker for resistance to radiotherapy.
Collapse
Affiliation(s)
- Wenfang Cheng
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Xiuling Shi
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Mingqiang Lin
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Qiwei Yao
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Jiayu Ma
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Jiancheng Li
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
17
|
Sorace AG, Elkassem AA, Galgano SJ, Lapi SE, Larimer BM, Partridge SC, Quarles CC, Reeves K, Napier TS, Song PN, Yankeelov TE, Woodard S, Smith AD. Imaging for Response Assessment in Cancer Clinical Trials. Semin Nucl Med 2020; 50:488-504. [PMID: 33059819 PMCID: PMC7573201 DOI: 10.1053/j.semnuclmed.2020.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The use of biomarkers is integral to the routine management of cancer patients, including diagnosis of disease, clinical staging and response to therapeutic intervention. Advanced imaging metrics with computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET) are used to assess response during new drug development and in cancer research for predictive metrics of response. Key components and challenges to identifying an appropriate imaging biomarker are selection of integral vs integrated biomarkers, choosing an appropriate endpoint and modality, and standardization of the imaging biomarkers for cooperative and multicenter trials. Imaging biomarkers lean on the original proposed quantified metrics derived from imaging such as tumor size or longest dimension, with the most commonly implemented metrics in clinical trials coming from the Response Evaluation Criteria in Solid Tumors (RECIST) criteria, and then adapted versions such as immune-RECIST (iRECIST) and Positron Emission Tomography Response Criteria in Solid Tumors (PERCIST) for immunotherapy response and PET imaging, respectively. There have been many widely adopted biomarkers in clinical trials derived from MRI including metrics that describe cellularity and vascularity from diffusion-weighted (DW)-MRI apparent diffusion coefficient (ADC) and Dynamic Susceptibility Contrast (DSC) or dynamic contrast enhanced (DCE)-MRI (Ktrans, relative cerebral blood volume (rCBV)), respectively. Furthermore, Fluorodexoyglucose (FDG), fluorothymidine (FLT), and fluoromisonidazole (FMISO)-PET imaging, which describe molecular markers of glucose metabolism, proliferation and hypoxia have been implemented into various cancer types to assess therapeutic response to a wide variety of targeted- and chemotherapies. Recently, there have been many functional and molecular novel imaging biomarkers that are being developed that are rapidly being integrated into clinical trials (with anticipation of being implemented into clinical workflow in the future), such as artificial intelligence (AI) and machine learning computational strategies, antibody and peptide specific molecular imaging, and advanced diffusion MRI. These include prostate-specific membrane antigen (PSMA) and trastuzumab-PET, vascular tumor burden extracted from contrast-enhanced CT, diffusion kurtosis imaging, and CD8 or Granzyme B PET imaging. Further excitement surrounds theranostic procedures such as the combination of 68Ga/111In- and 177Lu-DOTATATE to use integral biomarkers to direct care and personalize therapy. However, there are many challenges in the implementation of imaging biomarkers that remains, including understand the accuracy, repeatability and reproducibility of both acquisition and analysis of these imaging biomarkers. Despite the challenges associated with the biological and technical validation of novel imaging biomarkers, a distinct roadmap has been created that is being implemented into many clinical trials to advance the development and implementation to create specific and sensitive novel imaging biomarkers of therapeutic response to continue to transform medical oncology.
Collapse
Affiliation(s)
- Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL.
| | - Asser A Elkassem
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Samuel J Galgano
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL; Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL
| | - Benjamin M Larimer
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | | | - C Chad Quarles
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ
| | - Kirsten Reeves
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; Cancer Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Tiara S Napier
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; Cancer Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Patrick N Song
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX; Department of Diagnostic Medicine, University of Texas at Austin, Austin, TX; Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX
| | - Stefanie Woodard
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Andrew D Smith
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
18
|
Gillman JA, Pantel AR, Mankoff DA, Edmonds CE. Update on Quantitative Imaging for Predicting and Assessing Response in Oncology. Semin Nucl Med 2020; 50:505-517. [PMID: 33059820 PMCID: PMC9788668 DOI: 10.1053/j.semnuclmed.2020.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Molecular imaging has revolutionized clinical oncology by imaging-specific facets of cancer biology. Through noninvasive measurements of tumor physiology, targeted radiotracers can serve as biomarkers for disease characterization, prognosis, response assessment, and predicting long-term response/survival. In turn, these imaging biomarkers can be utilized to tailor therapeutic regimens to tumor biology. In this article, we review biomarker applications for response assessment and predicting long-term outcomes. 18F-fluorodeoxyglucose (FDG), a measure of cellular glucose metabolism, is discussed in the context of lymphoma and breast and lung cancer. FDG has gained widespread clinical acceptance and has been integrated into the routine clinical care of several malignancies, most notably lymphoma. The novel radiotracers 16α-18F-fluoro-17β-estradiol and 18F-fluorothymidine are reviewed in application to the early prediction of response assessment of breast cancer. Through illustrative examples, we explore current and future applications of molecular imaging biomarkers in the advancement of precision medicine.
Collapse
Affiliation(s)
- Jennifer A Gillman
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Austin R Pantel
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - David A Mankoff
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Christine E Edmonds
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
19
|
Hirata A, Hayano K, Ohira G, Imanishi S, Hanaoka T, Murakami K, Aoyagi T, Shuto K, Matsubara H. Volumetric histogram analysis of apparent diffusion coefficient for predicting pathological complete response and survival in esophageal cancer patients treated with chemoradiotherapy. Am J Surg 2020; 219:1024-1029. [PMID: 31387687 DOI: 10.1016/j.amjsurg.2019.07.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/23/2019] [Accepted: 07/27/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The purpose of the study was to evaluate whether histogram analysis of apparent diffusion coefficient (ADC) can predict pathological complete response (pCR) and survival in patients with esophageal squamous cell carcinoma (ESCC) after chemoradiotherapy (CRT). METHODS We retrospectively identified 58 patients with ESCC who underwent surgery after CRT between 2007 and 2016. Associations of pretreatment histogram derived ADC parameters with pathological response and survival were analyzed. RESULTS Tumors achieved pCR (10 patients, 17.2%) showed significant lower ADC, higher kurtosis, and higher skewness than those of non-pCR (p = 0.005, 0.007, <0.001, respectively). Receiver operating characteristics analysis demonstrated skewness was the best predictor for pCR (AUC = 0.86), with a cut off value of 0.50 (accuracy, 86.2%). In Kaplan-Meier analysis, patients with higher skewness tumors (≥0.50) showed a significantly better recurrence free survival (p = 0.032, log-rank). CONCLUSIONS Histogram analysis of ADC can enable prediction of pCR and survival in ESCC patients treated with preoperative CRT. A SHORT SUMMARY ADC histogram analysis can be an imaging biomarker for esophageal cancer patients treated with CRT.
Collapse
Affiliation(s)
- Atsushi Hirata
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Japan
| | - Koichi Hayano
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Japan.
