1
|
Bond MJG, Mijnals C, Bolhuis K, van Amerongen MJ, Engelbrecht MRW, Hermans JJ, van Lienden KP, May AM, Swijnenburg RJ, Punt CJA. Prognostic value of radiologic and pathological response in colorectal cancer liver metastases upon systemic induction treatment: subgroup analysis of the CAIRO5 trial. ESMO Open 2024; 9:104075. [PMID: 39667310 PMCID: PMC11697041 DOI: 10.1016/j.esmoop.2024.104075] [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: 08/27/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND RECIST may not be optimal for assessing treatment response with current systemic regimens. We evaluated RECIST, morphologic, and pathologically documented response (pathological response) in patients with initially unresectable colorectal cancer liver-only metastases (CRLM). PATIENTS AND METHODS Four hundred and eighty-nine patients from the phase III CAIRO5 trial were included who were treated with FOLFOX/FOLFIRI/FOLFOXIRI and bevacizumab or panitumumab. The association of the different response tools with overall survival (OS) was evaluated for all patients, and with early recurrence (<6 months) for patients after complete local treatment. RESULTS In the overall population, suboptimal [hazard ratio (HR) 1.10, 95% confidence interval (CI) 0.83-1.47] and optimal (HR 0.95, 95% CI 0.74-1.22) morphologic response were not associated with OS compared with no response. RECIST partial response (HR 0.61, 95% CI 0.49-0.76) and progressive disease (HR 5.77, 95% CI 3.97-8.39) were associated with OS compared with stable disease. In 242 patients who underwent local treatment, suboptimal (HR 1.22, 95% CI 0.76-1.96) and optimal (HR 1.28, 95% CI 0.89-1.86) morphologic response were not associated with OS compared with no response. RECIST partial response was not significantly associated with OS (HR 0.73, 95% CI 0.52-1.01), whereas progressive disease was (HR 19.74, 95% CI 5.75-67.78), compared with stable disease. While major pathological response (HR 0.66, 95% CI 0.44-0.99) was associated with OS, partial pathological response (HR 0.82, 95% CI 0.57-1.19) was not, compared with no pathological response. Pathological response, but not morphologic response and RECIST, was significantly associated with early recurrence (P < 0.001) which occurred in 13/58 (22%) patients with major response, 29/61 (48%) patients with partial response, and 51/88 (58%) patients with no response. CONCLUSIONS Our results show that RECIST but not morphologic response was prognostic for OS. In patients eligible for local treatment, neither RECIST nor morphologic response were associated with early recurrence. Pathological response was associated with early recurrence but is only available post-operatively. Hence, novel preoperative parameters are warranted to predict early recurrence and prevent potentially futile liver surgery.
Collapse
Affiliation(s)
- M J G Bond
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | - C Mijnals
- Department of Pathology, OLVG, Amsterdam, The Netherlands
| | - K Bolhuis
- Department of Gastrointestinal Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M J van Amerongen
- Department of Radiology, Sint Maartenskliniek, Nijmegen, The Netherlands
| | - M R W Engelbrecht
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| | - J J Hermans
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - K P van Lienden
- Department of Radiology, Sint Antonius Hospital, Nieuwegein, The Netherlands
| | - A M May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R-J Swijnenburg
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - C J A Punt
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
2
|
Ni X, Wu J, Pan J, Li X, Fang B, Wei Y, Ye D, Liang F, Zhu Y. Heterogeneity of Radiological Progression Patterns and Association with Outcomes in Patients with Metastatic Prostate Cancer. Eur Urol Oncol 2024; 7:897-905. [PMID: 38151441 DOI: 10.1016/j.euo.2023.11.019] [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: 10/04/2023] [Revised: 10/30/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND With an increasing number of clinical trials using radiographic progression-free survival (rPFS) instead of overall survival as the primary study endpoint, the heterogeneity of different radiological progression patterns in rPFS and postprogression survival (PPS) remains unclear. OBJECTIVE Herein, we investigate the proportion of various radiological progression patterns in patients with metastatic hormone-sensitive prostate cancer (mHSPC) and metastatic castration-resistant prostate cancer (mCRPC), and further explore the differences in rPFS and PPS between patients exhibiting single- or multicategory progression patterns. DESIGN, SETTING, AND PARTICIPANTS This post hoc, retrospective secondary analysis was based on individual patient data from LATITUDE (phase 3 randomized mHSPC study) and COU-AA-302 (phase 3 randomized mCRPC study). Patients with complete imaging follow-up data and radiological progression were included in the analysis. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The rPFS and PPS in LATITUDE and COU-AA-302 were evaluated. The proportion of patients exhibiting each progression pattern was calculated, and a survival analysis was conducted using the Kaplan-Meier method. RESULTS AND LIMITATIONS Of the 489 mHSPC patients studied, 366 experienced single-category progression, while the remaining 123 patients (25.2%) exhibited simultaneous occurrence of different progressive events (multicategory radiological progression). Of the 534 mCRPC patients studied, 390 experienced single-category progression, while the remaining 144 patients (27.0%) experienced multicategory progressive events. Among mCRPC patients, the rPFS of bone progression was the shortest. In contrast, among mHSPC patients, the rPFS of target lesion enlargement is the shortest, followed by bone progression. Notably, patients experiencing a single-category progression pattern displayed comparable rPFS to but significantly longer PPS than those experiencing multicategory progression patterns (PPS mHSPC cohort: 21.5 vs 6.9 mo, p < 0.0001; mCRPC cohort: 23.6 vs 15.7 mo, p < 0.0001). The study is limited by its hypothesis-generating nature. Therefore, the observed phenomena in our research necessitate validation through future prospective studies. CONCLUSIONS Patients who experience multicategory radiological progression represent a significant proportion, accounting for approximately 25% of all men with mHSPC or mCRPC. Patients with multicategory radiological progression patterns had similar rPFS to but significantly shorter PPS than those experiencing single-category progression patterns. In future clinical trials and clinical practice, radiological progression patterns should be recognized as a crucial determinant of prognosis, while also serving as the stratification or inclusion criteria for second-line treatment clinical trials. PATIENT SUMMARY In this study, we observed that among men with metastatic prostate cancer, those who experienced two or more radiological events during a single visit had a worse prognosis than those who experienced isolated radiological events.
Collapse
Affiliation(s)
- Xudong Ni
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Jian Pan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Fei Liang
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, China; Clinical Research Unit, Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China.
| |
Collapse
|
3
|
Tabari A, D’Amore B, Noh J, Gee MS, Daye D. Quantitative peritumoral magnetic resonance imaging fingerprinting improves machine learning-based prediction of overall survival in colorectal cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:74-84. [PMID: 38464383 PMCID: PMC10918231 DOI: 10.37349/etat.2024.00205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 12/28/2023] [Indexed: 03/12/2024] Open
Abstract
Aim To investigate magnetic resonance imaging (MRI)-based peritumoral texture features as prognostic indicators of survival in patients with colorectal liver metastasis (CRLM). Methods From 2007-2015, forty-eight patients who underwent MRI within 3 months prior to initiating treatment for CRLM were identified. Clinicobiological prognostic variables were obtained from electronic medical records. Ninety-four metastatic hepatic lesions were identified on T1-weighted post-contrast images and volumetrically segmented. A total of 112 radiomic features (shape, first-order, texture) were derived from a 10 mm region surrounding each segmented tumor. A random forest model was applied, and performance was tested by receiver operating characteristic (ROC). Kaplan-Meier analysis was utilized to generate the survival curves. Results Forty-eight patients (male:female = 23:25, age 55.3 years ± 18 years) were included in the study. The median lesion size was 25.73 mm (range 8.5-103.8 mm). Microsatellite instability was low in 40.4% (38/94) of tumors, with Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutation detected in 68 out of 94 (72%) tumors. The mean survival was 35 months ± 21 months, and local disease progression was observed in 35.5% of patients. Univariate regression analysis identified 42 texture features [8 first order, 5 gray level dependence matrix (GLDM), 5 gray level run time length matrix (GLRLM), 5 gray level size zone matrix (GLSZM), 2 neighboring gray tone difference matrix (NGTDM), and 17 gray level co-occurrence matrix (GLCM)] independently associated with metastatic disease progression (P < 0.03). The random forest model achieved an area under the curve (AUC) of 0.88. Conclusions MRI-based peritumoral heterogeneity features may serve as predictive biomarkers for metastatic disease progression and patient survival in CRLM.
Collapse
Affiliation(s)
- Azadeh Tabari
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Brian D’Amore
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Janice Noh
- Department of informatics, Boston University, Boston, MA 02114, USA
| | - Michael S. Gee
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Dania Daye
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Mang A, Zou W, Rolny V, Reck M, Cigoianu D, Schulze K, Holdenrieder S, Socinski MA, Shames DS, Wehnl B, Patil NS. Combined use of CYFRA 21-1 and CA 125 predicts survival of patients with metastatic NSCLC and stable disease in IMpower150. Tumour Biol 2024; 46:S177-S190. [PMID: 37545290 DOI: 10.3233/tub-230001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Patients with non-small cell lung cancer (NSCLC) and stable disease (SD) have an unmet clinical need to help guide early treatment adjustments. OBJECTIVE To evaluate the potential of tumor biomarkers to inform on survival outcomes in NSCLC SD patients. METHODS This post hoc analysis included 480 patients from the IMpower150 study with metastatic NSCLC, treated with chemotherapy, atezolizumab and bevacizumab combinations, who had SD at first CT scan (post-treatment initiation). Patients were stratified into high- and low-risk groups (overall survival [OS] and progression-free survival [PFS] outcomes) based on serum tumor biomarker levels. RESULTS The CYFRA 21-1 and CA 125 biomarker combination predicted OS and PFS in patients with SD. Risk of death was ~4-fold higher for the biomarker-stratified high-risk versus low-risk SD patients (hazard ratio [HR] 3.80; 95% confidence interval [CI] 3.02-4.78; p < 0.0001). OS in patients with the low- and high-risk SD was comparable to that in patients with the CT-defined partial response (PR; HR 1.10; 95% CI 0.898-1.34) and progressive disease (PD) (HR 1.05; 95% CI 0.621-1.77), respectively. The findings were similar with PFS, and consistent across treatment arms. CONCLUSIONS Biomarker testing shows potential for providing prognostic information to help direct treatment in NSCLC patients with SD. Prospective clinical studies are warranted.ClinicalTrials.gov: NCT02366143.
Collapse
Affiliation(s)
- Anika Mang
- Roche Diagnostics GmbH, Penzberg, Germany
| | - Wei Zou
- Oncology Biomarkers Development, Genentech, San Francisco, CA, USA
| | | | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany
| | | | - Katja Schulze
- Oncology Biomarkers Development, Genentech, San Francisco, CA, USA
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | | | - David S Shames
- Oncology Biomarkers Development, Genentech, San Francisco, CA, USA
| | | | - Namrata S Patil
- Oncology Biomarkers Development, Genentech, San Francisco, CA, USA
| |
Collapse
|
5
|
Abbas E, Fanni SC, Bandini C, Francischello R, Febi M, Aghakhanyan G, Ambrosini I, Faggioni L, Cioni D, Lencioni RA, Neri E. Delta-radiomics in cancer immunotherapy response prediction: A systematic review. Eur J Radiol Open 2023; 11:100511. [PMID: 37520768 PMCID: PMC10371799 DOI: 10.1016/j.ejro.2023.100511] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023] Open
Abstract
Background The new immunotherapies have not only changed the oncological therapeutic approach but have also made it necessary to develop new imaging methods for assessing the response to treatment. Delta radiomics consists of the analysis of radiomic features variation between different medical images, usually before and after therapy. Purpose This review aims to evaluate the role of delta radiomics in the immunotherapy response assessment. Methods A systematic search was performed in PubMed, Scopus, and Web Of Science using "delta radiomics AND immunotherapy" as search terms. The included articles' methodological quality was measured using the Radiomics Quality Score (RQS) tool. Results Thirteen articles were finally included in the systematic review. Overall, the RQS of the included studies ranged from 4 to 17, with a mean RQS total of 11,15 ± 4,18 with a corresponding percentage of 30.98 ± 11.61 %. Eleven articles out of 13 performed imaging at multiple time points. All the included articles performed feature reduction. No study carried out prospective validation, decision curve analysis, or cost-effectiveness analysis. Conclusions Delta radiomics has been demonstrated useful in evaluating the response in oncologic patients undergoing immunotherapy. The overall quality was found law, due to the lack of prospective design and external validation. Thus, further efforts are needed to bring delta radiomics a step closer to clinical implementation.
