1
|
Taya M, Hou X, Veneris JT, Kazi N, Larson MC, Maurer MJ, Heinzen EP, Chen H, Lastra R, Oberg AL, Weroha SJ, Fleming GF, Conzen SD. Investigation of selective glucocorticoid receptor modulation in high-grade serous ovarian cancer PDX models. J Gynecol Oncol 2024; 36:36.e4. [PMID: 38909640 DOI: 10.3802/jgo.2025.36.e4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/18/2024] [Accepted: 05/07/2024] [Indexed: 06/25/2024] Open
Abstract
OBJECTIVE In ovarian cancer (OvCa), tumor cell high glucocorticoid receptor (GR) has been associated with poor patient prognosis. In vitro, GR activation inhibits chemotherapy-induced OvCa cell death in association with transcriptional upregulation of genes encoding anti-apoptotic proteins. A recent randomized phase II study demonstrated improvement in progression-free survival (PFS) for heavily pre-treated OvCa patients randomized to receive therapy with a selective GR modulator (SGRM) plus chemotherapy compared to chemotherapy alone. We hypothesized that SGRM therapy would improve carboplatin response in OvCa patient-derived xenograft (PDX). METHODS Six high-grade serous (HGS) OvCa PDX models expressing GR mRNA (NR3C1) and protein were treated with chemotherapy +/- SGRM. Tumor size was measured longitudinally by peritoneal transcutaneous ultrasonography. RESULTS One of the 6 GR-positive PDX models showed a significant improvement in PFS with the addition of a SGRM. Interestingly, the single model with an improved PFS was least carboplatin sensitive. Possible explanations for the modest SGRM activity include the high carboplatin sensitivity of 5 of the PDX tumors and the potential that SGRMs activate the tumor invasive immune cells in patients (absent from immunocompromised mice). The level of tumor GR protein expression alone appears insufficient for predicting SGRM response. CONCLUSION The significant improvement in PFS shown in 1 of the 6 models after treatment with a SGRM plus chemotherapy underscores the need to determine predictive biomarkers for SGRM therapy in HGS OvCa and to better identify patient subgroups that are most likely to benefit from adding GR modulation to chemotherapy.
Collapse
Affiliation(s)
- Manisha Taya
- Division of Hematology and Oncology, UT Southwestern, Dallas, TX, USA
| | - Xiaonan Hou
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jennifer T Veneris
- Department of Medicine, Section of Hematology and Oncology, The University of Chicago, Chicago, IL, USA
| | - Nina Kazi
- Division of Hematology and Oncology, UT Southwestern, Dallas, TX, USA
| | - Melissa C Larson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Matthew J Maurer
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ethan P Heinzen
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | - Hao Chen
- Department of Pathology, UT Southwestern, Dallas, TX, USA
| | - Ricardo Lastra
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Ann L Oberg
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - S John Weroha
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Gini F Fleming
- Department of Medicine, Section of Hematology and Oncology, The University of Chicago, Chicago, IL, USA
| | - Suzanne D Conzen
- Division of Hematology and Oncology, UT Southwestern, Dallas, TX, USA.
| |
Collapse
|
2
|
Ambegoda P, Wei HC, Jang SRJ. The role of immune cells in resistance to oncolytic viral therapy. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:5900-5946. [PMID: 38872564 DOI: 10.3934/mbe.2024261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Resistance to treatment poses a major challenge for cancer therapy, and oncoviral treatment encounters the issue of viral resistance as well. In this investigation, we introduce deterministic differential equation models to explore the effect of resistance on oncolytic viral therapy. Specifically, we classify tumor cells into resistant, sensitive, or infected with respect to oncolytic viruses for our analysis. Immune cells can eliminate both tumor cells and viruses. Our research shows that the introduction of immune cells into the tumor-virus interaction prevents all tumor cells from becoming resistant in the absence of conversion from resistance to sensitivity, given that the proliferation rate of immune cells exceeds their death rate. The inclusion of immune cells leads to an additional virus-free equilibrium when the immune cell recruitment rate is sufficiently high. The total tumor burden at this virus-free equilibrium is smaller than that at the virus-free and immune-free equilibrium. Therefore, immune cells are capable of reducing the tumor load under the condition of sufficient immune strength. Numerical investigations reveal that the virus transmission rate and parameters related to the immune response significantly impact treatment outcomes. However, monotherapy alone is insufficient for eradicating tumor cells, necessitating the implementation of additional therapies. Further numerical simulation shows that combination therapy with chimeric antigen receptor (CAR T-cell) therapy can enhance the success of treatment.