| | - Gaku Ohira
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Japan
| | - Shunsuke Imanishi
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Japan
| | - Toshiharu Hanaoka
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Japan
| | - Kentaro Murakami
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Japan
| | - Tomoyoshi Aoyagi
- Department of Surgery, Funabashi Municipal Medical Center, Japan
| | - Kiyohiko Shuto
- Department of Surgery, Teikyo University Chiba Medical Center, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Japan
| |
Collapse
|
20
|
Metabolic Tumor Volume Change Predicts Long-term Survival and Histological Response to Preoperative Chemotherapy in Locally Advanced Esophageal Cancer. Ann Surg 2020; 270:1090-1095. [PMID: 29727327 DOI: 10.1097/sla.0000000000002808] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Here, we assess the ability of metabolic tumor volume (MTV) as measured by F-fluorodeoxyglucose-positron emission tomography/computed tomography (F-FDG PET/CT) to evaluate neoadjuvant chemotherapy response for patients with locally advanced esophageal cancer (EC). BACKGROUND Optimal methods to evaluate treatment response for EC patients have not yet been established. Although previous studies have reported the value of standardized uptake value (SUV), the accuracy of predicting histological response or long-term survival in EC is limited. METHODS In all, 102 EC patients without distant metastasis who underwent F-FDG PET/CT both before and after the preoperative chemotherapy series were analyzed. RESULTS The median primary tumor MTV values before and after preoperative chemotherapy were 22.55 (range 0.4-183.1) and 2.75 (0-52.9), respectively, and the median MVT reduction rate was 86.5%. We found the most significant difference in survival between PET responders and nonresponders with a cut-off value of 60% MTV reduction, using a 10% stepwise cut-off analysis [2-year progression-free survival (PFS): 79.2 vs 44.4%; hazard ratio (HR) 3.397; P < 0.0001). With this cut-off value, histological response (P = 0.0091), tumor location (P = 0.0102), pT (P = 0.0011), and pN (P = 0.0110) were significantly associated with PET response. Univariate analysis of PFS indicated a correlation between PFS and tumor size, cT, decrease of primary lesion by CT, SUVmax reduction rate, MTV reduction rate, pT, pN, and pM. Multivariate analysis further identified pM (HR 3.063; P = 0.0279) and MTV reduction rate (HR 2.471; P = 0.0263) to be independent prognostic predictors, but not decrease of primary lesion by CT or SUVmax reduction rate. CONCLUSION MTV change is clinically useful in predicting both long-term survival and histological response to preoperative chemotherapy in EC patients, after determining the optimal cut-off value based on survival analysis.
Collapse
|
21
|
Lin J, Liu Z, Liao S, Li E, Wu X, Zeng W. Elevation of long non-coding RNA GAS5 and knockdown of microRNA-21 up-regulate RECK expression to enhance esophageal squamous cell carcinoma cell radio-sensitivity after radiotherapy. Genomics 2019; 112:2173-2185. [PMID: 31866421 DOI: 10.1016/j.ygeno.2019.12.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/04/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Lately, lncRNAs have been proposed to function in the radio-sensitivity of tumor cells, yet the role of lncRNA GAS5 in that of esophageal squamous cell carcinoma (ESCC) has scarcely been studied. This study aims to examine GAS5's effects on ESCC cell radio-sensitivity. METHODS GAS5, miR-21 and RECK expression in radiation-sensitive and radiation-resistant ESCC tissues, and TE-1 and TE-1-R cells was determined. TE-1 and TE-1-R cells were treated with pcDNA-GAS5 or miR-21 inhibitors to figure out their roles in ESCC cell proliferation, radio-sensitivity, and apoptosis via gain- and loss-of-function experiments. RESULTS We found underexpressed GAS5 and RECK, and overexpressed miR-21 in ESCC. GAS5 elevation and miR-21 inhibition reduced viability and the colony formation ability, and enhanced the apoptosis of ESCC cells under radiation. CONCLUSION Our study reveals that GAS5 elevation up-regulates RECK expression by down-regulating miR-21 to increase ESCC cell apoptosis after radiation therapy, thus enhancing cell radio-sensitivity.
Collapse
Affiliation(s)
- Jing Lin
- Department of Oncology, The First Affiliated Hospital of Shantou Univresity Medical College, Shantou 515041, China.
| | - Zewa Liu
- Department of Oncology, The First Affiliated Hospital of Shantou Univresity Medical College, Shantou 515041, China
| | - Shasha Liao
- Department of Oncology, Shantou Longhu people's Hospital, Shantou 515041, Guangdong, China
| | - E Li
- Department of Oncology, Shantou Longhu people's Hospital, Shantou 515041, Guangdong, China
| | - Xiaohua Wu
- Department of Oncology, Shantou Longhu people's Hospital, Shantou 515041, Guangdong, China
| | - Wanting Zeng
- Division of Medical University College, London WCIE 6BT, United Kingdom
| |
Collapse
|
22
|
de Gouw DJJM, Klarenbeek BR, Driessen M, Bouwense SAW, van Workum F, Fütterer JJ, Rovers MM, Ten Broek RPG, Rosman C. Detecting Pathological Complete Response in Esophageal Cancer after Neoadjuvant Therapy Based on Imaging Techniques: A Diagnostic Systematic Review and Meta-Analysis. J Thorac Oncol 2019; 14:1156-1171. [PMID: 30999111 DOI: 10.1016/j.jtho.2019.04.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 03/14/2019] [Accepted: 03/17/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Up to 32% of patients with esophageal cancer show a pathological complete response (ypCR) after neoadjuvant therapy. To prevent overtreatment, the indication to perform esophagectomy in these patients should be reconsidered. Implementing an organ-preserving strategy for patients with ypCR requires an accurate assessment of residual disease after neoadjuvant treatment. The aim of this study was to systematically review the effectiveness of imaging techniques used for detection of ypCR after neoadjuvant therapy but before resection in patients with esophageal cancer. METHODS A systematic literature search of the Medline, Embase, and Cochrane Library databases was performed from January 1, 2000, to December 13, 2017. Eligible studies were diagnostic studies that compared results of imaging modalities after neoadjuvant therapy to histopathological findings in the resection specimen after esophagectomy. Methodological quality was assessed by the Cochrane Quality Assessment of Diagnostic Accuracy Studies, version 2, model. Primary outcome measures were true positive, false-positive, false-negative, and true negative values of imaging techniques predicting ypCR. A meta-analysis was performed by pooling sensitivities and specificities by using a bivariate model. RESULTS A total of 4420 articles were identified. After exclusion of irrelevant titles and abstracts, 360 articles were reviewed in full text. In total, four imaging modalities (computed tomography [CT], positron emission tomography [PET-CT], endoscopic ultrasound [EUS], and magnetic resonance imaging [MRI]) were used for restaging. The meta-analysis was conducted with data from 56 studies involving 3625 patients. The pooled sensitivities of CT, PET-CT, EUS, and MRI for detecting ypCR were 0.35, 0.62, 0.01 and 0.80, respectively, whereas the pooled specificities were 0.83, 0.73, 0.99, and 0.83, respectively. The positive predictive value in detecting ypCR was 0.47 for CT, 0.41 for PET-CT, not applicable for EUS, and 0.61 for MRI. CONCLUSION Current imaging modalities such as CT, PET-CT, and EUS seem to be insufficiently accurate to identify complete responders. More accurate diagnostic tests are needed to improve restaging accuracy for patients with esophageal cancer.
Collapse
Affiliation(s)
- Didi J J M de Gouw
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands.
| | | | - Mitchell Driessen
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stefan A W Bouwense
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frans van Workum
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jurgen J Fütterer
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maroeska M Rovers
- Department of Health Evidence and Operating Rooms, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Camiel Rosman
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
23
|
Xie T, Ye Z, Pang P, Shao G. Quantitative Multiparametric MRI May Augment the Response to Radiotherapy in Mid-Treatment Assessment of Patients with Esophageal Carcinoma. Oncol Res Treat 2019; 42:326-333. [PMID: 31064001 DOI: 10.1159/000499322] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 02/28/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The purpose of this study was to assess the mid-treatment response to radiotherapy (RT) using dynamic contrast-enhanced MRI (DCE-MRI) and diffusion-weighted imaging (DWI) in patients with esophageal cancer (EC). METHODS 42 patients with squamous EC were prepared for DCE-MRI and DWI scans both before treatment (NRT) and after the fifth radiotherapy (5th RT). The patients were classified into two groups (complete response [CR] and partial response [PR]) according to tumor regression after treatment. The quantitative parameters of DCE-MRI (Ktrans, Kep, Ve, and ADC) were measured. A receiver operating characteristic curve (ROC) was used to detect the efficiency of the above parameters. RESULTS After 1 month of RT, 29 patients were classified as CR and 11 patients were classified as PR. In the NRT group, the p values of Ktrans, Kep, Ve, and ADC were 0.004, 0.078, 0.0008, and <0.0001, respectively. After the 5th RT, the p values of the above parameters were <0.001, 0.005, 0.108, and 0.365, respectively. In the NRT group, the areas under the ROC curves of Ktrans, Ve, and ADC were 0.790, 0.617, and 0.737; the sensitivity values were 89.3, 92.5, and 90.0%; the specificity values were 69.4, 27.5, and 50.0%. In the 5th RT group, the areas under the ROC curves of Ktrans and Kep were 0.816 and 0.804; the sensitivity values were 71.2 and 95.0%; the specificity values were 81.6 and 50.0%. CONCLUSION DCE-MRI combined with DWI is effective in the early prediction of radiotherapeutic response of EC after the 5th RT other than after the traditional final treatment.