Collapse
Affiliation(s)
- Engy Abbas
- The Joint Department of Medical Imaging, University of Toronto, University Health Network, Sinai Health System, Women’s College Hospital, 610 University Ave, Toronto, ON, Canada M5G 2M9
| | | | - Claudio Bandini
- Department of Translational Research, Academic Radiology, University of Pisa, Pisa, Italy
| | - Roberto Francischello
- Department of Translational Research, Academic Radiology, University of Pisa, Pisa, Italy
| | - Maria Febi
- Department of Translational Research, Academic Radiology, University of Pisa, Pisa, Italy
| | - Gayane Aghakhanyan
- Department of Translational Research, Academic Radiology, University of Pisa, Pisa, Italy
| | - Ilaria Ambrosini
- Department of Translational Research, Academic Radiology, University of Pisa, Pisa, Italy
| | - Lorenzo Faggioni
- Department of Translational Research, Academic Radiology, University of Pisa, Pisa, Italy
| | - Dania Cioni
- Department of Translational Research, Academic Radiology, University of Pisa, Pisa, Italy
| | | | - Emanuele Neri
- The Joint Department of Medical Imaging, University of Toronto, University Health Network, Sinai Health System, Women’s College Hospital, 610 University Ave, Toronto, ON, Canada M5G 2M9
- Department of Translational Research, Academic Radiology, University of Pisa, Pisa, Italy
| |
Collapse
|
6
|
Nishino M, Wang X, Ricciuti B, Tseng SC, Park H, Alessi JV, Vaz VR, Hatabu H, Lin X, Christiani DC, Awad MM. Advanced non-small-cell lung cancer treated with first-line pembrolizumab plus chemotherapy: tumor response dynamics as a marker for survival. Eur Radiol 2023; 33:7284-7293. [PMID: 37099174 PMCID: PMC10896107 DOI: 10.1007/s00330-023-09658-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 04/27/2023]
Abstract
OBJECTIVES The study investigated tumor burden dynamics on computed tomography (CT) scans in patients with advanced non-small-cell lung cancer (NSCLC) during first-line pembrolizumab plus chemotherapy, to provide imaging markers for overall survival (OS). METHODS The study included 133 patients treated with first-line pembrolizumab plus platinum-doublet chemotherapy. Serial CT scans during therapy were assessed for tumor burden dynamics during therapy, which were studied for the association with OS. RESULTS There were 67 responders, with overall response rate of 50%. The tumor burden change at the best overall response ranged from - 100.0% to + 132.1% (median of - 30%). Higher response rates were associated with younger age (p < 0.001) and higher programmed cell death-1 (PD-L1) expression levels (p = 0.01). Eighty-three patients (62%) showed tumor burden below the baseline burden throughout therapy. Using an 8-week landmark analysis, OS was longer in patients with tumor burden below the baseline burden in the first 8 weeks than in those who experienced ≥ 0% increase (median OS: 26.8 vs. 7.6 months, hazard ratio (HR): 0.36, p < 0.001). Tumor burden remained below their baseline throughout therapy was associated with significantly reduced hazards of death (HR: 0.72, p = 0.03) in the extended Cox models, after adjusting for other clinical variables. Pseudoprogression was noted in only one patient (0.8%). CONCLUSIONS Tumor burden staying below the baseline burden throughout the therapy was predictive of prolonged overall survival in patients with advanced NSCLC treated with first-line pembrolizumab plus chemotherapy, and may be used as a practical marker for therapeutic decisions in this widely used combination regimen. CLINICAL RELEVANCE STATEMENT The analysis of tumor burden dynamics on serial CT scans in reference to the baseline burden can provide an additional objective guide for treatment decision making in patients treated with first-line pembrolizumab plus chemotherapy for their advanced NSCLC. KEY POINTS • Tumor burden remaining below baseline burden during therapy predicted longer survival during first-line pembrolizumab plus chemotherapy. • Pseudoprogression was noted in 0.8%, demonstrating the rarity of the phenomenon. • Tumor burden dynamics may serve as an objective marker for treatment benefit to guide treatment decisions during first-line pembrolizumab plus chemotherapy.
Collapse
Affiliation(s)
- Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital and Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Ave., MA, 02215, Boston, USA.
| | - Xinan Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - Biagio Ricciuti
- Department of Medical Oncology and Department of Medicine, Dana-Farber Cancer Institute and Brigham and Women's Hospital, 450 Brookline Ave., Boston, MA, 02215, USA
| | - Shu-Chi Tseng
- Department of Radiology, Brigham and Women's Hospital and Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Ave., MA, 02215, Boston, USA
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Hyesun Park
- Department of Radiology, Brigham and Women's Hospital and Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Ave., MA, 02215, Boston, USA
| | - Joao V Alessi
- Department of Medical Oncology and Department of Medicine, Dana-Farber Cancer Institute and Brigham and Women's Hospital, 450 Brookline Ave., Boston, MA, 02215, USA
| | - Victor R Vaz
- Department of Medical Oncology and Department of Medicine, Dana-Farber Cancer Institute and Brigham and Women's Hospital, 450 Brookline Ave., Boston, MA, 02215, USA
| | - Hiroto Hatabu
- Department of Radiology, Brigham and Women's Hospital and Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Ave., MA, 02215, Boston, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
- Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02115, USA
| | - Mark M Awad
- Department of Medical Oncology and Department of Medicine, Dana-Farber Cancer Institute and Brigham and Women's Hospital, 450 Brookline Ave., Boston, MA, 02215, USA
| |
Collapse
|
7
|
Ansari G, Mirza-Aghazadeh-Attari M, Mohseni A, Madani SP, Shahbazian H, Pawlik TM, Kamel IR. Response Assessment of Primary Liver Tumors to Novel Therapies: an Imaging Perspective. J Gastrointest Surg 2023; 27:2245-2259. [PMID: 37464140 DOI: 10.1007/s11605-023-05762-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/11/2023] [Indexed: 07/20/2023]
Abstract
The latest developments in cancer immunotherapy, namely the introduction of immune checkpoint inhibitors, have led to a fundamental change in advanced cancer treatments. Imaging is crucial to identify tumor response accurately and delineate prognosis in immunotherapy-treated patients. Simultaneously, advances in image acquisition techniques, notably functional and molecular imaging, have facilitated more accurate pretreatment evaluation, assessment of response to therapy, and monitoring for tumor recurrence. Traditional approaches to assessing tumor progression, such as RECIST, rely on changes in tumor size, while new strategies for evaluating tumor response to therapy, such as the mRECIST and the EASL, rely on tumor enhancement. Moreover, the assessment of tumor volume, enhancement, cellularity, and perfusion are some novel techniques that have been investigated. Validation of these novel approaches should rely on comparing their results with those of standard evaluation methods (EASL, mRECIST) while considering the ultimate outcome, which is patient survival. More recently, immunotherapy has been used in the management of primary liver tumors. However, little is known about its efficacy. This article reviews imaging modalities and techniques for assessing tumor response and survival in immunotherapy-treated patients with primary hepatic malignancies.
Collapse
Affiliation(s)
- Golnoosh Ansari
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Mohammad Mirza-Aghazadeh-Attari
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Alireza Mohseni
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Seyedeh Panid Madani
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Haneyeh Shahbazian
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA
| | - Timothy M Pawlik
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, James Comprehensive Cancer Center, Columbus, OH, USA
| | - Ihab R Kamel
- Russell H. Morgan Department of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, MRI 143, Baltimore, MD, 21287, USA.
| |
Collapse
|
8
|
Li Y, Flavell RR, Juarez R, Chow M, Wu C, Tsai K, Daud A, Behr SC. Retrospective study of the incidence of sarcoidosis-like reaction in patients treated with immunotherapy. Clin Radiol 2023; 78:e131-e136. [PMID: 36344282 DOI: 10.1016/j.crad.2022.09.127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/06/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
AIM To assess the frequency of radiographically evident drug-induced sarcoidosis-like reaction (DISR) in patients treated with anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) therapy, anti-programmed cell death protein 1 (PD-1) therapy, or a combination of both in a single centre. MATERIALS AND METHODS The images and medical records of 457 patients with metastatic melanoma or head and neck cancer treated with either anti-CTLA-4 therapy, anti-PD-1 therapy, or a combination of both at University of California medical centre were reviewed retrospectively and the incidence of radiological manifestations of DISR was assessed among these treatment groups. RESULTS Radiological manifestations of DISR were found in 19/457 patients (4.1%). The mean interval from the initiation of immunotherapy to development of DISR was 5.5 months (range 2.3-13.5 months). Mean interval from radiological detection of DISR to imaging evidence of resolution was 5.8 months (range 1.6-18.3 months). Three patients out of 81 (3.7%), 11/297 (3.7%), and 5/79 (6.3%) developed sarcoidosis-like reaction after treatment with anti-CTLA-4 antibody, anti-PD-1 antibody, and a combination of both, respectively. Most patients with DISR were asymptomatic and did not require systemic therapy. Most patients did not demonstrate concomitant increased maximum standardised uptake value (SUVmax) in other organs on their integrated 2-[18F]-fluoro-2-deoxy-d-glucose (FDG) positron-emission tomography (PET)/computed tomography (CT). CONCLUSIONS In the present retrospective study of patients treated with immune checkpoint inhibitors (ICIs), DISR occurred in approximately 3.7% of patients treated with either anti-CTLA-4 or anti-PD-1 antibody and 6.3% of patients treated with a combination of both.
Collapse
Affiliation(s)
- Y Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, USA
| | - R R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, USA
| | - R Juarez
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, USA
| | - M Chow
- Department of Medicine, University of California, San Francisco, 1825 5(th) St, San Francisco, CA 94143, USA
| | - C Wu
- Department of Medicine, University of California, San Francisco, 1825 5(th) St, San Francisco, CA 94143, USA
| | - K Tsai
- Department of Medicine, University of California, San Francisco, 1825 5(th) St, San Francisco, CA 94143, USA
| | - A Daud
- Department of Medicine, University of California, San Francisco, 1825 5(th) St, San Francisco, CA 94143, USA
| | - S C Behr
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, USA.
| |
Collapse
|
9
|
Parsonidis P, Beis G, Iliopoulos AC, Papasotiriou I. Adoptive transfer of activated immune cells against solid tumors: A preliminary study. Cell Immunol 2022; 382:104616. [PMID: 36219944 DOI: 10.1016/j.cellimm.2022.104616] [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: 07/14/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND This study presents preliminary results concerning the effectiveness of a novel immunotherapy in cancer. The proposed adoptive cellular therapy product contains a mixture of effector immune cells, specifically macrophages, NK cells, dendritic cells, cytotoxic T lymphocytes and monoclonal antibody producing plasma cells. METHODS The results were based on both descriptive and inferential statistical analysis of data concerning 17 cancer patients. Particularly, performance scales such as clinical condition, Karnofsky-Index, ECOG index and symptom's scale were evaluated post therapy administration (4 months). Furthermore, circulating tumor cells (CTCs) and a specific tumor marker (EpCAM) were measured pre- and post-cellular therapy. RESULTS The results revealed a positive evaluation for clinical condition (70.59 %), Karnofsky-Index (88.23 %), ECOG index (94.12 %), and symptoms' scale (64.70 %). In addition, statistically significant reductions were found for both CTCs (p = 0.0016) and EpCAM positive cells (p = 0.0005), post-therapy, which were related to large size effects, namely 0.77 and 0.85, respectively. No cytokine storm, anaphylaxis or severe adverse events were observed with 4 months follow up evaluation. CONCLUSIONS These preliminary results indicate that the proposed cellular therapy can be considered for further studies in clinical trials.
Collapse
Affiliation(s)
| | - Georgios Beis
- Research Genetic Cancer Centre S.A., Florina, Greece
| | | | | |
Collapse
|
10
|
Monitoring of Current Cancer Therapy by Positron Emission Tomography and Possible Role of Radiomics Assessment. Int J Mol Sci 2022; 23:ijms23169394. [PMID: 36012657 PMCID: PMC9409366 DOI: 10.3390/ijms23169394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/31/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Evaluation of cancer therapy with imaging is crucial as a surrogate marker of effectiveness and survival. The unique response patterns to therapy with immune-checkpoint inhibitors have facilitated the revision of response evaluation criteria using FDG-PET, because the immune response recalls reactive cells such as activated T-cells and macrophages, which show increased glucose metabolism and apparent progression on morphological imaging. Cellular metabolism and function are critical determinants of the viability of active cells in the tumor microenvironment, which would be novel targets of therapies, such as tumor immunity, metabolism, and genetic mutation. Considering tumor heterogeneity and variation in therapy response specific to the mechanisms of therapy, appropriate response evaluation is required. Radiomics approaches, which combine objective image features with a machine learning algorithm as well as pathologic and genetic data, have remarkably progressed over the past decade, and PET radiomics has increased quality and reliability based on the prosperous publications and standardization initiatives. PET and multimodal imaging will play a definitive role in personalized therapeutic strategies by the precise monitoring in future cancer therapy.
Collapse
|
11
|
Liu Y, Ni M, Li L, Wang J, Tu Z, Zhou H, Zhang S. A novel four-gene signature predicts immunotherapy response of patients with different cancers. J Clin Lab Anal 2022; 36:e24494. [PMID: 35588138 PMCID: PMC9279975 DOI: 10.1002/jcla.24494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/10/2022] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) therapy has demonstrated favorable clinical efficacy, particularly for advanced or difficult-to-treat cancer types. However, this therapy is ineffective for many patients displaying lack of immune response or resistance to ICB. This study aimed to establish a novel four-gene signature (CD8A, CD8B, TCF7, and LEF1) to provide a prognostic immunotherapy biomarker for different cancers. METHODS Transcriptome profiles and clinical data were obtained from The Cancer Genome Atlas database. Multivariate Cox regression analysis was used to establish a four-gene signature. The R package estimate was used to obtain the immune score for every patient. RESULTS Risk scores of the novel four-gene signature could effectively divided all patients into high- and low-risk groups, with distinct outcomes. The immune score calculated via the estimate package demonstrated that the four-gene signature was significantly associated with the immune infiltration level. Furthermore, the four-gene signature could predict the response to atezolizumab immunotherapy in patients with metastatic urothelial cancer. CONCLUSIONS The novel four-gene signature developed in this study is a good prognostic biomarker, as it could identify many kinds of patients with cancer who are likely to respond to and benefit from immunotherapy.