Collapse
Affiliation(s)
- Prathibha Ambegoda
- Department of Mathematics & Statistics, Texas Tech University, Lubbock, TX, USA
| | - Hsiu-Chuan Wei
- Department of Applied Mathematics, Feng Chia University, Taichung, Taiwan
| | - Sophia R-J Jang
- Department of Mathematics & Statistics, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
3
|
Melton CA, Freese P, Zhou Y, Shenoy A, Bagaria S, Chang C, Kuo CC, Scott E, Srinivasan S, Cann G, Roychowdhury-Saha M, Chang PY, Singh AH. A Novel Tissue-Free Method to Estimate Tumor-Derived Cell-Free DNA Quantity Using Tumor Methylation Patterns. Cancers (Basel) 2023; 16:82. [PMID: 38201510 PMCID: PMC10777919 DOI: 10.3390/cancers16010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/07/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Estimating the abundance of cell-free DNA (cfDNA) fragments shed from a tumor (i.e., circulating tumor DNA (ctDNA)) can approximate tumor burden, which has numerous clinical applications. We derived a novel, broadly applicable statistical method to quantify cancer-indicative methylation patterns within cfDNA to estimate ctDNA abundance, even at low levels. Our algorithm identified differentially methylated regions (DMRs) between a reference database of cancer tissue biopsy samples and cfDNA from individuals without cancer. Then, without utilizing matched tissue biopsy, counts of fragments matching the cancer-indicative hyper/hypo-methylated patterns within DMRs were used to determine a tumor methylated fraction (TMeF; a methylation-based quantification of the circulating tumor allele fraction and estimate of ctDNA abundance) for plasma samples. TMeF and small variant allele fraction (SVAF) estimates of the same cancer plasma samples were correlated (Spearman's correlation coefficient: 0.73), and synthetic dilutions to expected TMeF of 10-3 and 10-4 had estimated TMeF within two-fold for 95% and 77% of samples, respectively. TMeF increased with cancer stage and tumor size and inversely correlated with survival probability. Therefore, tumor-derived fragments in the cfDNA of patients with cancer can be leveraged to estimate ctDNA abundance without the need for a tumor biopsy, which may provide non-invasive clinical approximations of tumor burden.
Collapse
|
4
|
Cappello G, Romano V, Neri E, Fournier L, D'Anastasi M, Laghi A, Zamboni GA, Beets-Tan RGH, Schlemmer HP, Regge D. A European Society of Oncologic Imaging (ESOI) survey on the radiological assessment of response to oncologic treatments in clinical practice. Insights Imaging 2023; 14:220. [PMID: 38117394 PMCID: PMC10733253 DOI: 10.1186/s13244-023-01568-6] [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: 07/16/2023] [Accepted: 11/08/2023] [Indexed: 12/21/2023] Open
Abstract
OBJECTIVES To present the results of a survey on the assessment of treatment response with imaging in oncologic patient, in routine clinical practice. The survey was promoted by the European Society of Oncologic Imaging to gather information for the development of reporting models and recommendations. METHODS The survey was launched on the European Society of Oncologic Imaging website and was available for 3 weeks. It consisted of 5 sections, including 24 questions related to the following topics: demographic and professional information, methods for lesion measurement, how to deal with diminutive lesions, how to report baseline and follow-up examinations, which previous studies should be used for comparison, and role of RECIST 1.1 criteria in the daily clinical practice. RESULTS A total of 286 responses were received. Most responders followed the RECIST 1.1 recommendations for the measurement of target lesions and lymph nodes and for the assessment of tumor response. To assess response, 48.6% used previous and/or best response study in addition to baseline, 25.2% included the evaluation of all main time points, and 35% used as the reference only the previous study. A considerable number of responders used RECIST 1.1 criteria in daily clinical practice (41.6%) or thought that they should be always applied (60.8%). CONCLUSION Since standardized criteria are mainly a prerogative of clinical trials, in daily routine, reporting strategies are left to radiologists and oncologists, which may issue local and diversified recommendations. The survey emphasizes the need for more generally applicable rules for response assessment in clinical practice. CRITICAL RELEVANCE STATEMENT Compared to clinical trials which use specific criteria to evaluate response to oncological treatments, the free narrative report usually adopted in daily clinical practice may lack clarity and useful information, and therefore, more structured approaches are needed. KEY POINTS · Most radiologists consider standardized reporting strategies essential for an objective assessment of tumor response in clinical practice. · Radiologists increasingly rely on RECIST 1.1 in their daily clinical practice. · Treatment response evaluation should require a complete analysis of all imaging time points and not only of the last.
Collapse
Affiliation(s)
- Giovanni Cappello
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, Str. Prov.le 142 km 3.95, 10060, Candiolo (Turin), Italy.
| | - Vittorio Romano
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, Str. Prov.le 142 km 3.95, 10060, Candiolo (Turin), Italy
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Emanuele Neri
- Department of Translational Research, Academic Radiology, University of Pisa, 56124, Pisa, Italy
| | - Laure Fournier
- Radiology Department, Hôpital Européen Georges Pompidou, AP-HP, Université de Paris, 20 Rue Leblanc, 75015, Paris, France
| | - Melvin D'Anastasi
- Medical Imaging Department, Mater Dei Hospital, University of Malta, Msida, 2090, MSD, Malta
| | - Andrea Laghi
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Sant'Andrea University Hospital, Via Di Grottarossa, 1035-1039, 00189, Rome, Italy
| | - Giulia A Zamboni
- Department of Diagnostics and Public Health, Institute of Radiology, University of Verona, Policlinico GB Rossi, P.Le LA Scuro 10, 37134, Verona, Italy
| | - Regina G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, P.O. Box 90203, 1006 BE, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| | - Heinz-Peter Schlemmer
- Department of Radiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Daniele Regge
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, Str. Prov.le 142 km 3.95, 10060, Candiolo (Turin), Italy
- Academic Radiology, Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Via Roma 67, Pisa, 56126, Italy
| |
Collapse
|
5
|
Suter Y, Notter M, Meier R, Loosli T, Schucht P, Wiest R, Reyes M, Knecht U. Evaluating automated longitudinal tumor measurements for glioblastoma response assessment. FRONTIERS IN RADIOLOGY 2023; 3:1211859. [PMID: 37745204 PMCID: PMC10513769 DOI: 10.3389/fradi.2023.1211859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/25/2023] [Indexed: 09/26/2023]
Abstract
Automated tumor segmentation tools for glioblastoma show promising performance. To apply these tools for automated response assessment, longitudinal segmentation, and tumor measurement, consistency is critical. This study aimed to determine whether BraTumIA and HD-GLIO are suited for this task. We evaluated two segmentation tools with respect to automated response assessment on the single-center retrospective LUMIERE dataset with 80 patients and a total of 502 post-operative time points. Volumetry and automated bi-dimensional measurements were compared with expert measurements following the Response Assessment in Neuro-Oncology (RANO) guidelines. The longitudinal trend agreement between the expert and methods was evaluated, and the RANO progression thresholds were tested against the expert-derived time-to-progression (TTP). The TTP and overall survival (OS) correlation was used to check the progression thresholds. We evaluated the automated detection and influence of non-measurable lesions. The tumor volume trend agreement calculated between segmentation volumes and the expert bi-dimensional measurements was high (HD-GLIO: 81.1%, BraTumIA: 79.7%). BraTumIA achieved the closest match to the expert TTP using the recommended RANO progression threshold. HD-GLIO-derived tumor volumes reached the highest correlation between TTP and OS (0.55). Both tools failed at an accurate lesion count across time. Manual false-positive removal and restricting to a maximum number of measurable lesions had no beneficial effect. Expert supervision and manual corrections are still necessary when applying the tested automated segmentation tools for automated response assessment. The longitudinal consistency of current segmentation tools needs further improvement. Validation of volumetric and bi-dimensional progression thresholds with multi-center studies is required to move toward volumetry-based response assessment.