Collapse
Affiliation(s)
- Tieming Xie
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhimin Ye
- Department of Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Peipei Pang
- Life Sciences, GE Healthcare, Hangzhou, China
| | - Guoliang Shao
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China,
| |
Collapse
|
24
|
Kim HD, Kim BJ, Kim HS, Kim JH. Comparison of the morphologic criteria (RECIST) and metabolic criteria (EORTC and PERCIST) in tumor response assessments: a pooled analysis. Korean J Intern Med 2019; 34:608-617. [PMID: 29334722 PMCID: PMC6506740 DOI: 10.3904/kjim.2017.063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 07/10/2017] [Accepted: 07/19/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND/AIMS The Positron Emission Tomography Response Criteria in Solid Tumors (PERCIST) or European Organization for Research and Treatment of Cancer (EORTC) criteria are used to assess metabolic tumor responses. However, tumor responses have shown considerable discrepancies between the morphologic criteria (Response Evaluation Criteria in Solid Tumors [RECIST]) and metabolic criteria. We performed this pooled study to compare the RECIST and metabolic criteria in the assessment of tumor responses. METHODS Electronic databases were searched for eligible articles with the terms "RECIST," "PERCIST," or "EORTC criteria." The level of concordance in the tumor responses between the two criteria was estimated using κ statistics. RESULTS A total of 216 patients were collected from eight studies comparing the RECIST and EORTC criteria. The agreement of tumor responses between the two criteria was moderate (κ = 0.447). Eighty-six patients (39.8%) showed disagreement: tumor response was upgraded in 70 patients and downgraded in 16 when adopting the EORTC criteria. The EORTC criteria significantly increased the overall response rate (53% vs. 28%, p < 0.0001). The agreement of tumor responses between the RECIST and PERCIST was deemed fair (κ = 0.389). Of 407 patients from nine studies, 181 (44.5%) showed a discrepancy: using the PERCIST, tumor response were upgraded in 151 patients and downgraded in 30. When adopting the PERCIST, the overall response rate was also significantly increased from 30% to 55% (p < 0.0001). CONCLUSION This pooled analysis demonstrates that the concordance of tumor responses between the morphologic criteria and metabolic criteria is not excellent. When adopting the metabolic criteria instead of the RECIST, overall response rates were significantly increased.
Collapse
Affiliation(s)
- Hong Deok Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Seoul, Korea
| | - Bum Jun Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Seoul, Korea
| | - Hyeong Su Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Seoul, Korea
| | - Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Seoul, Korea
| |
Collapse
|
25
|
Greally M, Chou JF, Molena D, Rusch VW, Bains MS, Park BJ, Wu AJ, Goodman KA, Kelsen DP, Janjigian YY, Ilson DH, Ku GY. Positron-Emission Tomography Scan-Directed Chemoradiation for Esophageal Squamous Cell Carcinoma: No Benefit for a Change in Chemotherapy in Positron-Emission Tomography Nonresponders. J Thorac Oncol 2019; 14:540-546. [PMID: 30391577 PMCID: PMC6640852 DOI: 10.1016/j.jtho.2018.10.152] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Preoperative or definitive chemoradiation is an accepted treatment for locally advanced esophageal squamous cell carcinoma (ESCC). The MUNICON study showed that positron-emission tomography (PET) response following induction chemotherapy was predictive of outcomes in patients with gastroesophageal junction adenocarcinoma. We evaluated the predictive value of PET following induction chemotherapy in ESCC patients and assessed the impact of changing chemotherapy during radiation in PET nonresponders. METHODS We retrospectively reviewed all patients with locally advanced ESCC who received induction chemotherapy and chemoradiation; all patients had a PET before and after induction chemotherapy. Survival was calculated from date of repeat PET using Kaplan-Meier analysis and compared between groups using the log-rank test. RESULTS Of 111 patients, 70 (63%) were PET responders (defined as a 35% or more decrease in maximum standard uptake value) to induction chemotherapy. PET responders received the same chemotherapy during radiation. Of 41 PET nonresponders, 16 continued with the same chemotherapy and 25 were changed to alternative chemotherapy with radiation. Median progression-free survival (70.1 months versus 7.1 months, p < 0.01) and overall survival (84.8 months versus 17.2 months, p < 0.01) were improved for PET responders versus nonresponders. Median progression-free survival and overall survival for PET nonresponders who changed chemotherapy versus those who did not were 6.4 months versus 8.3 months (p = 0.556) and 14.1 versus 17.2 months (p = 0.81), respectively. CONCLUSIONS PET after induction chemotherapy highly predicts for outcomes in ESCC patients who receive chemoradiation. However, our results suggest that PET nonresponders do not benefit from changing chemotherapy during radiation. Future trials should use PET nonresponse after induction chemotherapy to identify poor prognosis patients for novel therapies.
Collapse
Affiliation(s)
- Megan Greally
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Joanne F Chou
- Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Valerie W Rusch
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Manjit S Bains
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bernard J Park
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Abraham J Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Karyn A Goodman
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - David P Kelsen
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yelena Y Janjigian
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David H Ilson
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Geoffrey Y Ku
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
26
|
Hynninen J, Laasik M, Vallius T, Kemppainen J, Grönroos S, Virtanen J, Casado J, Hautaniemi S, Grenman S, Seppänen M, Auranen A. Clinical Value of 18 F-fluorodeoxyglucose Positron Emission Tomography/Computed Tomography in Response Evaluation after Primary Treatment of Advanced Epithelial Ovarian Cancer. Clin Oncol (R Coll Radiol) 2018; 30:507-514. [DOI: 10.1016/j.clon.2018.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 04/09/2018] [Indexed: 01/08/2023]
|
27
|
Riyahi S, Choi W, Liu CJ, Zhong H, Wu AJ, Mechalakos JG, Lu W. Quantifying local tumor morphological changes with Jacobian map for prediction of pathologic tumor response to chemo-radiotherapy in locally advanced esophageal cancer. Phys Med Biol 2018; 63:145020. [PMID: 29911659 PMCID: PMC6064042 DOI: 10.1088/1361-6560/aacd22] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We proposed a framework to detect and quantify local tumor morphological changes due to chemo-radiotherapy (CRT) using a Jacobian map and to extract quantitative radiomic features from the Jacobian map to predict the pathologic tumor response in locally advanced esophageal cancer patients. In 20 patients who underwent CRT, a multi-resolution BSpline deformable registration was performed to register the follow-up (post-CRT) CT to the baseline CT image. The Jacobian map (J) was computed as the determinant of the gradient of the deformation vector field. The Jacobian map measured the ratio of local tumor volume change where J < 1 indicated tumor shrinkage and J > 1 denoted expansion. The tumor was manually delineated and corresponding anatomical landmarks were generated on the baseline and follow-up images. Intensity, texture and geometry features were then extracted from the Jacobian map of the tumor to quantify tumor morphological changes. The importance of each Jacobian feature in predicting pathologic tumor response was evaluated by both univariate and multivariate analysis. We constructed a multivariate prediction model by using a support vector machine (SVM) classifier coupled with a least absolute shrinkage and selection operator (LASSO) for feature selection. The SVM-LASSO model was evaluated using ten-times repeated 10-fold cross-validation (10 × 10-fold CV). After registration, the average target registration error was 4.30 ± 1.09 mm (LR:1.63 mm AP:1.59 mm SI:3.05 mm) indicating registration error was within two voxels and close to 4 mm slice thickness. Visually, the Jacobian map showed smoothly-varying local shrinkage and expansion regions in a tumor. Quantitatively, the average median Jacobian was 0.80 ± 0.10 and 1.05 ± 0.15 for responder and non-responder tumors, respectively. These indicated that on average responder tumors had 20% median volume shrinkage while non-responder tumors had 5% median volume expansion. In univariate analysis, the minimum Jacobian (p = 0.009, AUC = 0.98) and median Jacobian (p = 0.004, AUC = 0.95) were the most significant predictors. The SVM-LASSO model achieved the highest accuracy when these two features were selected (sensitivity = 94.4%, specificity = 91.8%, AUC = 0.94). Novel features extracted from the Jacobian map quantified local tumor morphological changes using only baseline tumor contour without post-treatment tumor segmentation. The SVM-LASSO model using the median Jacobian and minimum Jacobian achieved high accuracy in predicting pathologic tumor response. The Jacobian map showed great potential for longitudinal evaluation of tumor response.