Collapse
Affiliation(s)
- Yuanli Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Mingyue Ni
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Lamei Li
- Department of DermatologyAnhui Provincial Hospital Affiliated to Anhui Medical UniversityHefeiChina
| | - Junyan Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
- Department of Clinical Medicine (5+3 Programme)Anhui Medical UniversityHefeiChina
| | - Zhenzhen Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Haisheng Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Siping Zhang
- Department of DermatologyAnhui Provincial Hospital Affiliated to Anhui Medical UniversityHefeiChina
| |
Collapse
|
12
|
Liu WL, Zhang YQ, Li LT, Zhu YY, Ming ZH, Chen WL, Yang RQ, Li RH, Chen M, Zhang GJ. Application of molecular imaging in immune checkpoints therapy: From response assessment to prognosis prediction. Crit Rev Oncol Hematol 2022; 176:103746. [PMID: 35752425 DOI: 10.1016/j.critrevonc.2022.103746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/30/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Recently, immune checkpoint therapy (ICT) represented by programmed cell death1 (PD-1) and its major ligands, programmed death ligand 1 (PD-L1), has achieved significant success. Detection of PD-L1 by immunohistochemistry (IHC) is a classic method to guide the treatment of ICT patients. However, PD-L1 expression in the tumor microenvironment is highly complex. Thus, PD-L1 IHC is inadequate to fully understand the relevance of PD-L1 levels in the whole body and their dynamics to improve therapeutic outcomes. Intriguingly, numerous studies have revealed that molecular imaging technologies could potentially meet this need. Therefore, the purpose of this narrative review is to summarize the preclinical and clinical application of ICT guided by molecular imaging technology, and to explore the future opportunities and practical difficulties of these innovations.
Collapse
Affiliation(s)
- Wan-Ling Liu
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Yong-Qu Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Liang-Tao Li
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Yuan-Yuan Zhu
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Zi-He Ming
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Wei-Ling Chen
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Rui-Qin Yang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China
| | - Rong-Hui Li
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Department of Medical Oncology, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China
| | - Min Chen
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China.
| | - Guo-Jun Zhang
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer (Xiang'an Hospital of Xiamen University), 2000 East Xiang'an Road, Xiamen, China; Xiamen Key Laboratory for Endocrine Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, 2000 East Xiang'an Road, Xiamen, China; Xiamen Research Center of Clinical Medicine in Breast & Thyroid Cancers, 2000 East Xiang'an Road, Xiamen, China; Cancer Research Center, School of Medicine, Xiamen University, 4221 South Xiang'an Road, Xiamen, China.
| |
Collapse
|
13
|
Papasotiriou I, Beis G, Iliopoulos AC, Apostolou P. Supportive Oligonucleotide Therapy (SOT) as an Alternative Treatment Option in Cancer: A Preliminary Study. In Vivo 2022; 36:898-906. [PMID: 35241548 DOI: 10.21873/invivo.12779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM An early evaluation concerning the effectiveness of supportive oligonucleotide therapy (SOT) in cancer as a monotherapy and in combination with other types of treatment. PATIENTS AND METHODS This study evaluated the clinical condition and performance status (Karnofsky-Index) of 95 patients, post-SOT administration. Furthermore, circulating tumor cells (CTCs) from 47 patients' pre- and post-SOT administration were measured and analyzed by repeated-measures ANOVA. RESULTS Improvement of the clinical condition was observed in all patients who used SOT (77.89%), SOT in combination with other therapy (69.77%) and SOT as a monotherapy or no information was given concerning another therapy (84.31%). Positive results for Karnofsky-Index were also observed in 71.58%, 61.36%, and 80.39%, respectively. Finally, statistically significant reductions in CTCs were observed for both SOT as a monotherapy and SOT as an adjunctive therapy. CONCLUSION The preliminary results indicate that SOT therapy can be used both as monotherapy as well as in combination with other therapies for cancer.
Collapse
Affiliation(s)
| | - Georgios Beis
- Research Genetic Cancer Centre S.A., Industrial Area of Florina, Florina, Greece
| | - Aggelos C Iliopoulos
- Research Genetic Cancer Centre S.A., Industrial Area of Florina, Florina, Greece
| | - Panagiotis Apostolou
- Research Genetic Cancer Centre S.A., Industrial Area of Florina, Florina, Greece
| |
Collapse
|
14
|
PET imaging of lung and pleural cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00206-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
15
|
Papasotiriou I, Hatzidaki E. Personalized dendritic cell vaccination in cancer therapy: An evidence-based research study. J Cancer Res Ther 2022; 19:S52-S58. [PMID: 37147983 DOI: 10.4103/jcrt.jcrt_522_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Although chemotherapy is considered to be the golden standard, it does not come without a price. Toxicities and resistance are frequently limiting its effectiveness. Immunotherapy has emerged as a safer therapeutic alternative but still has a long way until it has proven to be of equal efficacy. A type of immunotherapy is dendritic cell (DC) vaccination. Aims and Objectives We have developed a novel platform for the generation of autologous DCs that have been activated against peptides that are personalized for each patient individually. The aim of the study was to clinically evaluate this platform. Materials and Methods Our platform and our algorithm for the determination of the immunogenic peptides has been tested. DC generation was verified both morphologically and by CD80/86 expression. Peptide antigenicity was determined using a number of T-cell epitope prediction algorithms. Response to therapy was evaluated using response evaluation criteria in solid tumors (RECIST) criteria by the doctors involved. Immune status was also evaluated before and after DC vaccination and correlated with circulated tumor cell count. Results It was found that DC vaccine increased immune activation while correlated with decreased circulating tumor cell counts. Clinical evaluation by the determination of immune markers may be a superior tool than using RECIST criteria. Conclusion Dendritic cell therapies could prove to be a valuable tool in cancer treatment.
Collapse
|
16
|
Evaluation of Response to Immune Checkpoint Inhibitors Using a Radiomics, Lesion-Level Approach. Cancers (Basel) 2021; 13:cancers13236050. [PMID: 34885160 PMCID: PMC8657103 DOI: 10.3390/cancers13236050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/28/2021] [Accepted: 11/28/2021] [Indexed: 11/17/2022] Open
Abstract
Conventional methods to determine the response to immune checkpoint inhibitors (ICIs) are limited by the unique responses to an ICI. We performed a radiomics approach for all measurable lesions to identify radiomic variables that could distinguish hyperprogressive disease (HPD) on baseline CT scans and classify a dissociated response (DR). One hundred and ninety-six patients with advanced lung cancer, treated with ICI monotherapy, who underwent at least three CT scans, were retrospectively enrolled. For all 621 measurable lesions, HPDv was determined from baseline CT scans using the tumor growth kinetics (TGK) ratio, and radiomics features were extracted. Multivariable logistic regression analysis of radiomics features was performed to discriminate DR. Radiomics features that significantly discriminated HPDv on baseline CT differed according to organ. Of the 196 patients, 54 (27.6%) had a DR and 142 (72.4%) did not have a DR. Overall survival in the group with a DR was significantly inferior to that in the group without a DR (log rank test, p = 0.04). Our study shows that lesion-level analysis using radiomics features has great potential for discriminating HPDv and understanding heterogeneous tumor progression, including a DR, after ICI treatment.
Collapse
|
17
|
Nishino M, Lu J, Hino T, Vokes NI, Jänne PA, Hatabu H, Johnson BE. Tumor Growth Rate After Nadir Is Associated With Survival in Patients With EGFR-Mutant Non-Small-Cell Lung Cancer Treated With Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor. JCO Precis Oncol 2021; 5:1603-1610. [PMID: 34994646 PMCID: PMC9848598 DOI: 10.1200/po.21.00172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/24/2021] [Accepted: 09/09/2021] [Indexed: 01/25/2023] Open
Abstract
PURPOSE To investigate the association between tumor volume growth rate after the nadir and survival in patients with EGFR-mutant advanced non-small-cell lung cancer (NSCLC) treated with erlotinib. MATERIALS AND METHODS Seventy-one patients with EGFR-mutant advanced NSCLC treated with erlotinib were studied for computed tomography tumor volume kinetics during therapy. The tumor growth rate after nadir was obtained using a previously published analytic module for longitudinal volume tracking to study its relationship with overall survival (OS). RESULTS The median tumor volume for the cohort was 19,842 mm3 at baseline and 4,083 mm3 at nadir. The median time to nadir was 6.2 months. The tumor growth rate after nadir for logeV (the natural logarithm of tumor volume measured in mm3) was 0.11/mo on average for the cohort (SE: 0.014), which was very similar to the previously validated reference value of 0.12/mo to define slow and fast tumor growth. The OS of 48 patients with slow tumor growth (≤ 0.12/mo) was significantly longer compared with 23 patients with fast tumor growth (> 0.12/mo; median OS: 37.8 v 25.0 months; P = .0012). In Cox models, tumor growth rate was also associated with survival (regression coefficient: 3.9903; P = .0024; faster rate leads to increased hazards), after adjusting for time to nadir (regression coefficient: -0.0863; P = .0008; longer time to nadir leads to decreased hazards) and smoking history. CONCLUSION In patients with EGFR-mutant advanced NSCLC treated with erlotinib, slower tumor growth rates after nadir were associated with longer OS, providing a rationale for using tumor growth rates to guide precision therapy for lung cancer.
Collapse
Affiliation(s)
- Mizuki Nishino
- Department of Imaging, Dana Farber Cancer
Institute, Boston, MA
- Department of Radiology, Brigham and
Women's Hospital, Boston, MA
| | - Junwei Lu
- Department of Biostatistics, Harvard Chan
School of Public Health, Boston, MA
| | - Takuya Hino
- Department of Radiology, Brigham and
Women's Hospital, Boston, MA
| | - Natalie I. Vokes
- Department of Medical Oncology, Dana
Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and
Women's Hospital, Boston, MA
| | - Pasi A. Jänne
- Department of Medical Oncology, Dana
Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and
Women's Hospital, Boston, MA
| | - Hiroto Hatabu
- Department of Imaging, Dana Farber Cancer
Institute, Boston, MA
- Department of Radiology, Brigham and
Women's Hospital, Boston, MA
| | - Bruce E. Johnson
- Department of Medical Oncology, Dana
Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and
Women's Hospital, Boston, MA
| |
Collapse
|
18
|
Nishino M, Lu J, Hino T, Vokes NI, Jänne PA, Hatabu H, Johnson BE. Tumor Growth Rate After Nadir Is Associated With Survival in Patients With EGFR-Mutant Non-Small-Cell Lung Cancer Treated With Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor. JCO Precis Oncol 2021. [PMID: 34994646 DOI: 10.1200/po.20.00478:501-509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
PURPOSE To investigate the association between tumor volume growth rate after the nadir and survival in patients with EGFR-mutant advanced non-small-cell lung cancer (NSCLC) treated with erlotinib. MATERIALS AND METHODS Seventy-one patients with EGFR-mutant advanced NSCLC treated with erlotinib were studied for computed tomography tumor volume kinetics during therapy. The tumor growth rate after nadir was obtained using a previously published analytic module for longitudinal volume tracking to study its relationship with overall survival (OS). RESULTS The median tumor volume for the cohort was 19,842 mm3 at baseline and 4,083 mm3 at nadir. The median time to nadir was 6.2 months. The tumor growth rate after nadir for logeV (the natural logarithm of tumor volume measured in mm3) was 0.11/mo on average for the cohort (SE: 0.014), which was very similar to the previously validated reference value of 0.12/mo to define slow and fast tumor growth. The OS of 48 patients with slow tumor growth (≤ 0.12/mo) was significantly longer compared with 23 patients with fast tumor growth (> 0.12/mo; median OS: 37.8 v 25.0 months; P = .0012). In Cox models, tumor growth rate was also associated with survival (regression coefficient: 3.9903; P = .0024; faster rate leads to increased hazards), after adjusting for time to nadir (regression coefficient: -0.0863; P = .0008; longer time to nadir leads to decreased hazards) and smoking history. CONCLUSION In patients with EGFR-mutant advanced NSCLC treated with erlotinib, slower tumor growth rates after nadir were associated with longer OS, providing a rationale for using tumor growth rates to guide precision therapy for lung cancer.