Collapse
Affiliation(s)
- Yannick Suter
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Michelle Notter
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
- Cantonal Hospital of Graubünden, Chur, Switzerland
| | - Raphael Meier
- Support Center for Advanced Neuroimaging, Inselspital, Bern, Switzerland
| | - Tina Loosli
- Support Center for Advanced Neuroimaging, Inselspital, Bern, Switzerland
| | | | - Roland Wiest
- Support Center for Advanced Neuroimaging, Inselspital, Bern, Switzerland
| | - Mauricio Reyes
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Urspeter Knecht
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
- Radiology Department, Spital Emmental, Burgdorf, Switzerland
| |
Collapse
|
6
|
Gong AJ, Ruchalski K, Kim HJ, Douek M, Gutierrez A, Patel M, Sai V, Coy H, Villegas B, Raman S, Goldin J. RECIST 1.1 Target Lesion Categorical Response in Metastatic Renal Cell Carcinoma: A Comparison of Conventional versus Volumetric Assessment. Radiol Imaging Cancer 2023; 5:e220166. [PMID: 37656041 PMCID: PMC10546365 DOI: 10.1148/rycan.220166] [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: 11/21/2022] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 09/02/2023]
Abstract
Purpose To investigate Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1) approximations of target lesion tumor burden by comparing categorical treatment response according to conventional RECIST versus actual tumor volume measurements of RECIST target lesions. Materials and Methods This is a retrospective cohort study of individuals with metastatic renal cell carcinoma enrolled in a clinical trial (from 2003 to 2017) and includes individuals who underwent baseline and at least one follow-up chest, abdominal, and pelvic CT study and with at least one target lesion. Target lesion volume was assessed by (a) Vmodel, a spherical model of conventional RECIST 1.1, which was extrapolated from RECIST diameter, and (b) Vactual, manually contoured volume. Volumetric responses were determined by the sum of target lesion volumes (Vmodel-sum TL and Vactual-sum TL, respectively). Categorical volumetric thresholds were extrapolated from RECIST. McNemar tests were used to compare categorical volume responses. Results Target lesions were assessed at baseline (638 participants), week 9 (593 participants), and week 17 (508 participants). Vmodel-sum TL classified more participants as having progressive disease (PD), compared with Vactual-sum TL at week 9 (52 vs 31 participants) and week 17 (57 vs 39 participants), with significant overall response discordance (P < .001). At week 9, 25 (48%) of 52 participants labeled with PD by Vmodel-sum TL were classified as having stable disease by Vactual-sum TL. Conclusion A model of RECIST 1.1 based on a single diameter measurement more frequently classified PD compared with response assessment by actual measured tumor volume. Keywords: Urinary, Kidney, Metastases, Oncology, Tumor Response, Volume Analysis, Outcomes Analysis ClinicalTrials.gov registration no. NCT01865747 © RSNA, 2023 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Amanda J. Gong
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Kathleen Ruchalski
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Hyun J. Kim
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Michael Douek
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Antonio Gutierrez
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Maitraya Patel
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Victor Sai
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Heidi Coy
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Bianca Villegas
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Steven Raman
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| | - Jonathan Goldin
- From the David Geffen School of Medicine, University of California,
Los Angeles, Calif (A.J.G., K.R., H.J.K., M.D., A.G., M.P., V.S., H.C., S.R.,
J.G.); Department of Radiological Sciences, UCLA, Los Angeles, Calif (K.R.,
H.J.K., M.D., A.G., M.P., V.S., S.R., J.G.); and UCLA Center for Computer Vision
and Imaging Biomarkers, 924 Westwood Blvd, Ste 615, Los Angeles, CA 90024
(A.J.G., H.J.K., H.C., B.V., J.G.)