Collapse
Affiliation(s)
- Sadegh Riyahi
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wookjin Choi
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chia-Ju Liu
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hualiang Zhong
- Department of Radiation Oncology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Abraham J. Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - James G. Mechalakos
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wei Lu
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
28
|
Esophageal cancer associated with a sarcoid-like reaction and systemic sarcoidosis in lymph nodes: supportive findings of [18F]-fluorodeoxyglucose positron emission tomography-computed tomography during neoadjuvant therapy. Surg Case Rep 2018; 4:62. [PMID: 29943286 PMCID: PMC6020089 DOI: 10.1186/s40792-018-0473-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/14/2018] [Indexed: 01/23/2023] Open
Abstract
Background In patients with esophageal cancer, differentiation between lymph node metastasis and lymphadenopathies from sarcoidosis or sarcoid-like reactions of lymph nodes is clinically important. Herein, we report two esophageal cancer cases with lymph node involvement of sarcoid-like reaction or sarcoidosis. Case presentation One patient received chemotherapy and the other chemoradiotherapy as initial treatments. In both cases, [18F]-fluorodeoxyglucose positron emission tomography–computed tomography (FDG-PET/CT) was performed before and after chemo(radio)therapy. After the treatment, FDG uptake was not detected in the primary tumor, but it was slightly reduced in the hilar and mediastinal lymph nodes in both cases. These non-identical responses to chemo(radio)therapy suggest the presence of sarcoid-like reaction of lymph nodes associated with squamous cell carcinoma of the esophagus. Curative surgical resection was performed as treatment. Conclusions These FDG-PET/CT findings may be helpful to distinguish between metastasis and sarcoidosis-associated lymphadenopathy in esophageal cancer.
Collapse
|
29
|
Feng L, Qi S, Lin M. Efficacy and survival rate of intensity-modulated radiotherapy combined with chemotherapy for elderly patients with locally advanced oropharyngeal cancer. Exp Ther Med 2018; 15:2475-2479. [PMID: 29467849 PMCID: PMC5792795 DOI: 10.3892/etm.2017.5682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 12/12/2017] [Indexed: 02/02/2023] Open
Abstract
The efficacy of intensity-modulated radiotherapy (IMRT) combined with chemotherapy in the treatment of elderly patients with locally advanced oropharyngeal cancer and its effect on survival rate were studied. Elderly patients (n=150) diagnosed with locally advanced oropharyngeal cancer by histopathology were selected and randomly divided into the observation group (n=75) and the control group (n=75). Patients in the observation group were treated with IMRT combined with chemotherapy, while those in the control group were treated with conventional radiotherapy and chemotherapy. The two groups were treated with docetaxel + cisplatin (TP regimen). All patients received 1 to 2 cycles of docetaxel + cisplatin-induced chemotherapy, and after the radiotherapy began, the chemotherapy with docetaxel was synchronously conducted. The recent efficacy (tumor regression condition was observed at 3 months after the treatment), 1-year, 3-year and 5-year overall survival (OS), local-regional control (LRC), progression-free survival (PFS), disease-free survival (DFS) and the incidence rate of adverse reactions of patients in the two groups were compared. In the observation group, 73 patients completed the radiotherapy and chemotherapy, while all the patients in the control group completed the treatments. The 1-year OS of the observation group and the control group was 97.3 and 85.3%, respectively. In the observation group, the 3-year LRC, OS, PFS and DFS of the observation group was 94.5, 91.8, 90.4 and 87.7%, respectively; the 5-year LRC, OS, PFS and DFS was 64.4, 56.2, 56.2 and 54.8%, respectively. In the control group, the 3-year LRC, OS, PFS and DFS was 86.7, 73.3, 82.7 and 68.0%, respectively; the 5-year LRC, OS, PFS and DFS were 54.7, 45.3, 44.0 and 56.7%, respectively. The differences were statistically significant (P<0.05). In the observation group, the number of leukocytes was decreased, and the incidence rates of acute oropharyngeal mucosa reaction and radiation dermatitis were significantly lower than those in the control group. The differences were statistically significant (P<0.05). In conclusion, IMRT combined with chemotherapy can improve the OS and the 3-year and 5-year LRC, PFS and DFS of elderly patients with locally advanced oropharyngeal cancer, reduce toxic and side effects, and improve patients' quality of life.
Collapse
Affiliation(s)
- Li Feng
- Customer Service Management Office, Shandong University Affiliated Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Shummei Qi
- Department of Stomatology, Jinan Maternity and Child Health Care Hospital, Jinan, Shandong 250001, P.R. China
| | - Ming Lin
- Department of Stomatology, Shandong University Affiliated Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
30
|
Odawara S, Kitajima K, Katsuura T, Kurahashi Y, Shinohara H, Yamakado K. Tumor response to neoadjuvant chemotherapy in patients with esophageal cancer assessed with CT and FDG-PET/CT - RECIST 1.1 vs. PERCIST 1.0. Eur J Radiol 2018; 101:65-71. [PMID: 29571803 DOI: 10.1016/j.ejrad.2018.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/09/2018] [Indexed: 10/18/2022]
Abstract
PURPOSE We compared the response classification systems Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 and Positron Emission Tomography Response Criteria in Solid Tumors (PERCIST) 1.0 for assessment of response to neoadjuvant chemotherapy in patients with esophageal cancer. MATERIALS AND METHODS Prior to planned surgical resection, 62 patients with esophageal cancer underwent fluorodeoxyglucose (FDG)-PET/CT and contrast-enhanced CT examinations before and after receiving neoadjuvant chemotherapy. Primary tumor largest diameter, maximum standardized uptake value (SUVmax), peak lean body mass SUV (SULpeak), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) were determined. Patients were divided into responders (grade 1b-3) and non-responders (grade 0-1a) according to pathological response. RESULTS Concordance between RECIST 1.1 and PERCIST 1.0 for response classification was seen in 28 (45.2%) patients. For 18 defined as responders, the number of metabolic responders (partial metabolic response + complete metabolic response) shown by PERCIST 1.0 was 17 and the number of anatomic responders (partial response + complete response) shown by RECIST 1.1 was 13. To distinguish responders from non-responders, the area under the receiver operating characteristics curve values for reduced primary tumor largest diameter, SUVmax, SULpeak, MTV, and TLG were 0.724, 0.775, 0.781, 0.756, and 0.759, respectively. An optimal percent decrease in largest diameter cut-off value of 39.2% was found to have 66.7% sensitivity and 70.5% specificity, while that for SULpeak of 55.8% was 77.8% and 75.0%, respectively. CONCLUSIONS As compared to RECIST 1.1, PERSIST 1.0 may be more suitable for evaluation of neoadjuvant therapeutic response to esophageal cancer.
Collapse
Affiliation(s)
- Soichi Odawara
- Department of Radiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Kazuhiro Kitajima
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan.
| | - Takayuki Katsuura
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan.
| | - Yasunori Kurahashi
- Division of Upper G.I. Surgery, Department of Surgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501 Japan.
| | - Hisashi Shinohara
- Division of Upper G.I. Surgery, Department of Surgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501 Japan.
| | - Koichiro Yamakado
- Department of Radiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| |
Collapse
|
31
|
O JH, Wahl RL. PERCIST in Perspective. Nucl Med Mol Imaging 2018; 52:1-4. [PMID: 29391906 PMCID: PMC5777964 DOI: 10.1007/s13139-017-0507-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/16/2017] [Accepted: 11/23/2017] [Indexed: 01/01/2023] Open
Abstract
Positron Emission tomography Response Criteria In Solid Tumors (PERCIST) version 1.0 was introduced in 2009 for objective assessment of tumor metabolic response using 18F-FDG PET/CT. Practical PERCIST: A Simplified Guide to PET Response Criteria in Solid Tumors 1.0 was published in 2016 to review and clarify some of the issues with the PERCIST. In this article, we reflect on the benefits and challenges of implementing PERCIST, and speculate on topics that could be discussed in PERCIST 1.1 in the future.