Collapse
Affiliation(s)
- Mizuki Nishino
- Department of Imaging, Dana Farber Cancer Institute, Boston, MA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA
| | - Junwei Lu
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA
| | - Takuya Hino
- Department of Radiology, Brigham and Women's Hospital, Boston, MA
| | - Natalie I Vokes
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Pasi A Jänne
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Hiroto Hatabu
- Department of Imaging, Dana Farber Cancer Institute, Boston, MA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA
| | - Bruce E Johnson
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
19
|
Collin A, Groza V, Missenard L, Chomy F, Colin T, Palussière J, Saut O. A Model-Strengthened Imaging Biomarker for Survival Prediction in EGFR-Mutated Non-small-cell Lung Carcinoma Patients Treated with Tyrosine Kinase Inhibitors. Bull Math Biol 2021; 83:68. [PMID: 33966172 DOI: 10.1007/s11538-021-00902-7] [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: 11/19/2020] [Accepted: 04/19/2021] [Indexed: 11/25/2022]
Abstract
Non-small-cell lung carcinoma is a frequent type of lung cancer with a bad prognosis. Depending on the stage and genomics, several therapeutical approaches are used. Tyrosine Kinase Inhibitors (TKI) may be successful for a time in the treatment of EGFR-mutated non-small cells lung carcinoma. Our objective is here to introduce a survival assessment as their efficacy in the long run is challenging to evaluate. The study includes 17 patients diagnosed with EGFR-mutated non-small cell lung cancer and exposed to an EGFR-targeting TKI with 3 computed tomography (CT) scans of the primary tumor (one before the TKI introduction and two after). An imaging biomarker based on evolution of texture heterogeneity between the first and the third exams is derived and computed from a mathematical model and patient data. Defining the overall survival as the time between the introduction of the TKI treatment and the patient death, we obtain a statistically significant correlation between the overall survival and our imaging marker ([Formula: see text]). Using the ROC curve, the patients are separated into two populations and the comparison of the survival curves is statistically significant ([Formula: see text]). The baseline exam seems to have a significant role in the prediction of response to TKI treatment. More precisely, our imaging biomarker defined using only the CT scan before the TKI introduction allows to determine a first classification of the population which is improved over time using the imaging marker as soon as more CT scans are available. This exploratory study leads us to think that it is possible to obtain a survival assessment using only few CT scans of the primary tumor.
Collapse
Affiliation(s)
- Annabelle Collin
- Univ. Bordeaux, CNRS, Bordeaux INP, IMB, UMR 5251, INRIA Monc, 33400, Talence, France
| | - Vladimir Groza
- Univ. Bordeaux, CNRS, Bordeaux INP, IMB, UMR 5251, INRIA Monc, 33400, Talence, France
| | | | | | - Thierry Colin
- Sophia Genetics, Cité de la Photonique, 33600, Pessac, France
| | | | - Olivier Saut
- Univ. Bordeaux, CNRS, Bordeaux INP, IMB, UMR 5251, INRIA Monc, 33400, Talence, France.
| |
Collapse
|
20
|
Greater reductions in blood flow after anti-angiogenic treatment in non-small cell lung cancer patients are associated with shorter progression-free survival. Sci Rep 2021; 11:6805. [PMID: 33762653 PMCID: PMC7991665 DOI: 10.1038/s41598-021-86405-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
To evaluate tumor blood flow using 15O-water positron emission tomography (PET) in patients with non-small cell lung cancer (NSCLC) before and after chemotherapy with bevacizumab, and to investigate the effects of bevacizumab on tumor blood flow changes and progression-free survival (PFS). Twelve patients with NSCLC were enrolled. Six patients underwent chemotherapy with bevacizumab and the other six without bevacizumab. 15O-water dynamic PET scans were performed within 1 week before the start of chemotherapy and within 1 week after the first day of chemotherapy. Tumor blood flow was analyzed quantitatively using a single one-tissue compartment model with the correction of pulmonary circulation blood volume and arterial blood volume via an image-derived input function. In the bevacizumab group, mean tumor blood flow was statistically significantly reduced post-chemotherapy (pre-chemotherapy 0.27 ± 0.14 mL/cm3/min, post-chemotherapy 0.18 ± 0.12 mL/cm3/min). In the no bevacizumab group, there was no significant difference between mean tumor perfusion pre-chemotherapy (0.42 ± 0.42 mL/cm3/min) and post-chemotherapy (0.40 ± 0.27 mL/cm3/min). In the bevacizumab group, there was a positive correlation between the blood flow ratio (tumor blood flow post-chemotherapy/tumor blood flow pre-chemotherapy) and PFS (correlation coefficient 0.94). Mean tumor blood flow decreases after bevacizumab administration and was positively correlated with longer PFS.
Collapse
|
21
|
Nishino M, Hong F, Ricciuti B, Hatabu H, Awad MM. Tumor Response Dynamics During First-Line Pembrolizumab Therapy in Patients With Advanced Non-Small-Cell Lung Cancer. JCO Precis Oncol 2021; 5:PO.20.00478. [PMID: 34250409 DOI: 10.1200/po.20.00478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/29/2021] [Accepted: 02/16/2021] [Indexed: 12/26/2022] Open
Abstract
The objectives of the study were to characterize the tumor burden dynamics on serial computed tomography scans in patients with advanced non-small-cell lung cancer treated with first-line pembrolizumab and to identify imaging markers for prolonged overall survival (OS). MATERIALS AND METHODS Eighty-eight patients treated with first-line pembrolizumab monotherapy were evaluated on serial computed tomography scans to characterize their quantitative tumor burden during therapy. Tumor burden dynamics were studied for the association with OS. RESULTS The overall response rate was 42% (37/88), with the median tumor burden changes at the best overall response of -18.3% (range, -100.0% to +103.6%). Response rates were higher in men than in women (P = .05) and in patients with higher programmed cell death ligand-1 expression levels (P = .02). Tumor burden stayed below the baseline burden throughout therapy in 55 patients (63%). In an 8-week landmark analysis, patients with tumor burden below the baseline burden during the first 8 weeks of therapy had longer OS compared with patients who had ≥ 0% increase (median OS, 30.7 v 16.2 months; hazard ratio [HR] = 0.44; P = .01). In the extended Cox models, patients whose tumor burden stayed below the baseline burden throughout therapy had significantly reduced hazards of death (HR = 0.41, P = .003, univariate; HR = 0.35, P = .02, multivariate). Only one patient (1.1%) experienced pseudoprogression with initial tumor increase and subsequent tumor regression. CONCLUSION In patients with advanced non-small-cell lung cancer treated with first-line single-agent pembrolizumab, tumor burden reduction below the baseline burden during therapy was an independent marker for prolonged OS, which may serve as a practical guide for treatment decisions.
Collapse
Affiliation(s)
- Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital, Boston, MA.,Department of Imaging, Dana-Farber Cancer Institute, Boston, MA
| | - Fangxin Hong
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Biagio Ricciuti
- Department of Medical Oncology and Department of Medicine, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, MA
| | - Hiroto Hatabu
- Department of Radiology, Brigham and Women's Hospital, Boston, MA.,Department of Imaging, Dana-Farber Cancer Institute, Boston, MA
| | - Mark M Awad
- Department of Medical Oncology and Department of Medicine, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
22
|
Metabolic Fingerprinting of Murine L929 Fibroblasts as a Cell-Based Tumour Suppressor Model System for Methionine Restriction. Int J Mol Sci 2021; 22:ijms22063039. [PMID: 33809777 PMCID: PMC8002350 DOI: 10.3390/ijms22063039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/27/2022] Open
Abstract
Since Otto Warburg reported in 1924 that cancer cells address their increased energy requirement through a massive intake of glucose, the cellular energy level has offered a therapeutic anticancer strategy. Methionine restriction (MetR) is one of the most effective approaches for inducing low-energy metabolism (LEM) due to the central position in metabolism of this amino acid. However, no simple in vitro system for the rapid analysis of MetR is currently available, and this study establishes the murine cell line L929 as such a model system. L929 cells react rapidly and efficiently to MetR, and the analysis of more than 150 different metabolites belonging to different classes (amino acids, urea and tricarboxylic acid cycle (TCA) cycles, carbohydrates, etc.) by liquid chromatography/mass spectrometry (LC/MS) defines a metabolic fingerprint and enables the identification of specific metabolites representing normal or MetR conditions. The system facilitates the rapid and efficient testing of potential cancer therapeutic metabolic targets. To date, MS studies of MetR have been performed using organisms and yeast, and the current LC/MS analysis of the intra- and extracellular metabolites in the murine cell line L929 over a period of 5 days thus provides new insights into the effects of MetR at the cellular metabolic level.
Collapse
|
23
|
Management of Pulmonary Nodules in Oncologic Patients: AJR Expert Panel Narrative Review. AJR Am J Roentgenol 2020; 216:1423-1431. [PMID: 33355489 DOI: 10.2214/ajr.20.24907] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cancer survivors are at higher risk than the general population for development of a new primary malignancy, most commonly lung cancer. Current lung cancer screening guidelines recommend low-dose chest CT for high-risk individuals, including patients with a history of cancer and a qualifying smoking history. However, major lung cancer screening trials have inconsistently included cancer survivors, and few studies have assessed management of lung nodules in this population. This narrative review highlights relevant literature and provides expert opinion for management of pulmonary nodules detected incidentally or by screening in oncologic patients. In patients with previously treated lung cancer, a new nodule most likely represents distant metastasis from the initial lung cancer or a second primary lung cancer; CT features such as nodule size and composition should guide decisions regarding biopsy, PET/CT, and CT surveillance. In patients with extrapulmonary cancers, nodule management requires individualized risk assessment; smoking is associated with increased odds of primary lung cancer, whereas specific primary cancer types are associated with increased odds of pulmonary metastasis. Nonneoplastic causes, such as infection, medication toxicity, and postradiation or postsurgical change, should also be considered. Future prospective studies are warranted to provide evidence-based data to assist clinical decision-making in this context.
Collapse
|
24
|
Tumor Volume Analysis as a Predictive Marker for Prolonged Survival in Anaplastic Lymphoma Kinase-rearranged Advanced Non-Small Cell Lung Cancer Patients Treated With Crizotinib. J Thorac Imaging 2020; 35:101-107. [PMID: 30985604 DOI: 10.1097/rti.0000000000000413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE Targeted inhibition of anaplastic lymphoma kinase (ALK) has been widely used for the treatment of advanced non-small cell lung cancer (NSCLC) with ALK rearrangements. We performed tumor volume analysis of ALK-rearranged advanced NSCLC treated with crizotinib to identify an early predictive marker for prolonged survival. MATERIALS AND METHODS Cases of 42 patients with ALK-rearranged advanced NSCLC (16 men, 26 women; median age: 55.7 y) treated with crizotinib as their first ALK-directed therapy were retrospectively studied. Tumor volume measurements of dominant lung lesions were performed on baseline computed tomography and follow-up computed tomography at 8 weeks of therapy. The relationships between the 8-week volume change (%) and overall survival (OS) were investigated. RESULTS The 8-week tumor volume change ranged from -99.3% to 117.5% (median: -57.7%). Using the 25th percentile of the 8-week volume change of -74%, 11 patients with >74% volume decrease at 8 weeks had a significantly longer OS compared with 31 patients with ≤74% decrease (median OS: 92.0 vs. 22.8 mo; P=0.0048). In multivariable analyses using Cox proportional hazards models, the 8-week volume decrease of >74% was significantly associated with longer OS (hazard ratio=0.14, 95% confidence interval: 0.03-0.59; Cox P=0.008) after adjusting for tumor stage (stage IV vs. recurrent NSCLC, hazard ratio=5.6, 95% confidence interval: 1.29-24.3; P=0.02). CONCLUSIONS The 8-week tumor volume decrease of >74% is significantly associated with longer OS in patients with ALK-rearranged NSCLC treated with crizotinib.
Collapse
|
25
|
Hu-Lieskovan S, Bhaumik S, Dhodapkar K, Grivel JCJB, Gupta S, Hanks BA, Janetzki S, Kleen TO, Koguchi Y, Lund AW, Maccalli C, Mahnke YD, Novosiadly RD, Selvan SR, Sims T, Zhao Y, Maecker HT. SITC cancer immunotherapy resource document: a compass in the land of biomarker discovery. J Immunother Cancer 2020; 8:e000705. [PMID: 33268350 PMCID: PMC7713206 DOI: 10.1136/jitc-2020-000705] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
Since the publication of the Society for Immunotherapy of Cancer's (SITC) original cancer immunotherapy biomarkers resource document, there have been remarkable breakthroughs in cancer immunotherapy, in particular the development and approval of immune checkpoint inhibitors, engineered cellular therapies, and tumor vaccines to unleash antitumor immune activity. The most notable feature of these breakthroughs is the achievement of durable clinical responses in some patients, enabling long-term survival. These durable responses have been noted in tumor types that were not previously considered immunotherapy-sensitive, suggesting that all patients with cancer may have the potential to benefit from immunotherapy. However, a persistent challenge in the field is the fact that only a minority of patients respond to immunotherapy, especially those therapies that rely on endogenous immune activation such as checkpoint inhibitors and vaccination due to the complex and heterogeneous immune escape mechanisms which can develop in each patient. Therefore, the development of robust biomarkers for each immunotherapy strategy, enabling rational patient selection and the design of precise combination therapies, is key for the continued success and improvement of immunotherapy. In this document, we summarize and update established biomarkers, guidelines, and regulatory considerations for clinical immune biomarker development, discuss well-known and novel technologies for biomarker discovery and validation, and provide tools and resources that can be used by the biomarker research community to facilitate the continued development of immuno-oncology and aid in the goal of durable responses in all patients.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Kavita Dhodapkar
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | - Sumati Gupta
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Brent A Hanks
- Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Yoshinobu Koguchi
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Amanda W Lund
- Oregon Health and Science University, Portland, Oregon, USA
| | | | | | | | | | - Tasha Sims
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | | |
Collapse
|
26
|
Tumor volume dynamics and tumor growth rate in ALK-rearranged advanced non-small-cell lung cancer treated with crizotinib. Eur J Radiol Open 2020; 7:100210. [PMID: 33102632 PMCID: PMC7569410 DOI: 10.1016/j.ejro.2019.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/12/2019] [Accepted: 12/15/2019] [Indexed: 01/16/2023] Open
Abstract
Purpose The purpose of the study is to investigate volumetric tumor burden dynamics and tumor growth rates in ALK-rearranged advanced NSCLC patients during crizotinib monotherapy. Methods The study included 44 ALK-rearranged advanced NSCLC patients treated with crizotinib monotherapy as their initial ALK-directed therapy, who had at least one measurable lung lesion and at least two follow-up CT scans, and experienced tumor volume increase while on crizotinib. The tumor volume (in mm3) of the dominant lung lesion was measured on serial CT scans during therapy for analysis of tumor growth rates after the volume nadir. Results A total of 231 volume measurements from the nadir to the end of crizotinib therapy or the last follow-up in 44 patients were analyzed in a linear mixed-effects model, fitting time (in months since baseline) as a random effect. When measured from the volume nadir, the tumor growth rate of the logarithm of tumor volume (logeV) was 0.04/month (SE = 0.012, P = 0.0011) in the unadjusted model. When adjusted for the baseline volume (logeV0), the growth rate was again 0.04/month (SE = 0.011, P = 0.0004). When adjusted for clinical variables and logeV0, the growth rate was 0.045/month (SE = 0.012, P = 0.0002), indicating that the tumor growth rate after nadir in this cohort remains very close to 0.04/month regardless of logeV0 or clinical factors. Conclusions Tumor volume growth rate after nadir in ALK-rearranged NSCLC patients treated with crizotinib was obtained, providing objective reference values that can inform physicians when deciding to keep their patients on ALK directed therapy with slowly progressing lung cancer.