| |
Collapse
|
7
|
Qiao C, Hu S, Wang D, Cao K, Wang Z, Wang X, Ma X, Li Z, Hou W. Effectiveness and safety of Shenqi Fuzheng injection combined with platinum-based chemotherapy for treatment of advanced non-small cell lung cancer: a systematic review and meta-analysis. Front Oncol 2023; 13:1198768. [PMID: 37731634 PMCID: PMC10507621 DOI: 10.3389/fonc.2023.1198768] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 08/08/2023] [Indexed: 09/22/2023] Open
Abstract
Objective To evaluate the efficacy and safety of Shenqi Fuzheng Injection (SFI) combined with platinum-based chemotherapy (PBC) for the treatment of advanced non-small cell lung cancer (NSCLC). Methods Seven electronic databases, including CNKI and Wanfang, were comprehensively searched to screen randomized controlled trials (RCTs) until May 1, 2022. The quality of each trial was evaluated according to the Cochrane Handbook for Systematic Reviews of Interventions, and systematic reviews were conducted according to the PRISMA guidelines. Statistical analysis was performed using Review Manager 5.3, and the results were expressed as relative risk (RR) and 95% confidence interval (95% CI). The primary outcome measures were objective response rate (ORR) and disease control rate (DCR). The secondary outcome measures were quality of life and toxicity. Subgroup analysis was performed according to the number of days of SFI single-cycle treatment and combined PBC regimen. Results A total of 44 RCTs involving 3475 patients were included in the study. The meta-analysis results showed that, compared with PBC alone, SFI combined with PBC significantly improved the ORR (RR = 1.27, 95% CI = 1.18-1.37, P < 0.00001), DCR (RR = 1.12, 95% CI = 1.08-1.15, P < 0.00001), and quality of life (RR = 1.41, 95% CI = 1.31-1.52, P < 0.00001). It also reduced chemotherapy-induced hemoglobin reduction (RR = 0.57, 95% CI = 0.48-0.67, P < 0.00001), leukopenia (RR = 0.61, 95% CI = 0.53-0.71, P < 0.00001), thrombocytopenia (RR = 0.62, 95% CI = 0.55-0.70, P < 0.00001), and simple bone marrow suppression (RR = 0.55, 95% CI = 0.41-0.73, P < 0.0001). Nausea and vomiting (RR = 0.63, 95% CI = 0.52-0.77, P < 0.00001), diarrhea (RR = 0.48, 95% CI = 0.37-0.64, P < 0.00001), and simple digestive tract reactions (RR = 0.63, 95% CI = 0.49-0.80, P = 0.0002) also decreased with the treatment of SFI. Conclusion SFI combined with PBC for the treatment of advanced NSCLC improved the ORR, DCR, and quality of life, and reduced the incidence of myelosuppression and gastrointestinal adverse reactions. However, considering the limitations of existing evidence, further verification using high-quality RCTs is required. Systematic review registration https://inplasy.com/inplasy-2022-7-0026, identifier INPLASY202270026.
Collapse
Affiliation(s)
- Chenxi Qiao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuaihang Hu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dandan Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kangdi Cao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Zhuo Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Xinyan Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Xiumei Ma
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zheng Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Hou
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Mundi PS, Dela Cruz FS, Grunn A, Diolaiti D, Mauguen A, Rainey AR, Guillan K, Siddiquee A, You D, Realubit R, Karan C, Ortiz MV, Douglass EF, Accordino M, Mistretta S, Brogan F, Bruce JN, Caescu CI, Carvajal RD, Crew KD, Decastro G, Heaney M, Henick BS, Hershman DL, Hou JY, Iwamoto FM, Jurcic JG, Kiran RP, Kluger MD, Kreisl T, Lamanna N, Lassman AB, Lim EA, Manji GA, McKhann GM, McKiernan JM, Neugut AI, Olive KP, Rosenblat T, Schwartz GK, Shu CA, Sisti MB, Tergas A, Vattakalam RM, Welch M, Wenske S, Wright JD, Hibshoosh H, Kalinsky K, Aburi M, Sims PA, Alvarez MJ, Kung AL, Califano A. A Transcriptome-Based Precision Oncology Platform for Patient-Therapy Alignment in a Diverse Set of Treatment-Resistant Malignancies. Cancer Discov 2023; 13:1386-1407. [PMID: 37061969 PMCID: PMC10239356 DOI: 10.1158/2159-8290.cd-22-1020] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/14/2023] [Accepted: 03/14/2023] [Indexed: 04/17/2023]
Abstract
Predicting in vivo response to antineoplastics remains an elusive challenge. We performed a first-of-kind evaluation of two transcriptome-based precision cancer medicine methodologies to predict tumor sensitivity to a comprehensive repertoire of clinically relevant oncology drugs, whose mechanism of action we experimentally assessed in cognate cell lines. We enrolled patients with histologically distinct, poor-prognosis malignancies who had progressed on multiple therapies, and developed low-passage, patient-derived xenograft models that were used to validate 35 patient-specific drug predictions. Both OncoTarget, which identifies high-affinity inhibitors of individual master regulator (MR) proteins, and OncoTreat, which identifies drugs that invert the transcriptional activity of hyperconnected MR modules, produced highly significant 30-day disease control rates (68% and 91%, respectively). Moreover, of 18 OncoTreat-predicted drugs, 15 induced the predicted MR-module activity inversion in vivo. Predicted drugs significantly outperformed antineoplastic drugs selected as unpredicted controls, suggesting these methods may substantively complement existing precision cancer medicine approaches, as also illustrated by a case study. SIGNIFICANCE Complementary precision cancer medicine paradigms are needed to broaden the clinical benefit realized through genetic profiling and immunotherapy. In this first-in-class application, we introduce two transcriptome-based tumor-agnostic systems biology tools to predict drug response in vivo. OncoTarget and OncoTreat are scalable for the design of basket and umbrella clinical trials. This article is highlighted in the In This Issue feature, p. 1275.