Collapse
Affiliation(s)
- Joo Hyun O
- Department of Radiology, The Catholic University of Korea, Seoul, South Korea
| | - Richard L. Wahl
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St. Louis, MO USA
| |
Collapse
|
32
|
Comparison of the RECIST and PERCIST criteria in solid tumors: a pooled analysis and review. Oncotarget 2017; 7:27848-54. [PMID: 27036043 PMCID: PMC5053692 DOI: 10.18632/oncotarget.8425] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/16/2016] [Indexed: 11/25/2022] Open
Abstract
The PET Response Criteria in Solid Tumors (PERCIST) is a new method for the quantitative assessment of metabolic changes in solid tumors. The assessments of tumor response between the RECIST and PERCIST have shown considerable difference in several studies. This pooled study was conducted to compare tumor response according to the two criteria in patients with solid tumors. We surveyed MEDLINE, EMBASE and PUBMED for articles with terms of the RECIST or PERCIST from 2009 and January 2016. There were six articles comparing the RECIST and PERCIST. A total of 268 patients were recruited; 81 with colorectal cancer, 60 with lung cancer, 48 with esophageal cancer, 28 with breast cancer, 14 with basal cell carcinoma, 12 with stomach cancer, 10 with head and neck cancer, and 16 with other rare cancers. The agreement of tumor response between the RECIST and PERCIST was moderate (k = 0.590). Of 268 patients, 101 (37.7%) showed discordance in the tumor responses between two criteria. When adopting the PERCIST, tumor response was upgraded in 85 patients and downgraded in 16. The estimated overall response rates were significantly different between two criteria (35.1% by RECIST vs. 54.1% by PERCIST, P < 0.0001). In conclusion, this pooled analysis demonstrates that the concordance of tumor responses between the RECIST and PERCIST criteria is not excellent. The PERCIST might be more suitable for assessing tumor response than the RECIST criteria.
Collapse
|
33
|
Kitajima K, Nakajo M, Kaida H, Minamimoto R, Hirata K, Tsurusaki M, Doi H, Ueno Y, Sofue K, Tamaki Y, Yamakado K. Present and future roles of FDG-PET/CT imaging in the management of gastrointestinal cancer: an update. NAGOYA JOURNAL OF MEDICAL SCIENCE 2017; 79:527-543. [PMID: 29238109 PMCID: PMC5719212 DOI: 10.18999/nagjms.79.4.527] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 09/21/2017] [Indexed: 02/08/2023]
Abstract
Positron emission tomography/computed tomography (PET/CT) integrated with 2-[18F]fluoro-2-deoxy-D-glucose (FDG) is a useful tool for acquisition of both glucose metabolism and anatomic imaging data, as only a single device and one diagnostic session is required, thus opening a new field in clinical oncologic imaging. FDG-PET/CT has been successfully used for initial staging, restaging, assessment of early treatment response, evaluation of metastatic disease response, and prognostication of intestinal cancer as well as various malignant tumors. We reviewed the current status and role of FDG-PET/CT for management of patients with esophageal cancer, gastric cancer, and colorectal cancer, with focus on both its usefulness and limitations.
Collapse
Affiliation(s)
- Kazuhiro Kitajima
- Department of Radiology, Division of Nuclear Medicine and PET Center, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masatoyo Nakajo
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Hayato Kaida
- Department of Radiology, Kinki University Faculty of Medicine, Osaka, Japan
| | - Ryogo Minamimoto
- Department of Radiology, Division of Nuclear Medicine, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kenji Hirata
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | - Hiroshi Doi
- Department of Radiation Oncology, Kinki University Faculty of Medicine, Osaka, Japan
| | - Yoshiko Ueno
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keitaro Sofue
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yukihisa Tamaki
- Department of Radiation Oncology, Shimane University School of Medicine, Izumo, Japan
| | - Koichiro Yamakado
- Department of Radiology, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
34
|
Usefulness of computed tomography density of a tumor in predicting the response of advanced esophageal cancer to preoperative chemotherapy. Surgery 2017; 162:823-835. [DOI: 10.1016/j.surg.2017.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/07/2017] [Accepted: 06/02/2017] [Indexed: 11/23/2022]
|
35
|
Goel R, Subramaniam RM, Wachsmann JW. PET/Computed Tomography Scanning and Precision Medicine: Esophageal Cancer. PET Clin 2017; 12:373-391. [PMID: 28867110 DOI: 10.1016/j.cpet.2017.05.001] [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: 12/13/2022]
Abstract
Esophageal cancer commonly has a poor prognosis, which requires an accurate diagnosis and early treatment to improve outcome. Other modalities for staging, such as endoscopic ultrasound imaging and computed tomography (CT) scans, have a role in diagnosis and staging. However, PET with fluorine-18 fluoro-2-deoxy-d-glucose/CT (FDG PET/CT) scanning allows for improved detection of distant metastatic disease and can help to prevent unnecessary interventions that would increase morbidity. FDG PET/CT scanning is valuable in the neoadjuvant chemotherapy assessment and predicting survival outcomes subsequent to surgery. FDG PET/CT scanning detects recurrent disease and metastases in follow-up.
Collapse
Affiliation(s)
- Reema Goel
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA
| | - Rathan M Subramaniam
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Department of Clinical Sciences, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA
| | - Jason W Wachsmann
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA.
| |
Collapse
|
36
|
Makino T, Yamasaki M, Tanaka K, Tatsumi M, Takiguchi S, Hatazawa J, Mori M, Doki Y. Importance of positron emission tomography for assessing the response of primary and metastatic lesions to induction treatments in T4 esophageal cancer. Surgery 2017; 162:836-845. [PMID: 28711321 DOI: 10.1016/j.surg.2017.06.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND There is no consensus strategy for treatment of T4 esophageal cancer, and because of this, a better evaluation of treatment response is crucial to establish personalized therapies. This study aimed to establish a useful system for evaluating treatment response in T4 esophageal cancer. METHODS This study included 130 patients with cT4 esophageal cancer without distant metastasis who underwent 18F-fluorodeoxyglucose-positron emission tomography before and after a series of induction treatments comprising chemoradiation or chemotherapy. We evaluated the maximal standardized uptake value and treatment response. RESULTS The mean ± standard deviation of standardized uptake value in the primary tumor before and after induction treatments were 13.8 ± 4.4 and 5.4 ± 4.1, respectively, and the mean standardized uptake value decrease was 58.4%. The most significant difference in survival between positron emission tomography-primary tumor responders and nonresponders was at a decrease of 60% standardized uptake value, based on every 10% stepwise cutoff analysis (2-year cause-specific survival: 60.2 vs 23.5%; hazard ratio = 2.705; P < .0001). With this cutoff value, the resectability (P = .0307), pathologic response (P = .0004), and pT stage (P < .0001) were associated with positron emission tomography-primary tumor response. Univariate analysis of 2-year cause-specific survival indicated a correlation between cause-specific survival and clinical stages according to TNM classification, esophageal perforation, positron emission tomography-primary tumor response, lymph node status evaluated by positron emission tomography before and after induction treatments, and operative resection. Multivariate analysis further identified positron emission tomography-primary tumor response (hazard ratio = 2.354; P = .0107), lymph node status evaluated by positron emission tomography after induction treatments (hazard ratio = 1.966; P = .0089), and operative resection (hazard ratio = 2.012; P = .0245) as independent prognostic predictors. CONCLUSION Positron emission tomography evaluation of the response of primary and metastatic lesions to induction treatments is important to formulate treatment strategies for cT4 esophageal cancer.