Collapse
|
27
|
Ji Z, Cui Y, Peng Z, Gong J, Zhu HT, Zhang X, Li J, Lu M, Lu Z, Shen L, Sun YS. Use of Radiomics to Predict Response to Immunotherapy of Malignant Tumors of the Digestive System. Med Sci Monit 2020; 26:e924671. [PMID: 33077705 PMCID: PMC7586759 DOI: 10.12659/msm.924671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Despite the promising results of immunotherapy in cancer treatment, new response patterns, including pseudoprogression and hyperprogression, have been observed. Radiomics is the automated extraction of high-fidelity, high-dimensional imaging features from standard medical images, allowing comprehensive visualization and characterization of the tissue of interest and corresponding microenvironment. This study assessed whether radiomics can predict response to immunotherapy in patients with malignant tumors of the digestive system. MATERIAL AND METHODS Computed tomography (CT) images of patients with malignant tumors of the digestive system obtained at baseline and after immunotherapy were subjected to radiomics analyses. Radiomics features were extracted from each image. The formula of the screened features and the final predictive model were obtained using the Least Absolute Shrinkage and Selection Operator (LASSO) algorithm. RESULTS Imaging analysis was feasible in 87 patients, including 3 with pseudoprogression and 7 with hyperprogression. One hundred ten radiomics features were obtained before and after treatment, including 109 features of the target lesions and 1 of the aorta. Four models were constructed, with the model constructed from baseline and post-treatment CT features having the best classification performance, with a sensitivity, specificity, and AUC of 83.3%, 88.9%, and 0.806, respectively. CONCLUSIONS Radiomics can predict the response of patients with malignant tumors of the digestive system to immunotherapy and can supplement conventional evaluations of response.
Collapse
Affiliation(s)
- Zhi Ji
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Yong Cui
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Hai-Tao Zhu
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Ming Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| | - Ying-Shi Sun
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China (mainland)
| |
Collapse
|
28
|
Positron Emission Tomography for Response Evaluation in Microenvironment-Targeted Anti-Cancer Therapy. Biomedicines 2020; 8:biomedicines8090371. [PMID: 32972006 PMCID: PMC7556039 DOI: 10.3390/biomedicines8090371] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/31/2022] Open
Abstract
Therapeutic response is evaluated using the diameter of tumors and quantitative parameters of 2-[18F] fluoro-2-deoxy-d-glucose positron emission tomography (FDG-PET). Tumor response to molecular-targeted drugs and immune checkpoint inhibitors is different from conventional chemotherapy in terms of temporal metabolic alteration and morphological change after the therapy. Cancer stem cells, immunologically competent cells, and metabolism of cancer are considered targets of novel therapy. Accumulation of FDG reflects the glucose metabolism of cancer cells as well as immune cells in the tumor microenvironment, which differs among patients according to the individual immune function; however, FDG-PET could evaluate the viability of the tumor as a whole. On the other hand, specific imaging and cell tracking of cancer cell or immunological cell subsets does not elucidate tumor response in a complexed interaction in the tumor microenvironment. Considering tumor heterogeneity and individual variation in therapeutic response, a radiomics approach with quantitative features of multimodal images and deep learning algorithm with reference to pathologic and genetic data has the potential to improve response assessment for emerging cancer therapy.
Collapse
|
29
|
Abstract
In the past decade, tumour flare reaction (TFR) was considered as a new side effect associated with immunomodulatory agents (IMiDs) and as a condition of chronic lymphocytic leukaemia (CLL). However, this phenomenon is also observed with immune checkpoint inhibitors in solid tumours. It is still poorly understood and its incidence is underestimated. TFR has been associated with morbidity, therefore, early recognition and management of patients with TFR is critical. An exhaustive literature research between 1985 and 2016 was performed using PubMed; American Society of Clinical Oncology and American Society of Hematology abstracts reporting TFR or pseudoprogression were identified. The incidence of TFR in CLL ranged from 28 to 58%. Tumour response in patients treated beyond progression was reported in 9.7% with ipilimumab, 10% with nivolumab, 6.7 and 12% with pembrolizumab, and in renal cell carcinoma 69% with nivolumab. Rare life-threatening or fatal cases were reported; symptoms were usually mild. Studies showed that treating patients beyond progression yielded tumour responses, considering TFR as predictive of response. Treatment with immunomodulatory agents is associated with TFR, often misinterpreted as progression. Therefore, the identification of appropriate clinical benefit criteria and the use of immune-related response criteria in prospective trials for a better understanding are compulsory.
Collapse
|
30
|
Nell E, Ober C, Rendahl A, Forrest L, Lawrence J. Volumetric tumor response assessment is inefficient without overt clinical benefit compared to conventional, manual veterinary response assessment in canine nasal tumors. Vet Radiol Ultrasound 2020; 61:592-603. [PMID: 32702179 DOI: 10.1111/vru.12895] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/27/2020] [Accepted: 05/07/2020] [Indexed: 02/04/2023] Open
Abstract
Accurate assessment of tumor response to therapy is critical in guiding management of veterinary oncology patients and is most commonly performed using response evaluation criteria in solid tumors criteria. This process can be time consuming and have high intra- and interobserver variability. The primary aim of this serial measurements, secondary analysis study was to compare manual linear tumor response assessment to semi-automated, contoured response assessment in canine nasal tumors. The secondary objective was to determine if tumor measurements or clinical characteristics, such as stage, would correlate to progression-free interval. Three investigators evaluated paired CT scans of skulls of 22 dogs with nasal tumors obtained prior to and following radiation therapy. The automatically generated tumor volumes were not useful for canine nasal tumors in this study, characterized by poor intraobserver agreement between automatically generated contours and hand-adjusted contours. The radiologist's manual linear method of determining response evaluation criteria in solid tumors categorization and tumor volume is significantly faster (P < .0001) but significantly underestimates nasal tumor volume (P < .05) when compared to a contour-based method. Interobserver agreement was greater for volume determination using the contour-based method when compared to response evaluation criteria in solid tumors categorization utilizing the same method. However, response evaluation criteria in solid tumors categorization and percentage volume change were strongly correlated, providing validity to response evaluation criteria in solid tumors as a rapid method of tumor response assessment for canine nasal tumors. No clinical characteristics or tumor measurements were significantly associated with progression-free interval.
Collapse
Affiliation(s)
- Esther Nell
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, USA
| | - Christopher Ober
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, USA
| | - Aaron Rendahl
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, USA
| | - Lisa Forrest
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, USA
| |
Collapse
|
31
|
Dual EGFR blockade with cetuximab and erlotinib combined with anti-VEGF antibody bevacizumab in advanced solid tumors: a phase 1 dose escalation triplet combination trial. Exp Hematol Oncol 2020; 9:7. [PMID: 32337094 PMCID: PMC7171918 DOI: 10.1186/s40164-020-00159-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 02/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background Angiogenesis and activation of the epidermal growth factor (EGFR) pathway play an essential role in tumor proliferation and metastasis. Targeting angiogenesis or EGFR alone does not yield adequate tumor control in most solid tumors. Overcoming intrinsic and/or acquired resistance may need a doublet or triplet therapy strategy. Herein, we report the safety and feasibility of dual EGFR blockade with EGFR monoclonal antibody and EGFR tyrosine kinase inhibitor combined with anti-VEGF antibody in advanced solid tumors. Methods We conducted a phase I study combining erlotinib, cetuximab, and bevacizumab. Patients with advanced or metastatic solid tumors (excluding colorectal and non-small cell lung cancers) were analyzed for safety, toxicity profile, and response. Anti-tumor activity was evaluated per response evaluation criteria in solid tumors (RECIST 1.0). Results Thirty-six patients received treatment on a range of dose-levels. The most frequent tumor types enrolled were cervical (n = 10), head and neck squamous cell (n = 10), and follicular thyroid (n = 4) cancers. The most common treatment-related grade ≥ 2 adverse events were rash (56%), hypomagnesemia (17%), pruritus (11%), diarrhea (8%), and tumor-related bleeding (8%). Seventeen of 19 patients (89%) treated at the maximum tolerated dose did not present treatment-related dose-limiting toxicity. Fifteen (63%) of the 24 evaluable patients achieved a disease control (stable disease ≥ 4 months (n = 14) and partial response (n = 1). The median number of prior lines of therapies was 3 (range 1–10). Conclusions The triplet combination of erlotinib, cetuximab, and bevacizumab was well tolerated, conferring clinical benefit in heavily pretreated patients. Future studies are warranted with second or third-generation EGFR tyrosine kinase triplet combinations in the EGFR pathway aberrant patients. Trial Registration: ClinicalTrials.gov Identifier: NCT00543504. Sponsor(s): National Cancer Institute (NCI), MD Anderson Cancer Center
Collapse
|
32
|
Imaging and clinical correlates with regorafenib in metastatic colorectal cancer. Cancer Treat Rev 2020; 86:102020. [PMID: 32278232 DOI: 10.1016/j.ctrv.2020.102020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/11/2022]
Abstract
In colorectal cancer (CRC), imaging is important in determining tumor stage, selecting treatment strategies, and in assessing response to therapy. However, some challenges remain with established imaging techniques, such as computed tomography, and with some commonly used response criteria, such as Response Evaluation Criteria in Solid Tumors, which measures change in size of several target lesions instead of change in tumor morphology or metabolic function. In addition, these assessments are not typically conducted until after 8 weeks of treatment, meaning that potential non-responders are often not identified in a timely manner. Regorafenib, an oral tyrosine kinase inhibitor indicated for the treatment of metastatic CRC, blocks the activity of several protein kinases involved in angiogenesis, oncogenesis, metastasis, and tumor immunity. Timely differentiation of regorafenib responders from non-responders using appropriate imaging techniques that recognize not only changes in tumor size but also changes in tumor density or vasculature, may reduce unnecessary drug-related toxicity in patients who are unlikely to respond to treatment. This review discusses the latest developments in computed tomography, magnetic resonance imaging, and positron emission tomography tumor imaging modalities, and how these aid in identifying patients with metastatic CRC who are responders or non-responders to regorafenib treatment.
Collapse
|
33
|
Quantitative magnetic resonance imaging (q-MRI) for the assessment of soft-tissue sarcoma treatment response: a narrative case review of technique development. Clin Imaging 2020; 63:83-93. [PMID: 32163847 DOI: 10.1016/j.clinimag.2020.02.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 02/18/2020] [Accepted: 02/25/2020] [Indexed: 11/20/2022]
Abstract
Soft-tissue sarcomas are a heterogeneous class of tumors that exhibit varying degrees of cellularity and cystic degeneration in response to neoadjuvant chemotherapy. This creates unique challenges in the radiographic assessment of treatment response when relying on conventional markers such as tumor diameter (RECIST criteria). In this case series, we provide a narrative discussion of technique development for whole tumor volume quantitative magnetic resonance imaging (q-MRI), highlighting cases from a small pilot study of 8 patients (9 tumors) pre- and post-neoadjuvant chemotherapy. One of the methods of q-MRI analysis (the "constant-cutoff" technique) was able to predict responders versus non-responders based on percent necrosis and viable tumor volume calculations (p = 0.05), respectively. Our results suggest that q-MRI of whole tumor volume contrast enhancement may have a role in tumor response assessment, although further validation is needed.