Collapse
Affiliation(s)
- Prabhjot S. Mundi
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Filemon S. Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Adina Grunn
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Daniel Diolaiti
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Allison R. Rainey
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Kristina Guillan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Armaan Siddiquee
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Daoqi You
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Ronald Realubit
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Charles Karan
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Michael V. Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Eugene F. Douglass
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Melissa Accordino
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Suzanne Mistretta
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Frances Brogan
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Jeffrey N. Bruce
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurological Surgery, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - Cristina I. Caescu
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Richard D. Carvajal
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Katherine D Crew
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Guarionex Decastro
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, 160 Fort Washington Ave, New York, NY USA 10032
| | - Mark Heaney
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Brian S Henick
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Dawn L Hershman
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168th St. NY, NY 10032
| | - June Y. Hou
- Department of Obstetrics & Gynecology, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Fabio M. Iwamoto
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurology, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - Joseph G. Jurcic
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Ravi P. Kiran
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Surgery, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Michael D Kluger
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Surgery, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Teri Kreisl
- Novartis Five Cambridge, MA 02142, United States
| | - Nicole Lamanna
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Andrew B. Lassman
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurology, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - Emerson A. Lim
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Gulam A. Manji
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Guy M McKhann
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurological Surgery, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - James M. McKiernan
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, 160 Fort Washington Ave, New York, NY USA 10032
| | - Alfred I Neugut
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168th St. NY, NY 10032
| | - Kenneth P. Olive
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Todd Rosenblat
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Gary K. Schwartz
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Catherine A Shu
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Michael B. Sisti
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurological Surgery, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
- Department of Otolaryngology Head and Neck Surgery, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
- Department of Radiation Oncology, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY 10032, United States
| | - Ana Tergas
- Department of Obstetrics & Gynecology, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Reena M Vattakalam
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Obstetrics & Gynecology, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Mary Welch
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurology, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - Sven Wenske
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, 160 Fort Washington Ave, New York, NY USA 10032
| | - Jason D. Wright
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Obstetrics & Gynecology, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Kevin Kalinsky
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Winship Cancer Institute of Emory University and Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365-C Clifton Road NE, Atlanta, GA 30322, United States
| | - Mahalaxmi Aburi
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, 701 W 168th Street, New York, NY USA 10032
| | - Mariano J. Alvarez
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- DarwinHealth Inc. New York
| | - Andrew L. Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Andrea Califano
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, 701 W 168th Street, New York, NY USA 10032
- Department of Biomedical Informatics, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
- J.P. Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| |
Collapse
|
9
|
Tutino F, Giovannini E, Chiola S, Giovacchini G, Ciarmiello A. Assessment of Response to Immunotherapy in Patients with Hodgkin Lymphoma: Towards Quantifying Changes in Tumor Burden Using FDG-PET/CT. J Clin Med 2023; 12:jcm12103498. [PMID: 37240602 DOI: 10.3390/jcm12103498] [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: 03/10/2023] [Revised: 04/25/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Immune checkpoint inhibitors are currently the standard of care for many advanced solid tumors, and they have been recently approved for the treatment of relapsed/refractory Hodgkin lymphoma and primary mediastinal B cell lymphoma. Assessments of the response to immunotherapy may be complicated by the occurrence of the flare/pseudoprogression phenomenon, consisting of initial tumor enlargement and even the appearance of new lesions, followed by a response, which may initially be indistinguishable from true progression. There have been efforts to characterize and capture the new patterns of response observed during immunotherapy, namely, pseudoprogression and delayed response, and several immune-related response criteria have been proposed. Confirming progression on a subsequent scan and measuring the total tumor burden are both common in immune-related criteria. Due to the peculiarity of hematologic malignancies, lymphoma-specific immune-related criteria have been developed (LYRIC), and they have been evaluated in research studies in comparison to the Lugano Classification. In this review work, we illustrate the evolution of the response criteria in lymphomas from the first CT-based criteria to the development of the PET-based Lugano Classification, further refined to take into account the flare phenomenon encountered during immunotherapy. We also describe the additional contribution of PET-derived volumetric parameters to the interpretation of responses during immunotherapy.
Collapse
Affiliation(s)
- Francesca Tutino
- Nuclear Medicine Unit, Ospedale Civile Sant'Andrea, Via Vittorio Veneto 170, 19124 La Spezia, Italy
| | - Elisabetta Giovannini
- Nuclear Medicine Unit, Ospedale Civile Sant'Andrea, Via Vittorio Veneto 170, 19124 La Spezia, Italy
| | - Silvia Chiola
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Giampiero Giovacchini
- Nuclear Medicine Unit, Ospedale Civile Sant'Andrea, Via Vittorio Veneto 170, 19124 La Spezia, Italy
| | - Andrea Ciarmiello
- Nuclear Medicine Unit, Ospedale Civile Sant'Andrea, Via Vittorio Veneto 170, 19124 La Spezia, Italy
| |
Collapse
|
10
|
Intra- and Inter-Reader Variations in Lung Nodule Measurements: Influences of Nodule Size, Location, and Observers. Diagnostics (Basel) 2022; 12:diagnostics12102319. [PMID: 36292008 PMCID: PMC9600531 DOI: 10.3390/diagnostics12102319] [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: 08/29/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Background: Accurate measurement of lung-nodule size is necessary, but whether a three-dimensional volume measurement is better or more reliable than the one-dimensional method is still unclear. This study aimed to investigate the intra- and inter-reader variations according to nodule type, size, three-dimensional volume measurements, and one-dimensional linear measurements. (2) Methods: This retrospective study included computed tomography (CT) examinations of lung nodules and volume measurements performed from October to December 2016. Two radiologists independently performed all measurements. Intra-class correlation coefficients (ICC) and Bland-Altman plots were used for analysis. (3) Results: The overall variability in the calculated volume was larger than when using the semiautomatic volume measurement. Nodules <6 mm tended to have larger variability than nodules ≥6 mm in both one-dimensional and calculated volume measurements. The isolated type showed smaller variability in both intra- and inter-reader comparisons. The juxta-vascular type showed the largest variability in both one-dimensional and calculated volume measurements. The variability was decreased when using the 3D volume semiautomated software. (4) Conclusions: The present study suggests that 3D semiautomatic volume measurements showed lower variability than the calculated volume measurement. Nodule size and location influence measurement variability. The intra- and inter-reader variabilities in nodule volume measurement were considerable.