Collapse
Affiliation(s)
- Tomoki Makino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mitsuaki Tatsumi
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Jun Hatazawa
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
37
|
Swinburne NC, Biederman DM, Besa C, Tabori NE, Fischman AM, Patel RS, Nowakowski FS, Gunasekaran G, Schwartz ME, Lookstein RA, Kim E. Radioembolization for Unresectable Intrahepatic Cholangiocarcinoma: Review of Safety, Response Evaluation Criteria in Solid Tumors 1.1 Imaging Response and Survival. Cancer Biother Radiopharm 2017; 32:161-168. [DOI: 10.1089/cbr.2017.2189] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
| | - Derek M. Biederman
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Cecilia Besa
- Division of Body Imaging, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nora E. Tabori
- Division of Interventional Radiology, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aaron M. Fischman
- Division of Interventional Radiology, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rahul S. Patel
- Division of Interventional Radiology, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Francis Scott Nowakowski
- Division of Interventional Radiology, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ganesh Gunasekaran
- Department of Liver Surgery, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Myron E. Schwartz
- Department of Liver Surgery, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Robert A. Lookstein
- Division of Interventional Radiology, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Edward Kim
- Division of Interventional Radiology, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
38
|
Bang JI, Lim Y, Paeng JC, Han SW, Park S, Lee JM, Kim HJ, Cheon GJ, Lee DS, Chung JK, Kim TY, Kang KW. Comparison of Quantitative Methods on FDG PET/CT for Treatment Response Evaluation of Metastatic Colorectal Cancer. Nucl Med Mol Imaging 2017; 51:147-153. [PMID: 28559939 DOI: 10.1007/s13139-016-0449-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/21/2016] [Accepted: 08/30/2016] [Indexed: 11/26/2022] Open
Abstract
PURPOSE FDG PET is effective in treatment response evaluation of cancer. However, there is no standard method for quantitative evaluation of FDG PET, particularly regarding cytostatic drugs. We compared various FDG PET quantitative methods in terms of response determination. METHODS A total of 39 refractory metastatic colorectal cancer patients who received a multikinase inhibitor treatment were included. Baseline and posttreatment FDG PET/CT scans were performed before and two cycles after treatment. Standardized uptake value (SUV) and total lesion glycolysis (TLG) values using various margin thresholds (30-70 % of maximum SUV with increment 10 %, twice mean SUV of blood pool, SUV 3.0, and SUV 4.0) were measured, with measurement target of the hottest lesion or a maximum of five hottest lesions. Treatment response by the PERCIST criteria was also determined. Predictive values of the PET indexes were evaluated in terms of the treatment response determined by the RECIST 1.1 criteria. RESULTS The agreement rate was 38 % between response determined by the PERCIST and the RECIST criteria (κ = 0.381). When patients were classified into disease control group (PR, SD) and non-control group (PD) by the RECIST criteria, percent changes of TLG with various margin thresholds (particularly, 30-50 % of maximum SUV) exhibited significant differences between the two groups, and high diagnostic power for the response by the RECIST criteria. TLG-based criteria, which used a margin threshold of 50 % of maximum SUV, exhibited a high agreement with the RECIST criteria compared with the PERCIST criteria (κ = 0.606). CONCLUSION In metastatic colorectal cancer, FDG PET/CT could be effective for treatment response evaluation by using TLG measured by margin thresholds of 30-50 % of maximum SUV. Further studies are warranted regarding the optimal cutoff values for this method.
Collapse
Affiliation(s)
- Ji-In Bang
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Yoojoo Lim
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Jin Chul Paeng
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Sae-Won Han
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Sohyun Park
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Jung Min Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Hyun Joo Kim
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - June-Key Chung
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Tae-You Kim
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| |
Collapse
|
39
|
Dubreuil J, Cachin F, Berriolo-Ridinger A, Skanjeti A. Critères d’interprétation en imagerie cancérologique solide : RECIST, PERCIST…. MEDECINE NUCLEAIRE-IMAGERIE FONCTIONNELLE ET METABOLIQUE 2017. [DOI: 10.1016/j.mednuc.2017.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
40
|
Pinker K, Riedl C, Weber WA. Evaluating tumor response with FDG PET: updates on PERCIST, comparison with EORTC criteria and clues to future developments. Eur J Nucl Med Mol Imaging 2017; 44:55-66. [PMID: 28361188 DOI: 10.1007/s00259-017-3687-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 12/19/2022]
Abstract
Eighteen years ago, the EORTC PET criteria standardized for the first time response assessment by FDG PET. Response assessment by FDG PET has been further developed and refined by PERCIST (PET response criteria in solid tumors). This review describes the data underlying these two systems for assessing tumor response on FDG PET/CT. It also summarizes recent clinical studies that have compared EORTC criteria and PERCIST with each other as well as with the anatomically based "response criteria in solid tumors" (RECIST). These studies have shown that response assessment by EORTC criteria and PERCIST leads to very similar response classifications. In contrast, there are significant differences between response assessment by PERCIST and RECIST. Preliminary data also suggest that response assessment by PERCIST is better correlated with patient outcome and may be a better predictor for the effectiveness of new anti-cancer therapies than RECIST. If correct, this could have a significant impact on oncologic drug development. However, confirmation of the better predictive value of response assessment by PERCIST by data from randomized trials is still lacking.
Collapse
Affiliation(s)
- Katja Pinker
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, 1250 1st Av, New York, NY, 10065, USA
| | - Christopher Riedl
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, 1250 1st Av, New York, NY, 10065, USA
| | - Wolfgang A Weber
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, 1250 1st Av, New York, NY, 10065, USA.
| |
Collapse
|
41
|
Blum Murphy MA, Elimova E, Ajani JA. Current concepts and future potential in neoadjuvant chemotherapy for esophageal cancer. Expert Rev Gastroenterol Hepatol 2016; 10:383-92. [PMID: 26560689 DOI: 10.1586/17474124.2016.1116936] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Many trials have evaluated preoperative chemotherapy for the treatment of locally advanced esophageal cancer (LAEC). Most studies were small with conflicting results and no clear evidence of survival advantage. However, two large trials that included squamous cell carcinomas and adenocarcinomas of the esophagus produced opposite outcomes with one showing limited benefit and the other showing none. Recent meta-analyses suggest only a modest benefit from induction chemotherapy in the treatment of LAEC. Two factors associated with prolonged survival are: (1) an R0 resection and (2) pathological complete remission. Preoperative chemotherapy is preferred in Europe for adenocarcinomas; however, chemoradiation has been the treatment of choice in the US. The individualization and optimization of therapy for esophageal cancer patients may come from an in-depth understanding of molecular biology and the development of predictive biomarkers. The use of targeted and immunotherapy agents in the preoperative setting are also promising and warrant further evaluation.
Collapse
Affiliation(s)
- M A Blum Murphy
- a Department of Gastrointestinal Medical Oncology , University of Texas, MD Anderson Cancer Center , Houston , TX , USA
| | - Elena Elimova
- a Department of Gastrointestinal Medical Oncology , University of Texas, MD Anderson Cancer Center , Houston , TX , USA
| | - Jaffer A Ajani
- a Department of Gastrointestinal Medical Oncology , University of Texas, MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
42
|
Schneider BJ, Shah MA, Klute K, Ocean A, Popa E, Altorki N, Lieberman M, Schreiner A, Yantiss R, Christos PJ, Palmer R, You D, Viale A, Kermani P, Scandura JM. Phase I Study of Epigenetic Priming with Azacitidine Prior to Standard Neoadjuvant Chemotherapy for Patients with Resectable Gastric and Esophageal Adenocarcinoma: Evidence of Tumor Hypomethylation as an Indicator of Major Histopathologic Response. Clin Cancer Res 2016; 23:2673-2680. [PMID: 27836862 DOI: 10.1158/1078-0432.ccr-16-1896] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 10/05/2016] [Accepted: 10/25/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Epigenetic silencing of tumor suppressor genes (TSG) is an acquired abnormality observed in cancer and is prototypically linked to DNA methylation. We postulated that pretreatment (priming) with 5-azacitidine would increase the efficacy of chemotherapy by reactivating TSGs. This study was conducted to identify a tolerable dose of 5-azacitidine prior to EOX (epirubicin, oxaliplatin, capecitabine) neoadjuvant chemotherapy in patients with locally advanced esophageal/gastric adenocarcinoma (EGC).Experimental Design: Eligible patients had untreated, locally advanced, resectable EGC, ECOG 0-2, and adequate organ function. 5-Azacitidine (V, 75 mg/m2) was given subcutaneously for 3 (dose level, DL 1) or 5 (DL 2) days prior to each 21-day cycle of EOX (E, 50 mg/m2; O, 130 mg/m2; X, 625 mg/m2 twice daily for 21 days). Standard 3+3 methodology guided V dose escalation. DNA methylation at control and biomarker regions was measured by digital droplet, bisulfite qPCR in tumor samples collected at baseline and at resection.Results: All subjects underwent complete resection of residual tumor (R0). Three of the 12 patients (25%) achieved a surgical complete response and 5 had partial responses. The overall response rate was 67%. The most common toxicities were gastrointestinal and hematologic. Hypomethylation of biomarker genes was observed at all dose levels and trended with therapeutic response.Conclusions: Neoadjuvant VEOX was well-tolerated with significant clinical and epigenetic responses, with preliminary evidence that priming with V prior to chemotherapy may augment chemotherapy efficacy. The recommended phase II trial schedule is 5-azacitidine 75 mg/m2 for 5 days followed by EOX chemotherapy every 21 days. Clin Cancer Res; 23(11); 2673-80. ©2016 AACR.