Collapse
|
34
|
Vail DM, LeBlanc AK, Jeraj R. Advanced Cancer Imaging Applied in the Comparative Setting. Front Oncol 2020; 10:84. [PMID: 32117739 PMCID: PMC7019008 DOI: 10.3389/fonc.2020.00084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/16/2020] [Indexed: 11/13/2022] Open
Abstract
The potential for companion (pet) species with spontaneously arising tumors to act as surrogates for preclinical development of advanced cancer imaging technologies has become more apparent in the last decade. The utility of the companion model specifically centers around issues related to body size (including spatial target/normal anatomic characteristics), physical size and spatial distribution of metastasis, tumor heterogeneity, the presence of an intact syngeneic immune system and a syngeneic tumor microenvironment shaped by the natural evolution of the cancer. Companion species size allows the use of similar equipment, hardware setup, software, and scan protocols which provide the opportunity for standardization and harmonization of imaging operating procedures and quality assurance across imaging protocols, imaging hardware, and the imaged species. Murine models generally do not replicate the size and spatial distribution of human metastatic cancer and these factors strongly influence image resolution and dosimetry. The following review will discuss several aspects of comparative cancer imaging in more detail while providing several illustrative examples of investigational approaches performed or currently under exploration at our institutions. Topics addressed include a discussion on interested consortia; image quality assurance and harmonization; image-based biomarker development and validation; contrast agent and radionuclide tracer development; advanced imaging to assess and predict response to cytotoxic and immunomodulatory anticancer agents; imaging of the tumor microenvironment; development of novel theranostic approaches; cell trafficking assessment via non-invasive imaging; and intraoperative imaging to inform surgical oncology decision making. Taken in totality, these comparative opportunities predict that safety, diagnostic and efficacy data generated in companion species with naturally developing and progressing cancers would better recapitulate the human cancer condition than that of artificial models in small rodent systems and ultimately accelerate the integration of novel imaging technologies into clinical practice. It is our hope that the examples presented should serve to provide those involved in cancer investigations who are unfamiliar with available comparative methodologies an understanding of the potential utility of this approach.
Collapse
Affiliation(s)
- David M Vail
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Robert Jeraj
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States.,Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
35
|
Travis WD, Dacic S, Wistuba I, Sholl L, Adusumilli P, Bubendorf L, Bunn P, Cascone T, Chaft J, Chen G, Chou TY, Cooper W, Erasmus JJ, Ferreira CG, Goo JM, Heymach J, Hirsch FR, Horinouchi H, Kerr K, Kris M, Jain D, Kim YT, Lopez-Rios F, Lu S, Mitsudomi T, Moreira A, Motoi N, Nicholson AG, Oliveira R, Papotti M, Pastorino U, Paz-Ares L, Pelosi G, Poleri C, Provencio M, Roden AC, Scagliotti G, Swisher SG, Thunnissen E, Tsao MS, Vansteenkiste J, Weder W, Yatabe Y. IASLC Multidisciplinary Recommendations for Pathologic Assessment of Lung Cancer Resection Specimens After Neoadjuvant Therapy. J Thorac Oncol 2020; 15:709-740. [PMID: 32004713 DOI: 10.1016/j.jtho.2020.01.005] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/25/2019] [Accepted: 01/04/2020] [Indexed: 12/14/2022]
Abstract
Currently, there is no established guidance on how to process and evaluate resected lung cancer specimens after neoadjuvant therapy in the setting of clinical trials and clinical practice. There is also a lack of precise definitions on the degree of pathologic response, including major pathologic response or complete pathologic response. For other cancers such as osteosarcoma and colorectal, breast, and esophageal carcinomas, there have been multiple studies investigating pathologic assessment of the effects of neoadjuvant therapy, including some detailed recommendations on how to handle these specimens. A comprehensive mapping approach to gross and histologic processing of osteosarcomas after induction therapy has been used for over 40 years. The purpose of this article is to outline detailed recommendations on how to process lung cancer resection specimens and to define pathologic response, including major pathologic response or complete pathologic response after neoadjuvant therapy. A standardized approach is recommended to assess the percentages of (1) viable tumor, (2) necrosis, and (3) stroma (including inflammation and fibrosis) with a total adding up to 100%. This is recommended for all systemic therapies, including chemotherapy, chemoradiation, molecular-targeted therapy, immunotherapy, or any future novel therapies yet to be discovered, whether administered alone or in combination. Specific issues may differ for certain therapies such as immunotherapy, but the grossing process should be similar, and the histologic evaluation should contain these basic elements. Standard pathologic response assessment should allow for comparisons between different therapies and correlations with disease-free survival and overall survival in ongoing and future trials. The International Association for the Study of Lung Cancer has an effort to collect such data from existing and future clinical trials. These recommendations are intended as guidance for clinical trials, although it is hoped they can be viewed as suggestion for good clinical practice outside of clinical trials, to improve consistency of pathologic assessment of treatment response.
Collapse
Affiliation(s)
- William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Sanja Dacic
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ignacio Wistuba
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Prasad Adusumilli
- Thoracic Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lukas Bubendorf
- Department of Pathology, University of Basel, Basel, Switzerland
| | - Paul Bunn
- Medical Oncology, Colorado University School of Medicine, Aurora, Colorado
| | - Tina Cascone
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Jamie Chaft
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gang Chen
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | | | - Wendy Cooper
- Department of Pathology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Jeremy J Erasmus
- Department of Radiology, MD Anderson Cancer Center, Houston, Texas
| | | | - Jin-Mo Goo
- Department of Radiology, Seoul National University College of Medicine, Seoul, South Korea
| | - John Heymach
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute at Mount Sinai, New York, New York
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Keith Kerr
- Department of Pathology, Aberdeen University Medical School, Aberdeen, Scotland
| | - Mark Kris
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Young T Kim
- Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Fernando Lopez-Rios
- Laboratorio de Dianas Terapeuticas, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai, China
| | - Tetsuya Mitsudomi
- Thoracic Surgery, Kinki University Faculty of Medicine, Osaka-Sayama, Japan
| | - Andre Moreira
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Noriko Motoi
- Department of Pathology, Mational Cancer Center, Tokyo, Japan
| | - Andrew G Nicholson
- Department of Pathology, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | | | - Mauro Papotti
- Department of Pathology, University of Turin, Torino, Italy
| | - Ugo Pastorino
- Thoracic Surgery Division, Istituto Nazionale Tumor, Milan, Italy
| | - Luis Paz-Ares
- Medical Oncology, National Oncology Research Center, Madrid, Spain
| | | | - Claudia Poleri
- Office of Pathology Consultants, Buenos Aries, Argentina
| | - Mariano Provencio
- Oncology Department, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Anja C Roden
- Department of Pathology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ming S Tsao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | | | - Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Yasushi Yatabe
- Department of Pathology, Mational Cancer Center, Tokyo, Japan
| |
Collapse
|
36
|
Abstract
Immunotherapy is one of the most promising treatments for multiple tumor types. The significant clinical benefits and durable responses of immunotherapy have led to the emergence of various immune-related clinical response patterns that extend beyond those achieved with cytotoxic agents. Various studies investigated the efficacy of immunotherapy, including the effect on tumor size, long-term survival benefits, and the ability to overcome the particularly challenging survival curves tailing phenomenon. The current immune-related methods guidelines, such as immune-related Response Criteria (irRC), immune-related Response Evaluation Criteria in Solid Tumors (irRECIST), immune Response Evaluation Criteria in Solid Tumors (iRECIST), and immune-modified Response Evaluation Criteria in Solid Tumors (imRECIST), could be well-adapted to identify the heterogeneity of responses that appear in patients receiving immunotherapy, such as pseudoprogression (PsPD) and hyperprogressive disease (HPD), and to some extent to overcome the limitation of evaluating the efficacy of immunotherapy on tumor size by imaging. Additionally, a second type of evaluation method was proposed based on survival, which includes milestone analysis and restricted mean survival time. Currently, milestone analysis is a complementary tool to summarize and interpret trial results along with more conventional measures of survival and other less established metrics. A golden standard evaluation method to distinguish the efficacy of immunotherapy may improve the process of imaging and aid survival-based efficacy evaluation in patients with solid tumors.
Collapse
Affiliation(s)
- Rilan Bai
- Cancer Center, the First Hospital of Jilin University, Jilin 130021, China
| | - Wenqian Li
- Cancer Center, the First Hospital of Jilin University, Jilin 130021, China
| | - Nawen Du
- Cancer Center, the First Hospital of Jilin University, Jilin 130021, China
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Jilin 130021, China
| |
Collapse
|
37
|
Villanueva F, Yuan C, Drane W, Dang L, Nguyen TC. Cancer Treatment Response to Checkpoint Inhibitors Is Associated with Cytomegalovirus Infection. Cureus 2020; 12:e6670. [PMID: 32104613 PMCID: PMC7026877 DOI: 10.7759/cureus.6670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) and programmed cell death-ligand 1 (PD-L1) checkpoint inhibitors induce tumor response by activating the patient’s own immune system to fight cancer. Tumors with high tumor mutational burden or those that express high levels of PD-1/PD-L1 are more responsive to PD1/PDL1 inhibitors. There is much interest in determining how to improve response to PD-1/PD-L1 inhibitors. We report a case of a patient with metastatic bladder cancer who was primarily resistant to treatment with PD-1/PD-L1 inhibitors, then had a complete response after developing cytomegalovirus infection.
Collapse
Affiliation(s)
| | - Cai Yuan
- Oncology, University of Florida, Gainesville, USA
| | - Walter Drane
- Radiology, University of Florida, Gainesville, USA
| | - Long Dang
- Oncology, Ochsner Health System, Baton Rouge, USA
| | | |
Collapse
|
38
|
Peisen F, Thaiss W, Tietze N, Rausch S, Amend B, Nikolaou K, Bedke J, Stenzl A, Kaufmann S. [Influence of immunomodulators on urological imaging]. Urologe A 2019; 58:1451-1460. [PMID: 31705144 DOI: 10.1007/s00120-019-01063-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have led to great advances in the therapy of metastatic renal cell and urothelial carcinoma. Currently ICI are approved for the first-line therapy of cisplatin-unfit patients (Atezolizumab, Pembrolizumab) and second-line therapy in patients with metastasized urothelial cancer (Atezolizumab, Nivolumab, Pembrolizumab). For the therapy of metastasized RCC, Nivolumab is approved as a second-line therapy and in combination with the CTLA‑4 antibody Ipilimumab as a first-line therapy. OBJECTIVES What does the optimized radiological follow-up and therapy response assessment for ICI, which differ in their pathways from common chemotherapeutics and anti-angiogenetic drugs, look like? What strategies are needed to meet the upcoming challenges concerning interpretation of the acquired images? METHODS A systematic literature search was carried out for urothelial and renal cell carcinoma. RESULTS Immune-related response criteria have been introduced to better characterize the imaging changes occurring under ICI, as monitoring response to immunotherapy still relies on RECIST. CONCLUSIONS To properly identify and predict response after treatment with ICI, additional studies with long-term follow-ups are needed. Because of the growing use of ICI, radiologists and urologist should be familiar with common imaging findings (such as pseudo progress) under immunotherapy to correctly interpret these findings in daily routine.
Collapse
Affiliation(s)
- F Peisen
- Klinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Tübingen, Tübingen, Deutschland
| | - W Thaiss
- Klinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Tübingen, Tübingen, Deutschland
| | - N Tietze
- Klinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Tübingen, Tübingen, Deutschland
| | - S Rausch
- Klinik für Urologie, Universitätsklinikum Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Deutschland
| | - B Amend
- Klinik für Urologie, Universitätsklinikum Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Deutschland
| | - K Nikolaou
- Klinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Tübingen, Tübingen, Deutschland
| | - J Bedke
- Klinik für Urologie, Universitätsklinikum Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Deutschland
| | - A Stenzl
- Klinik für Urologie, Universitätsklinikum Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Deutschland.
| | - S Kaufmann
- Klinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Tübingen, Tübingen, Deutschland
| |
Collapse
|
39
|
Jiao R, Luo H, Xu W, Ge H. Immune checkpoint inhibitors in esophageal squamous cell carcinoma: progress and opportunities. Onco Targets Ther 2019; 12:6023-6032. [PMID: 31551657 PMCID: PMC6677374 DOI: 10.2147/ott.s214579] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/12/2019] [Indexed: 12/24/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the common malignant tumors in the world. More than half of patients with ESCC were detected in advanced or metastatic disease at the time of initial diagnosis and lost the opportunities of surgery. Currently, surgical resection, radiotherapy, and chemotherapy are most utilized in clinical practice, however, they are associated with limited survival benefits. Recognition of the limitation of traditional antitumor strategies prompt the development of new means to treat human cancer. In recent years, studies on immune checkpoint inhibitors (eg PD-1/PD-L1 inhibitors, CTLA-4 inhibitors, etc.) in ESCC have shown promising results. In addition, the combination of immune checkpoint inhibitor and traditional antitumor strategies for ESCC has caused extensive interest, and the results are encouraging. Previous analysis indicated that tumor cell PD-L1 expression, tumor mutation load (TMB), microsatellite instability-high status (MSI-H), and other biomarkers have relatively correlated with the efficacy of immunotherapy. This review explores the recent studies investigating checkpoint inhibitors in ESCC.