Collapse
|
11
|
Garg T, Shrigiriwar A, Habibollahi P, Cristescu M, Liddell RP, Chapiro J, Inglis P, Camacho JC, Nezami N. Intraarterial Therapies for the Management of Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14143351. [PMID: 35884412 PMCID: PMC9322128 DOI: 10.3390/cancers14143351] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 12/11/2022] Open
Abstract
Image-guided locoregional therapies play a crucial role in the management of patients with hepatocellular carcinoma (HCC). Transarterial therapies consist of a group of catheter-based treatments where embolic agents are delivered directly into the tumor via their supplying arteries. Some of the transarterial therapies available include bland embolization (TAE), transarterial chemoembolization (TACE), drug-eluting beads-transarterial chemoembolization (DEB-TACE), selective internal radioembolization therapy (SIRT), and hepatic artery infusion (HAI). This article provides a review of pre-procedural, intra-procedural, and post-procedural aspects of each therapy, along with a review of the literature. Newer embolotherapy options and future directions are also briefly discussed.
Collapse
Affiliation(s)
- Tushar Garg
- Division of Vascular and Interventional Radiology, Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (T.G.); (R.P.L.)
| | - Apurva Shrigiriwar
- Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
| | - Peiman Habibollahi
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Mircea Cristescu
- Vascular and Interventional Radiology Division, Department of Radiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Robert P. Liddell
- Division of Vascular and Interventional Radiology, Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (T.G.); (R.P.L.)
| | - Julius Chapiro
- Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Peter Inglis
- Division of Vascular and Interventional Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Juan C. Camacho
- Department of Clinical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA;
- Vascular and Interventional Radiology, Radiology Associates of Florida, Sarasota, FL 34239, USA
| | - Nariman Nezami
- Division of Vascular and Interventional Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Experimental Therapeutics Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Correspondence:
| |
Collapse
|
12
|
Palgen JL, Perrillat-Mercerot A, Ceres N, Peyronnet E, Coudron M, Tixier E, Illigens BMW, Bosley J, L’Hostis A, Monteiro C. Integration of Heterogeneous Biological Data in Multiscale Mechanistic Model Calibration: Application to Lung Adenocarcinoma. Acta Biotheor 2022; 70:19. [PMID: 35796890 PMCID: PMC9261258 DOI: 10.1007/s10441-022-09445-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 06/15/2022] [Indexed: 11/26/2022]
Abstract
Mechanistic models are built using knowledge as the primary information source, with well-established biological and physical laws determining the causal relationships within the model. Once the causal structure of the model is determined, parameters must be defined in order to accurately reproduce relevant data. Determining parameters and their values is particularly challenging in the case of models of pathophysiology, for which data for calibration is sparse. Multiple data sources might be required, and data may not be in a uniform or desirable format. We describe a calibration strategy to address the challenges of scarcity and heterogeneity of calibration data. Our strategy focuses on parameters whose initial values cannot be easily derived from the literature, and our goal is to determine the values of these parameters via calibration with constraints set by relevant data. When combined with a covariance matrix adaptation evolution strategy (CMA-ES), this step-by-step approach can be applied to a wide range of biological models. We describe a stepwise, integrative and iterative approach to multiscale mechanistic model calibration, and provide an example of calibrating a pathophysiological lung adenocarcinoma model. Using the approach described here we illustrate the successful calibration of a complex knowledge-based mechanistic model using only the limited heterogeneous datasets publicly available in the literature.