Collapse
Affiliation(s)
- Bryan J Schneider
- Division of Hematology/Oncology, Department of Internal Medicine, Weill Cornell Medical College, New York, New York.
| | - Manish A Shah
- Division of Hematology/Oncology, Department of Internal Medicine, Weill Cornell Medical College, New York, New York
| | - Kelsey Klute
- Division of Hematology/Oncology, Department of Internal Medicine, Weill Cornell Medical College, New York, New York
| | - Allyson Ocean
- Division of Hematology/Oncology, Department of Internal Medicine, Weill Cornell Medical College, New York, New York
| | - Elizabeta Popa
- Division of Hematology/Oncology, Department of Internal Medicine, Weill Cornell Medical College, New York, New York
| | - Nasser Altorki
- Department of Thoracic Surgery, Weill Cornell Medical College, New York, New York
| | - Michael Lieberman
- Department of Surgery, Weill Cornell Medical College, New York, New York
| | - Andrew Schreiner
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Rhonda Yantiss
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Paul J Christos
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | - Romae Palmer
- Clinical Trials Office, Weill Cornell Medical College, New York, New York
| | - Daoqi You
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Agnes Viale
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Pouneh Kermani
- Division of Hematology/Oncology, Department of Internal Medicine, Weill Cornell Medical College, New York, New York
| | - Joseph M Scandura
- Division of Hematology/Oncology, Department of Internal Medicine, Weill Cornell Medical College, New York, New York
- Division of Regenerative Medicine, Department of Internal Medicine; Weill Cornell Medical College, New York, New York
| |
Collapse
|
43
|
Comparison of WHO, RECIST 1.1, EORTC, and PERCIST criteria in the evaluation of treatment response in malignant solid tumors. Nucl Med Commun 2016; 37:9-15. [PMID: 26440568 DOI: 10.1097/mnm.0000000000000401] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AIM To compare response assessment according to the WHO, RECIST 1.1, EORTC, and PERCIST criteria in patients diagnosed with malignant solid tumors and who had received cytotoxic chemotherapy to establish the strength of agreement between each criterion. MATERIALS AND METHODS Sixty patients with malignant solid tumors were included in this retrospective study. The baseline and the sequential follow-up fluorine-18-fluorodeoxyglucose PET/computed tomography (CT) of each patient were evaluated according to the WHO, RECIST 1.1, EORTC, and PERCIST criteria. PET/CT images were used for both metabolic and anatomic evaluation. The concurrent diagnostic CT and MRI images (performed within 1 week of PET/CT) were also utilized when needed. The results were compared using the κ-statistics. RESULTS The response and progression rates according to the WHO criteria were 37 and 38%, respectively. The same ratios were also found for RECIST 1.1 (κ=1). The response and progression rates according to the EORTC criteria were 47 and 40%, respectively. When PERCIST criteria were used, one patient with progressive disease was upgraded to stable disease (κ=0.976). As we found the same results with WHO and RECIST 1.1 criteria, we used WHO criteria to compare the anatomic and metabolic criteria. When we compared the WHO and EORTC criteria, there was an agreement in 80% of the patients (κ=0.711). With WHO and PERCIST criteria, there was an agreement in 81.6% of the patients (κ=0.736). CONCLUSION Significant agreement was detected when the WHO, RECIST 1.1, EORTC, and PERCIST criteria were compared both within as well as between each other.
Collapse
|
44
|
van Rossum PSN, Xu C, Fried DV, Goense L, Court LE, Lin SH. The emerging field of radiomics in esophageal cancer: current evidence and future potential. Transl Cancer Res 2016; 5:410-423. [PMID: 30687593 DOI: 10.21037/tcr.2016.06.19] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
'Radiomics' is the name given to the emerging field of extracting additional information from standard medical images using advanced feature analysis. This innovative form of quantitative image analysis appears to have future potential for clinical practice in patients with esophageal cancer by providing an additional layer of information to the standard imaging assessment. There is a growing body of evidence suggesting that radiomics may provide incremental value for staging, predicting treatment response, and predicting survival in esophageal cancer, for which the current work-up has substantial limitations. This review outlines the available evidence and future potential for the application of radiomics in the management of patients with esophageal cancer. In addition, an overview of the current evidence on the importance of reproducibility of image features and the substantial influence of varying smoothing scales, quantization levels, and segmentation methods is provided.
Collapse
Affiliation(s)
- Peter S N van Rossum
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston (Texas), USA.,Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cai Xu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston (Texas), USA.,Department of Radiation Oncology, Cancer Hospital & Institute, Chinese Academy of Medical Science, Beijing 100021, China
| | - David V Fried
- Department of Radiation Oncology, University of North Carolina, Chapel Hill (North Carolina), USA
| | - Lucas Goense
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Laurence E Court
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston (Texas), USA
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston (Texas), USA
| |
Collapse
|
45
|
O JH, Lodge MA, Wahl RL. Practical PERCIST: A Simplified Guide to PET Response Criteria in Solid Tumors 1.0. Radiology 2016; 280:576-84. [PMID: 26909647 PMCID: PMC4976461 DOI: 10.1148/radiol.2016142043] [Citation(s) in RCA: 329] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Positron Emission Tomography (PET) Response Criteria in Solid Tumors (PERCIST 1.0) describes in detail methods for controlling the quality of fluorine 18 fluorodeoxyglucose PET imaging conditions to ensure the comparability of PET images from different time points to allow quantitative expression of the changes in PET measurements and assessment of overall treatment response in PET studies. The steps for actual application of PERCIST are summarized. Several issues from PERCIST 1.0 that appear to require clarification, such as measurement of size and definition of unequivocal progression, also are addressed. (©) RSNA, 2016.