Collapse
Affiliation(s)
- Ruidi Jiao
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province450008, People’s Republic of China
| | - Hui Luo
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province450008, People’s Republic of China
| | - Wenbo Xu
- Department of Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan450008, People’s Republic of China
| | - Hong Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province450008, People’s Republic of China
| |
Collapse
|
40
|
Cohen EEW, Bell RB, Bifulco CB, Burtness B, Gillison ML, Harrington KJ, Le QT, Lee NY, Leidner R, Lewis RL, Licitra L, Mehanna H, Mell LK, Raben A, Sikora AG, Uppaluri R, Whitworth F, Zandberg DP, Ferris RL. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of squamous cell carcinoma of the head and neck (HNSCC). J Immunother Cancer 2019; 7:184. [PMID: 31307547 PMCID: PMC6632213 DOI: 10.1186/s40425-019-0662-5] [Citation(s) in RCA: 478] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023] Open
Abstract
Head and neck cancers, including those of the lip and oral cavity, nasal cavity, paranasal sinuses, oropharynx, larynx and nasopharynx represent nearly 700,000 new cases and 380,000 deaths worldwide per annum, and account for over 10,000 annual deaths in the United States alone. Improvement in outcomes are needed for patients with recurrent and or metastatic squamous cell carcinoma of the head and neck (HNSCC). In 2016, the US Food and Drug Administration (FDA) granted the first immunotherapeutic approvals - the anti-PD-1 immune checkpoint inhibitors nivolumab and pembrolizumab - for the treatment of patients with recurrent squamous cell carcinoma of the head and neck (HNSCC) that is refractory to platinum-based regimens. The European Commission followed in 2017 with approval of nivolumab for treatment of the same patient population, and shortly thereafter with approval of pembrolizumab monotherapy for the treatment of recurrent or metastatic HNSCC in adults whose tumors express PD-L1 with a ≥ 50% tumor proportion score and have progressed on or after platinum-containing chemotherapy. Then in 2019, the FDA granted approval for PD-1 inhibition as first-line treatment for patients with metastatic or unresectable, recurrent HNSCC, approving pembrolizumab in combination with platinum and fluorouracil for all patients with HNSCC and pembrolizumab as a single agent for patients with HNSCC whose tumors express a PD-L1 combined positive score ≥ 1. These approvals marked the first new therapies for these patients since 2006, as well as the first immunotherapeutic approvals in this disease. In light of the introduction of these novel therapies for the treatment of patients with head and neck cancer, The Society for Immunotherapy of Cancer (SITC) formed an expert committee tasked with generating consensus recommendations for emerging immunotherapies, including appropriate patient selection, therapy sequence, response monitoring, adverse event management, and biomarker testing. These consensus guidelines serve as a foundation to assist clinicians' understanding of the role of immunotherapies in this disease setting, and to standardize utilization across the field for patient benefit. Due to country-specific variances in approvals, availability and regulations regarding the discussed agents, this panel focused solely on FDA-approved drugs for the treatment of patients in the U.S.
Collapse
Affiliation(s)
- Ezra E W Cohen
- Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - R Bryan Bell
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, OR, USA
| | - Carlo B Bifulco
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, OR, USA
| | - Barbara Burtness
- Yale School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - Maura L Gillison
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Nancy Y Lee
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rom Leidner
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, OR, USA
| | | | - Lisa Licitra
- Fondazione IRCCS Istituto Nazionale dei Tumori Milan and University of Milan, Milan, Italy
| | - Hisham Mehanna
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham, UK
| | - Loren K Mell
- Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - Adam Raben
- Helen F. Graham Cancer Center, Newark, DE, USA
| | | | - Ravindra Uppaluri
- Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | |
Collapse
|
41
|
Allen BC, Florez E, Sirous R, Lirette ST, Griswold M, Remer EM, Wang ZJ, Bieszczad JE, Cox KL, Goenka AH, Howard-Claudio CM, Kang HC, Nandwana SB, Sanyal R, Shinagare AB, Henegan JC, Storrs J, Davenport MS, Ganeshan B, Vasanji A, Rini B, Smith AD. Comparative Effectiveness of Tumor Response Assessment Methods: Standard of Care Versus Computer-Assisted Response Evaluation. JCO Clin Cancer Inform 2019; 1:1-16. [PMID: 30657391 DOI: 10.1200/cci.17.00026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To compare the effectiveness of metastatic tumor response evaluation with computed tomography using computer-assisted versus manual methods. MATERIALS AND METHODS In this institutional review board-approved, Health Insurance Portability and Accountability Act-compliant retrospective study, 11 readers from 10 different institutions independently categorized tumor response according to three different therapeutic response criteria by using paired baseline and initial post-therapy computed tomography studies from 20 randomly selected patients with metastatic renal cell carcinoma who were treated with sunitinib as part of a completed phase III multi-institutional study. Images were evaluated with a manual tumor response evaluation method (standard of care) and with computer-assisted response evaluation (CARE) that included stepwise guidance, interactive error identification and correction methods, automated tumor metric extraction, calculations, response categorization, and data and image archiving. A crossover design, patient randomization, and 2-week washout period were used to reduce recall bias. Comparative effectiveness metrics included error rate and mean patient evaluation time. RESULTS The standard-of-care method, on average, was associated with one or more errors in 30.5% (6.1 of 20) of patients, whereas CARE had a 0.0% (0.0 of 20) error rate ( P < .001). The most common errors were related to data transfer and arithmetic calculation. In patients with errors, the median number of error types was 1 (range, 1 to 3). Mean patient evaluation time with CARE was twice as fast as the standard-of-care method (6.4 minutes v 13.1 minutes; P < .001). CONCLUSION CARE reduced errors and time of evaluation, which indicated better overall effectiveness than manual tumor response evaluation methods that are the current standard of care.
Collapse
Affiliation(s)
- Brian C Allen
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Edward Florez
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Reza Sirous
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Seth T Lirette
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Michael Griswold
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Erick M Remer
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Zhen J Wang
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Jacob E Bieszczad
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Kelly L Cox
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Ajit H Goenka
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Candace M Howard-Claudio
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Hyunseon C Kang
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Sadhna B Nandwana
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Rupan Sanyal
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Atul B Shinagare
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - J Clark Henegan
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Judd Storrs
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Matthew S Davenport
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Balaji Ganeshan
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Amit Vasanji
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Brian Rini
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| | - Andrew D Smith
- Brian C. Allen, Duke University Medical Center, Durham, NC; Edward Florez, Reza Sirous, Seth T. Lirette, Michael Griswold, Candace M. Howard-Claudio, J. Clark Henegan, Judd Storrs, and Andrew D. Smith, University of Mississippi Medical Center, Jackson, MS; Erick M. Remer and Brian Rini, The Cleveland Clinic; Amit Vasanji, ImageIQ, Cleveland; Jacob E. Bieszczad, University of Toledo Medical Center, Toledo, OH; Zhen J. Wang, University of California at San Francisco Medical Center, San Francisco, CA; Kelly L. Cox and Sadhna B. Nandwana, Emory University School of Medicine, Atlanta, GA; Ajit H. Goenka, The Mayo Clinic, Rochester, MN; Hyunseon C. Kang, University of Texas MD Anderson Cancer Center, Houston, TX; Rupan Sanyal, University of Alabama at Birmingham Medical Center, Birmingham, AL; Atul B. Shinagare, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard University, Boston, MA; Matthew S. Davenport, University of Michigan Health System, Ann Arbor, MI; and Balaji Ganeshan, University College of London, London, United Kingdom
| |
Collapse
|
42
|
Guigay J, Sâada-Bouzid E, Peyrade F, Michel C. Approach to the Patient with Recurrent/Metastatic Disease. Curr Treat Options Oncol 2019; 20:65. [PMID: 31240480 DOI: 10.1007/s11864-019-0664-z] [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] [Indexed: 01/20/2023]
Abstract
OPINION STATEMENT For most of patients with a recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC), the treatment remains palliative: The main objective is to reduce the symptoms related to the locoregional relapse, prolong life while maintaining quality of life, which is a big challenge. The systemic treatment needs to be adapted to the performance status, comorbidities, and sequelae of patients. For fit patients, the combination of platinum-based chemotherapy and cetuximab (EXTREME) is the standard of care in first-line treatment since 2008, as no other targeted therapy has been approved in this setting until now. The replacement of 5-FU with a taxane (docetaxel) in the EXTREME regimen has been explored in the large randomized international study TPExtreme which results are awaited in a few months. Depending on the study results on survival, response rate, and tolerance, the TPEx regimen may become a treatment option for patients with R/M HNSCC. Unfit patients are usually treated with platinum-free combinations or with the monotherapies which are recommended in second-line setting (methotrexate, taxanes, cetuximab). However, the irruption of new immunotherapies (e.g., checkpoint inhibitors (CPI)) is changing the guidelines. The tolerance of anti-PD-1 CPI is better than that of chemotherapy, and they seem to be a good option for unfit patients. Anti-PD-1 nivolumab and pembrolizumab are now approved for platinum refractory patients, providing prolonged survival in the case of response, and improvement in quality of life. New options arise in first-line setting with pembrolizumab alone or combined with chemotherapy. Patients with a high PD-L1 biomarker level seem to benefit more from immunotherapy. Other situations (e.g., PD-L1-low, PD-L1-negative, high tumor burden) may more likely to benefit from other combinations, such as cetuximab plus chemotherapy, to avoid local failures and life-threatening fast progression. In terms of perspectives, chemo-free and CPI-free approaches, using other immune oncology agents, should be the next steps.
Collapse
Affiliation(s)
- Joël Guigay
- Centre Antoine Lacassagne, Cancer research center, Medical Oncology Department, FHU Oncoage, University Côte d'Azur, 33 av. de Valombrose, 06189, Nice Cedex 2, France.
| | - Esma Sâada-Bouzid
- Centre Antoine Lacassagne, Cancer research center, Medical Oncology Department, FHU Oncoage, University Côte d'Azur, 33 av. de Valombrose, 06189, Nice Cedex 2, France
| | - Frédéric Peyrade
- Centre Antoine Lacassagne, Cancer research center, Medical Oncology Department, FHU Oncoage, University Côte d'Azur, 33 av. de Valombrose, 06189, Nice Cedex 2, France
| | - Cécile Michel
- Centre Antoine Lacassagne, Cancer research center, Medical Oncology Department, FHU Oncoage, University Côte d'Azur, 33 av. de Valombrose, 06189, Nice Cedex 2, France
| |
Collapse
|
43
|
Bak SH, Park H, Sohn I, Lee SH, Ahn MJ, Lee HY. Prognostic Impact of Longitudinal Monitoring of Radiomic Features in Patients with Advanced Non-Small Cell Lung Cancer. Sci Rep 2019; 9:8730. [PMID: 31217441 PMCID: PMC6584670 DOI: 10.1038/s41598-019-45117-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 05/31/2019] [Indexed: 01/10/2023] Open
Abstract
Tumor growth dynamics vary substantially in non-small cell lung cancer (NSCLC). We aimed to develop biomarkers reflecting longitudinal change of radiomic features in NSCLC and evaluate their prognostic power. Fifty-three patients with advanced NSCLC were included. Three primary variables reflecting patterns of longitudinal change were extracted: area under the curve of longitudinal change (AUC1), beta value reflecting slope over time, and AUC2, a value obtained by considering the slope and area over the longitudinal change of features. We constructed models for predicting survival with multivariate cox regression, and identified the performance of these models. AUC2 exhibited an excellent correlation between patterns of longitudinal volume change and a significant difference in overall survival time. Multivariate regression analysis based on cut-off values of radiomic features extracted from baseline CT and AUC2 showed that kurtosis of positive pixel values and surface area from baseline CT, AUC2 of density, skewness of positive pixel values, and entropy at inner portion were associated with overall survival. For the prediction model, the areas under the receiver operating characteristic curve (AUROC) were 0.948 and 0.862 at 1 and 3 years of follow-up, respectively. Longitudinal change of radiomic tumor features may serve as prognostic biomarkers in patients with advanced NSCLC.
Collapse
Affiliation(s)
- So Hyeon Bak
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Radiology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Hyunjin Park
- School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science, Suwon, Korea
| | - Insuk Sohn
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Seung Hak Lee
- Department of Electronic Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
| | - Myung-Ju Ahn
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.
| |
Collapse
|
44
|
Trinidad López C, De La Fuente Aguado J, Oca Pernas R, Delgado Sánchez-Gracián C, Santos Armentia E, Vaamonde Liste A, Prada González R, Souto Bayarri M. Evaluation of response to conventional chemotherapy and radiotherapy by perfusion computed tomography in non-small cell lung cancer (NSCLC). Eur Radiol Exp 2019; 3:23. [PMID: 31197486 PMCID: PMC6565789 DOI: 10.1186/s41747-019-0101-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/02/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND To evaluate changes in perfusion computed tomography (PCT) parameters induced by treatment with conventional chemotherapy (CCT) alone or with CCT and radiation therapy (RT) in patients with non-small cell lung cancer (NSCLC) and to determine whether these changes correlate with response as defined by the response evaluation criteria in solid tumours version 1.1 (RECIST-1.1). METHODS Fifty-three patients with a histological diagnosis of NSCLC prospectively underwent PCT of the whole tumour, before/after CCT or before/after CCT and RT. Blood flow (BF), blood volume (BV), permeability (PMB), and mean transit time (MTT) were compared before and after treatment and with the response as defined by RECIST-1.1. The relationship between changes in the perfusion parameters and in tumour size was also evaluated. RESULTS PCT parameters decreased after treatment, significantly for BV (p = 0.002) and MTT (p = 0.027). The 30 patients with partial response had a significant decrease of 21% for BV (p = 0.006) and 17% for MTT (p = 0.031). A non-significant decrease in all perfusion parameters was found in patients with stable disease (p > 0.137). In patients with progressive disease, MTT decreased by 10% (p = 0.465) and the other parameters did not significantly vary (p > 0.809). No significant correlation was found between changes in size and PCT parameters (p > 0.145). CONCLUSIONS Treatment of NSCLC with platinum derivatives, with or without RT, induces changes in PCT parameters. Partial response is associated with a significant decrease in BV and MTT, attributable to the effect of the treatment on tumour vascularisation.