Collapse
Affiliation(s)
| | | | - Nicoletta Ceres
- Novadiscovery, Pl. Giovanni da Verrazzano, Lyon, 69009 Rhône France
| | | | - Matthieu Coudron
- Novadiscovery, Pl. Giovanni da Verrazzano, Lyon, 69009 Rhône France
| | - Eliott Tixier
- Novadiscovery, Pl. Giovanni da Verrazzano, Lyon, 69009 Rhône France
| | - Ben M. W. Illigens
- Novadiscovery, Pl. Giovanni da Verrazzano, Lyon, 69009 Rhône France
- Dresden International University, Freiberger Str. 37, Dresden, 01067 Germany
| | - Jim Bosley
- Novadiscovery, Pl. Giovanni da Verrazzano, Lyon, 69009 Rhône France
| | - Adèle L’Hostis
- Novadiscovery, Pl. Giovanni da Verrazzano, Lyon, 69009 Rhône France
| | - Claudio Monteiro
- Novadiscovery, Pl. Giovanni da Verrazzano, Lyon, 69009 Rhône France
| |
Collapse
|
13
|
Wang B, Dong Y, Yu X, Li F, Wang J, Chen H, Niu Z, Song Y, Yuan Z, Tao Z. Safety and Efficacy of Stereotactic Ablative Radiotherapy for Ultra-Central Lung Cancer. Front Oncol 2022; 12:868844. [PMID: 35600391 PMCID: PMC9118536 DOI: 10.3389/fonc.2022.868844] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundUltra-central lung cancer (UCLC) is difficult to achieve surgical treatment. Over the past few years, stereotactic ablative radiotherapy (SABR) or stereotactic body radiotherapy (SBRT) obviously improved the clinical efficacy and survival of UCLC patients. However, the adapted scheme of radiation therapy is still controversial. For this, a single arm retrospective analysis was performed on UCLC patients treated with SBRT.Material and MethodsWe retrospectively studied primary UCLC patients who were treated with SBRT of 56 Gy/6-8f between 2010 and 2018. UCLC was defined as planning target volume (PTV) touching or overlapping the proximal bronchial tree, trachea, esophagus, heart, pulmonary vein, or pulmonary artery within 2 cm around the bronchial tree in all directions.ResultsA total of 58 patients whose median age was 68 years (range, 46-85) were included in our study, 79.3% of whom did not undergo any previous therapy. The median dose of the PTV was 77.8 Gy (range, 43.3-91.8), and the median PTV of tumors was 6.2 cm3 (range, 12.9-265.0). With a median follow-up of 57 months (range, 6-90 months), the median cumulative overall survival (OS) rate was 58 months (range, 2-105). In addition, the 1-year, 2-year and 5-year OS rates were 94.7%, 75.0% and 45.0%, respectively. In our univariable analysis (p=0.020) and multivariate analysis (p=0.004), the OS rate was associated with the PTV. The 5-year OS rates for PTV <53.0 cm3 and PTV ≥53.0 cm3 were 61.6% and 37.4%, respectively. Regarding toxicity after SBRT, there were two cases (3.5%) with grade ≥3 adverse events, of which 1 case died of sudden severe unexplained hemoptysis.ConclusionsPatients with UCLC can benefit from SBRT at a dose of 56 Gy/6-8f. On the other hand, smaller PTV was associated with superior outcomes, and the cure difference needs to be validated by prospective comparative trials.
Collapse
Affiliation(s)
- Bin Wang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yang Dong
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xuyao Yu
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fengtong Li
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jingsheng Wang
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Huaming Chen
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zeqian Niu
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yongchun Song
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhiyong Yuan
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- *Correspondence: Zhiyong Yuan, ; Zhen Tao,
| | - Zhen Tao
- Department of Radiation Oncology, CyberKnife Center and Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- *Correspondence: Zhiyong Yuan, ; Zhen Tao,
| |
Collapse
|
14
|
Ma LQ, Wu HX, Kong XQ, Fei ZD, Fang WN, Du KX, Chen F, Zhao D, Wu ZP. Which evaluation criteria of the short-term efficacy can better reflect the long-term outcomes for patients with nasopharyngeal carcinoma? Transl Oncol 2022; 20:101412. [PMID: 35395603 PMCID: PMC8987992 DOI: 10.1016/j.tranon.2022.101412] [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: 01/28/2022] [Revised: 03/17/2022] [Accepted: 03/27/2022] [Indexed: 12/08/2022] Open
Abstract
1D, 2D, and 3D measurements were all significantly correlated with PTV measurement. The 1D measurement more closely agreed with the PTV measurement than the 2D and 3D measurements. 1D tumor response assessment of the short-term efficacy can reflect the PFS for patients with nasopharyngeal carcinoma.
Purpose To compare the consistency of one-dimensional Response Evaluation Criteria in Solid Tumors (1D-RECIST), two-dimensional WHO criteria (2D-WHO), and three-dimensional (3D) measurement for therapeutic response assessment of nasopharyngeal carcinoma (NPC). Materials and methods Retrospective data of 288 newly diagnosed NPC patients were reviewed. Tumor size was assessed on magnetic resonance imaging (MRI) according to the 1D-RECIST, 2D-WHO, and 3D measurement criteria. Agreement between tumor responses was assessed using unweighted k statistics. The receiver operating characteristic (ROC) curve was used to determine the optimal cut-off point of the PTV. The Kaplan–Meier method and Cox regression were used for the survival analysis. Results The optimal cut-off point of the PTV for progression-free survival (PFS) was 29.6%. Agreement with PTV measurement was better for 1D measurement than for 2D and 3D measurements (kappa values of 0.646, 0.537, and 0.577 for 1D, 2D, and 3D measurements, respectively; P < 0.05). The area under the curve of the 1D measurement (AUC=0.596) was similar to that of the PTV measurement (AUC=0.621). Compared with 2D and 3D measurements, 1D measurement is superior for predicting prognosis in NPC (C-index of 0.672, 0.663, and 0.646 were for 1D, 2D, and 3D measurements, respectively; P < 0.005). Survival analysis showed that patients with non-responders had worse prognosis (P < 0.05). Conclusions The 1D measurement more closely agreed with the PTV measurement than the 2D and 3D measurements for predicting therapeutic responses in NPC. Therefore, we recommend using the less time-consuming 1D-RECIST criteria in routine clinical practice.