Collapse
|
46
|
Horvat TZ, Adel NG, Dang TO, Momtaz P, Postow MA, Callahan MK, Carvajal RD, Dickson MA, D'Angelo SP, Woo KM, Panageas KS, Wolchok JD, Chapman PB. Reply to A. Indini et al. J Clin Oncol 2016; 34:1018-9. [PMID: 26786917 DOI: 10.1200/jco.2015.65.7007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Troy Z Horvat
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nelly G Adel
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Thu-Oanh Dang
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Parisa Momtaz
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael A Postow
- Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Margaret K Callahan
- Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Richard D Carvajal
- Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Mark A Dickson
- Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Sandra P D'Angelo
- Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| | - Kaitlin M Woo
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center; Ludwig Institute for Cancer Research; and Weill Cornell Medical College, New York, NY
| | - Paul B Chapman
- Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY
| |
Collapse
|
47
|
Anderegg MCJ, de Groof EJ, Gisbertz SS, Bennink RJ, Lagarde SM, Klinkenbijl JHG, Dijkgraaf MGW, Bergman JJGHM, Hulshof MCCM, van Laarhoven HWM, van Berge Henegouwen MI. 18F-FDG PET-CT after Neoadjuvant Chemoradiotherapy in Esophageal Cancer Patients to Optimize Surgical Decision Making. PLoS One 2015; 10:e0133690. [PMID: 26529313 PMCID: PMC4631456 DOI: 10.1371/journal.pone.0133690] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 07/01/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Prognosis of esophageal cancer patients can be significantly improved by neoadjuvant chemoradiotherapy (nCRT). Given the aggressive nature of esophageal tumors, it is conceivable that in a significant portion of patients treated with nCRT, dissemination already becomes manifest during the period of nCRT. The aim of this retrospective study was to determine the value and diagnostic accuracy of PET-CT after neoadjuvant chemoradiotherapy to identify patients with metastases preoperatively in order to prevent non-curative surgery. METHODS From January 2011 until February 2013 esophageal cancer patients deemed eligible for a curative approach with nCRT and surgical resection underwent a PET-CT after completion of nCRT. If abnormalities on PET-CT were suspected metastases, histological proof was acquired. A clinical decision model was designed to assess the cost-effectiveness of this diagnostic strategy. RESULTS 156 patients underwent a PET-CT after nCRT. In 31 patients (19.9%) PET-CT showed abnormalities suspicious for dissemination, resulting in 17 cases of proven metastases (10.9%). Of the patients without proven metastases 133 patients were operated. In 6 of these 133 cases distant metastases were detected intraoperatively, corresponding to 4.5% false-negative results. The standard introduction of a post-neoadjuvant therapy PET-CT led to a reduction of overall health care costs per patient compared to a scenario without restaging with PET-CT ($34,088 vs. $36,490). CONCLUSION In 10.9% of esophageal cancer patients distant metastases were detected by standard PET-CT after neoadjuvant chemoradiotherapy. To avoid non-curative resections we advocate post-neoadjuvant therapy PET-CT as a cost-effective step in the standard work-up of candidates for surgery.
Collapse
Affiliation(s)
| | | | | | - Roel J. Bennink
- Nuclear Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Sjoerd M. Lagarde
- Department of Surgery, Academic Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
48
|
Miyata H, Yamasaki M, Makino T, Tatsumi M, Miyazaki Y, Takahashi T, Kurokawa Y, Takiguchi S, Mori M, Doki Y. Impact of number of [(18)F]fluorodeoxyglucose-PET-positive lymph nodes on survival of patients receiving neoadjuvant chemotherapy and surgery for oesophageal cancer. Br J Surg 2015; 103:97-104. [PMID: 26506079 DOI: 10.1002/bjs.9965] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/24/2015] [Accepted: 09/04/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND [(18) F]fluorodeoxyglucose (FDG)-PET has been used to evaluate the response of primary tumours to neoadjuvant therapy for oesophageal cancer. The clinical significance of the number of PET-positive nodes before and after therapy has not been investigated previously. METHODS [(18) F]FDG-PET was performed before and 2-3 weeks after completion of neoadjuvant chemotherapy to identify the number of PET-positive nodes, and these numbers were assessed in relation to metabolic changes in the primary tumour. RESULTS Of 302 patients in total, 90 had no PET-positive nodes, 83 had one, 59 had two and 70 patients had three or more positive nodes before therapy. After treatment, the numbers were: none in 207 patients, one in 59, two in 20 and three or more in 16 patients. The number of PET-positive nodes after treatment was influenced by both the number of PET-positive nodes before therapy and the response to preoperative therapy, and correlated with the number of metastatic lymph nodes. Overall survival was longer in patients who had no PET-positive nodes after treatment than in those who had one or more. Multivariable analysis identified the numbers of PET-positive nodes before and after chemotherapy as independent prognostic factors, together with clinical response, tumour depth and lymph node involvement. CONCLUSION The number of PET-positive nodes after treatment correlated with survival in patients with oesophageal cancer who underwent neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- H Miyata
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Surgery, Osaka Medical Centre for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - M Yamasaki
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - T Makino
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - M Tatsumi
- Departments of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Y Miyazaki
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - T Takahashi
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Y Kurokawa
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - S Takiguchi
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - M Mori
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Y Doki
- Departments of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
49
|
Zabor EC, Heller G, Schwartz LH, Chapman PB. Correlating Surrogate Endpoints with Overall Survival at the Individual Patient Level in BRAFV600E-Mutated Metastatic Melanoma Patients Treated with Vemurafenib. Clin Cancer Res 2015; 22:1341-7. [PMID: 26490313 DOI: 10.1158/1078-0432.ccr-15-1441] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/11/2015] [Indexed: 01/22/2023]
Abstract
PURPOSE Surrogate endpoints are needed that correlate with overall survival (OS). We analyzed individual patient tumor data from a phase III trial of vemurafenib versus dacarbazine (BRIM3) to identify criteria for tumor measures that correlated with OS. Correlates were validated using a separate data set from a phase II trial of vemurafenib (BRIM2). EXPERIMENTAL DESIGN Deidentified tumor measurements and OS data from BRIM3 and from BRIM2 were analyzed. Target tumor measurement data and nontarget tumor data were available from pretreatment, weeks 6,12, and every 9 weeks thereafter. In the BRIM3 data set, associations of OS with both early tumor response (first 12 weeks) and time to progression (TTP) were assessed. Different definitions of response and progression were explored. Findings were validated using the BRIM2 data set. RESULTS Thresholds of early response were explored ranging from any degree of tumor shrinkage to 100% tumor shrinkage. Correlation was weak at all thresholds tested. TTP, however, was more strongly correlated with OS. The strongest correlation was seen when progression was defined as ≥50% increase in the sum of tumor diameters or appearance of new tumors. This was confirmed by similar analyses in the BRIM2 cohort. CONCLUSIONS TTP defined as ≥50% increase in the sum of tumor diameters or appearance of new tumors was more strongly associated with OS than early tumor shrinkage in melanoma patients treated with RAF inhibitor. In future trials, consideration should be given to replacing response rate with TTP or PFS as preferable clinical endpoints in early-phase studies.
Collapse
Affiliation(s)
- Emily C Zabor
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Glenn Heller
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University College of Physicians and Surgeons and New York Presbyterian Hospital, New York, New York
| | - Paul B Chapman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
50
|
Yamasaki M, Miyata H, Miyazaki Y, Takahashi T, Kurokawa Y, Nakajima K, Takiguchi S, Mori M, Doki Y. Pattern of Lymphatic Spread of Esophageal Cancer at the Cervicothoracic Junction Based on the Tumor Location : Surgical Treatment of Esophageal Squamous Cell Carcinoma of the Cervicothoracic Junction. Ann Surg Oncol 2015; 22 Suppl 3:S750-7. [PMID: 26350372 DOI: 10.1245/s10434-015-4855-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND There is no consensus about the extent of lymphadenectomy for patients with esophageal squamous cell carcinoma at the cervicothoracic junction (CT-ESCC). The purpose of this study was to examine the pattern of lymph node spread in patients with CT-ESCC and the extent of lymphadenectomy that is necessary. METHODS We included 64 consecutive patients with CT-ESCC who underwent surgery. All patients were divided into two groups based on the location of the epicenter or anal edge of the primary tumor. Using the height of the epicenter, 27 and 37 patients were classified as having cervical-centered and thoracic-centered tumors, respectively; while, using the height of the anal edge, 38 and 26 patients had tumors that were cervical-localized and thoracic-invading, respectively. RESULTS In the patients with cervical-centered tumors, the incidences of metastasis and/or recurrences in the cervical paraesophageal, supraclavicular, and upper mediastinal nodes were 21.4-28.5 %. No patient had metastasis or recurrence in the middle and lower mediastinal and perigastric nodes. In patients with thoracic-centered tumors, the lymph node metastasis and/or recurrence spread to the cervical paraesophageal (41.7 %), supraclavicular (25 %), and upper mediastinal (55.6 %) nodes, as well as the middle (22.2 %) and lower mediastinal (8.3 %) and perigastric (19.4 %) nodes. There was no difference in the distribution and incidence of lymphatic spread between patients with the cervical-localized and thoracic-invading classifications. CONCLUSIONS Our results indicate a cervical and upper mediastinal lymphadenectomy is better indicated for patients with cervical-centered CT-ESCC, whereas patients with thoracic-centered CT-ESCC should be treated with a three-field lymphadenectomy.
Collapse
Affiliation(s)
- Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Hiroshi Miyata
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasuhiro Miyazaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|