Collapse
Affiliation(s)
- Carmen Trinidad López
- Department of Radiology, POVISA Hospital, 5 Salamanca st, 36208, Vigo, Pontevedra, Spain.
| | | | - Roque Oca Pernas
- Department of Radiology, Osatek, Urduliz Hospital, Vizcaya, Spain
| | | | - Eloisa Santos Armentia
- Department of Radiology, POVISA Hospital, 5 Salamanca st, 36208, Vigo, Pontevedra, Spain
| | - Antonio Vaamonde Liste
- Department of Statistics and Operational Research, Faculty of Economic and Business Sciences, Vigo University Spain, Vigo, Spain
| | - Raquel Prada González
- Department of Radiology, POVISA Hospital, 5 Salamanca st, 36208, Vigo, Pontevedra, Spain
| | - Miguel Souto Bayarri
- Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
45
|
Coy HJ, Douek ML, Ruchalski K, Kim HJ, Gutierrez A, Patel M, Sai V, Margolis DJA, Kaplan A, Brown M, Goldin J, Raman SS. Components of Radiologic Progressive Disease Defined by RECIST 1.1 in Patients with Metastatic Clear Cell Renal Cell Carcinoma. Radiology 2019; 292:103-109. [PMID: 31084479 DOI: 10.1148/radiol.2019182922] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Progression-free survival (PFS) determined by Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1) is the reference standard to assess efficacy of treatments in patients with clear cell renal cell carcinoma. Purpose To assess the most common components of radiologic progressive disease as defined by RECIST 1.1 in patients with clear cell renal cell carcinoma and how the progression events impact PFS. Materials and Methods This secondary analysis of the phase III METEOR trial conducted between 2013 and 2014 included patients with metastatic clear cell renal cell carcinoma, with at least one target lesion at baseline and one follow-up time point, who were determined according to RECIST 1.1 to have progressive disease. A chest, abdominal, and pelvic scan were acquired at each time point. Kruskal-Wallis analysis was used to test differences in median PFS among the RECIST 1.1 progression events. The Holm-Bonferroni method was used to compare the median PFS of the progression events for the family-wise error rate of 5% to adjust P values for multiple comparisons. Results Of the 395 patients (296 men, 98 women, and one patient with sex not reported; mean age, 61 years ± 10), 73 (18.5%) had progression due to non-target disease, 105 (26.6%) had new lesions, and 126 (31.9%) had progression of target lesions (defined by an increase in the sum of diameters). Patients with progression of non-target disease and those with new lesions had shorter PFS than patients with progression defined by the target lesions (median PFS, 2.8 months [95% confidence interval {CI}: 1.9 months, 3.7 months] and 3.6 months [95% CI: 3.3 months, 3.7 months] vs 5.4 months [95% CI: 5.0 months, 5.5 months], respectively [P < .01]). Conclusion The most common causes for radiologic progression of renal cell carcinoma were based on non-target disease and new lesions rather than change in target lesions, despite this being considered uncommon in the Response Evaluation Criteria in Solid Tumors version 1.1 literature. © RSNA, 2019 See also the editorial by Kuhl in this issue.
Collapse
Affiliation(s)
- Heidi J Coy
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Michael L Douek
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Kathleen Ruchalski
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Hyun J Kim
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Antonio Gutierrez
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Maitrya Patel
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Victor Sai
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Daniel J A Margolis
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Andrew Kaplan
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Matthew Brown
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Jonathan Goldin
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| | - Steven S Raman
- From the Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, 924 Westwood Blvd, Suite 615, Los Angeles, CA 90049 (H.J.C., M.L.D., K.R., H.J.K., A.G., M.P., V.S., A.K., M.B., J.G., S.S.R.); Department of Biostatistics, Fielding School of Public Health at UCLA, Los Angeles, CA (H.J.K.); Department of Radiology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, NY (D.M.); Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.); Department of Surgery, David Geffen School of Medicine at UCLA, Ronald Reagan UCLA Medical Center, Los Angeles, CA (S.S.R.)
| |
Collapse
|
46
|
Provencio M, Carcereny E, Artal Á. Consensus on the use of immune-related response criteria to evaluate the efficacy of immunotherapy in non-small cell lung cancer. Clin Transl Oncol 2019; 21:1464-1471. [PMID: 30903517 DOI: 10.1007/s12094-019-02072-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 02/23/2019] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Many methods used to assess the effectiveness of immune checkpoint (programmed death-ligand 1 or cytotoxic T-lymphocyte-associated protein 4) inhibitors for non-small cell lung cancer (NSCLC) are insufficient, as the therapeutic benefit of these agents is often underestimated. Consequently, immune-related evaluation criteria have been developed to better reflect their efficacy. The aim of this consensus was to obtain the opinion of lung cancer experts on the adequacy of immune-response criteria for evaluating the efficacy of these treatments. METHODS Through two rounds of a modified Delphi consensus, 18 Spanish lung cancer experts participated in a 15-item questionnaire regarding the use of immunotherapies for NSCLC and the assessment criteria used to evaluate their effectiveness. RESULTS Consensus was achieved on 80% of the items in the questionnaire. The panelists agreed that although the Response Evaluation Criteria in Solid Tumors (RECIST) are standard for the evaluation of solid tumors, immune-related response criteria would be useful for measuring the efficacy of immunotherapy. In addition, they considered that an overall survival (OS) rate at 2-5 years is the most useful end point for assessing the benefit of immunotherapy, as clinical benefit may extend beyond the RECIST criteria-defined progression of disease. CONCLUSIONS Although immune-related response criteria have been developed to better evaluate the efficacy of immunotherapy, their use has not been validated and is restricted to investigational applications. However, they may prove to be a useful tool for measuring the efficacy of immunotherapy agents in NSCLC, especially the OS rate at 2-5 years.
Collapse
Affiliation(s)
- M Provencio
- Medical Oncology Department, Hospital Universitario Puerta de Hierro Majadahonda, Calle Manuel de Falla 1, 28222, Majadahonda, Madrid, Spain.
| | - E Carcereny
- Medical Oncology Department, Hospital Instituto Catalán de Oncología Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Á Artal
- Medical Oncology Department, Hospital Universitario Miguel Servet, Zaragoza, Spain
| |
Collapse
|
47
|
Peng X, Wang B, Yang Y, Zhang Y, Liu Y, He Y, Zhang C, Fan H. Liver Tumor Spheroid Reconstitution for Testing Mitochondrial Targeted Magnetic Hyperthermia Treatment. ACS Biomater Sci Eng 2019; 5:1635-1644. [DOI: 10.1021/acsbiomaterials.8b01630] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Xuqi Peng
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xuefu Street No. 1, Xi’an, 710127, China
- School of Chemical Engineering, Northwest University, Xuefu Street No. 1, Xi’an, 710069, China
| | - Bingquan Wang
- State Key Laboratory of Cultivation Base for Photoelectric Technology and Functional Materials, Laboratory of Optoelectronic Technology of Shaanxi Province, National Center for International Research of Photoelectric Technology & Nanofunctional Materials and Application, Institute of Photonics and Photon-Technology, Northwest University, Xuefu Street No. 1, Xi’an 710127, China
| | - Yu Yang
- College of Life Science, Northwest University, Xuefu Street No. 1, Xi’an, 710069, China
| | - Yihan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xuefu Street No. 1, Xi’an, 710127, China
| | - Yonggang Liu
- Laboratory of Stem Cells and Tissue Engineering, College of Basic Medicine, Chongqing Medical University, Medical School Road NO. 1, Chongqing 400016, China
| | - Yuan He
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xuefu Street No. 1, Xi’an, 710127, China
| | - Ce Zhang
- State Key Laboratory of Cultivation Base for Photoelectric Technology and Functional Materials, Laboratory of Optoelectronic Technology of Shaanxi Province, National Center for International Research of Photoelectric Technology & Nanofunctional Materials and Application, Institute of Photonics and Photon-Technology, Northwest University, Xuefu Street No. 1, Xi’an 710127, China
| | - Haiming Fan
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xuefu Street No. 1, Xi’an, 710127, China
- School of Chemical Engineering, Northwest University, Xuefu Street No. 1, Xi’an, 710069, China
| |
Collapse
|
48
|
Nishino M, Hatabu H, Hodi FS. Imaging of Cancer Immunotherapy: Current Approaches and Future Directions. Radiology 2019; 290:9-22. [PMID: 30457485 PMCID: PMC6312436 DOI: 10.1148/radiol.2018181349] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/20/2022]
Abstract
Cancer immunotherapy using immune-checkpoint inhibitors has emerged as an effective treatment option for a variety of advanced cancers in the past decade. Because of the distinct mechanisms of immunotherapy that activate the host immunity to treat cancers, unconventional immune-related phenomena are encountered in terms of tumor response and progression, as well as drug toxicity. Imaging plays an important role in objectively characterizing immune-related tumor responses and progression and in detecting and monitoring immune-related adverse events. Moreover, emerging data suggest a promise for molecular imaging that can visualize the specific target molecules involved in immune-checkpoint pathways. In this article, the background and current status of cancer immunotherapy are summarized, and the current methods for imaging evaluations of immune-related responses and toxicities are reviewed along with their limitations and pitfalls. Emerging approaches with molecular imaging are also discussed as a future direction to address unmet needs.
Collapse
Affiliation(s)
- Mizuki Nishino
- From the Departments of Radiology (M.N., H.H.), Medical Oncology (F.S.H.), and Medicine (F.S.H.), Brigham and Women’s Hospital and Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215
| | - Hiroto Hatabu
- From the Departments of Radiology (M.N., H.H.), Medical Oncology (F.S.H.), and Medicine (F.S.H.), Brigham and Women’s Hospital and Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215
| | - F. Stephen Hodi
- From the Departments of Radiology (M.N., H.H.), Medical Oncology (F.S.H.), and Medicine (F.S.H.), Brigham and Women’s Hospital and Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215
| |
Collapse
|
49
|
Gupta M, Choudhury PS, Rawal S, Goel HC, Rao SA. Evaluation of response in patients of metastatic castration resistant prostate cancer undergoing systemic radiotherapy with lutetium177-prostate-specific membrane antigen: A comparison between response evaluation criteria in solid tumors, positron-emission tomography response criteria in solid tumors, European organization for research and treatment of cancer, and MDA criteria assessed by gallium 68-prostate-specific membrane antigen positron-emission tomography-computed tomography. Urol Ann 2019; 11:155-162. [PMID: 31040600 PMCID: PMC6476211 DOI: 10.4103/ua.ua_111_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Introduction We evaluated various morphological and molecular response criteria in metastatic castration-resistant prostate cancer (PCa) patient undergoing peptide receptor radioligand therapy (PRLT) with Lutetium177-prostate-specific membrane antigen (PSMA) by using Gallium 68-PSMA positron-emission tomography-computed tomography (Ga68-PSMA PET-CT). Methods A total of 46 pre- and 8-12 weeks' post-PRLT Ga68-PSMA PET-CT studies were reanalyzed (23 comparisons). Prostate-specific antigen drop of ≥50% and ≥25% increase was considered as partial response (PR) and progressive disease (PD), respectively, for biochemical response (BR) while change in-between was considered as stable disease (SD). Response evaluation criteria in solid tumors 1.1 (RECIST 1.1) and MD Anderson (MDA) criteria for morphological response while PET response criteria in solid tumors 1.0 (PERCIST 1.0) and European organization for research and treatment of cancer (EORTC) criteria for molecular response were used. Kappa coefficient was derived to see the level of agreement. Results The proportion of PD, PR, and SD by BR and RECIST criteria was 9 (39.13%), 3 (13.04%), and 11 (47.83%) and 5 (21.74%), 2 (8.70%), and 16 (69.57%), respectively. The proportion of PD, PR, and SD was same by PERCIST and EORTC criteria and which were 8 (34.78%), 5 (21.74%), and 10 (43.48%). The proportion of PD, PR, and SD by MDA criteria was 1 (4.35%), 1 (4.35%), and 21 (91.30%), respectively. Poor agreement between BR and both morphological criteria while a statistically significant agreement with both molecular criteria seen. Conclusion We concluded that molecular criteria performed better than morphological criteria in response assessment by Ga68-PSMA PET-CT in metastatic castration resistant PCa patients undergoing PRLT.
Collapse
Affiliation(s)
- Manoj Gupta
- Department of Nuclear Medicine, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | | | - Sudhir Rawal
- Department of Uro-Gynae Surgical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Harish Chandra Goel
- Amity Centre for Radiation Biology, Amity University, Noida, Uttar Pradesh, India
| | - Shriram Avinash Rao
- Department of Radiology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| |
Collapse
|
50
|
Grünwald V, Hornig M. Systemic and Sequential Therapy in Advanced Renal Cell Carcinoma. Urol Oncol 2019. [DOI: 10.1007/978-3-319-42623-5_64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|