Collapse
Affiliation(s)
- Li-Qin Ma
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou 350128, China.
| | - Hai-Xia Wu
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou 350128, China
| | - Xiang-Quan Kong
- Department of Radiation Oncology, Xiamen Humanity Hospital Fujian Medical University, Xiamen 361016, China
| | - Zhao-Dong Fei
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Wei-Ning Fang
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Kai-Xin Du
- Department of Radiation Oncology, Xiamen Humanity Hospital Fujian Medical University, Xiamen 361016, China
| | - Fei Chen
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou 350128, China
| | - Dan Zhao
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou 350128, China
| | - Zhu-Peng Wu
- College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou 350128, China
| |
Collapse
|
15
|
Lazow MA, Nievelstein MT, Lane A, Bandopadhayhay P, DeWire-Schottmiller M, Fouladi M, Glod JW, Greiner RJ, Hoffman LM, Hummel TR, Kilburn L, Leary S, Minturn JE, Packer R, Ziegler DS, Chaney B, Black K, de Blank P, Leach JL. Volumetric endpoints in diffuse intrinsic pontine glioma: comparison to cross-sectional measures and outcome correlations in the International DIPG/DMG Registry. Neuro Oncol 2022; 24:1598-1608. [PMID: 35148393 PMCID: PMC9435485 DOI: 10.1093/neuonc/noac037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Cross-sectional tumor measures are traditional clinical trial endpoints; however volumetric measures may better assess tumor growth. We determined the correlation and compared the prognostic impact of cross-sectional and volumetric measures of progressive disease (PD) among patients with DIPG. METHODS Imaging and clinical data were abstracted from the International DIPG Registry. Tumor volume and cross-sectional product (CP) were measured with mint Lesion™ software using manual contouring. Correlation between CP and volume (segmented and mathematical [ellipsoid] model) thresholds of PD were assessed by linear regression. Landmark analyses determined differences in survival (via log-rank) between patients classified as PD versus non-PD by CP and volumetric measurements at 1, 3, 5, 7, and 9 months postradiotherapy (RT). Hazard ratios (HR) for survival after these time points were calculated by Cox regression. RESULTS A total of 312 MRIs (46 patients) were analyzed. Comparing change from the previous smallest measure, CP increase of 25% (PD) correlated with a segmented volume increase of 30% (R2 = 0.710), rather than 40% (spherical model extrapolation). CP-determined PD predicted survival at 1 month post-RT (HR = 2.77), but not other time points. Segmented volumetric-determined PD (40% threshold) predicted survival at all imaging timepoints (HRs = 2.57, 2.62, 3.35, 2.71, 16.29), and 30% volumetric PD threshold predicted survival at 1, 3, 5, and 9 month timepoints (HRs = 2.57, 2.62, 4.65, 5.54). Compared to ellipsoid volume, segmented volume demonstrated superior survival associations. CONCLUSIONS Segmented volumetric assessments of PD correlated better with survival than CP or ellipsoid volume at most time points. Semiautomated tumor volume likely represents a more accurate, prognostically-relevant measure of disease burden in DIPG.
Collapse
Affiliation(s)
| | | | - Adam Lane
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | - Maryam Fouladi
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, Ohio, USA,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - John W Glod
- Cancer for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Robert J Greiner
- Division of Oncology, Penn State Health Children’s Hospital, Hershey, Pennsylvania, USA
| | - Lindsey M Hoffman
- Division of Oncology, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - Trent R Hummel
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lindsay Kilburn
- Division of Oncology, Children’s National Medical Center, Washington, DC, USA
| | - Sarah Leary
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Jane E Minturn
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Roger Packer
- Division of Oncology, Children’s National Medical Center, Washington, DC, USA
| | - David S Ziegler
- Kids Cancer Centre, Sydney Children’s Hospital, Sydney, NSW, Australia,School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW, Australia
| | - Brooklyn Chaney
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Katie Black
- Brain Tumor Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - James L Leach
- Corresponding Author: James L. Leach, MD, Department of Radiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue Cincinnati, OH 45229, USA ()
| |
Collapse
|
16
|
Fournier L, de Geus-Oei LF, Regge D, Oprea-Lager DE, D’Anastasi M, Bidaut L, Bäuerle T, Lopci E, Cappello G, Lecouvet F, Mayerhoefer M, Kunz WG, Verhoeff JJC, Caruso D, Smits M, Hoffmann RT, Gourtsoyianni S, Beets-Tan R, Neri E, deSouza NM, Deroose CM, Caramella C. Twenty Years On: RECIST as a Biomarker of Response in Solid Tumours an EORTC Imaging Group - ESOI Joint Paper. Front Oncol 2022; 11:800547. [PMID: 35083155 PMCID: PMC8784734 DOI: 10.3389/fonc.2021.800547] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Response evaluation criteria in solid tumours (RECIST) v1.1 are currently the reference standard for evaluating efficacy of therapies in patients with solid tumours who are included in clinical trials, and they are widely used and accepted by regulatory agencies. This expert statement discusses the principles underlying RECIST, as well as their reproducibility and limitations. While the RECIST framework may not be perfect, the scientific bases for the anticancer drugs that have been approved using a RECIST-based surrogate endpoint remain valid. Importantly, changes in measurement have to meet thresholds defined by RECIST for response classification within thus partly circumventing the problems of measurement variability. The RECIST framework also applies to clinical patients in individual settings even though the relationship between tumour size changes and outcome from cohort studies is not necessarily translatable to individual cases. As reproducibility of RECIST measurements is impacted by reader experience, choice of target lesions and detection/interpretation of new lesions, it can result in patients changing response categories when measurements are near threshold values or if new lesions are missed or incorrectly interpreted. There are several situations where RECIST will fail to evaluate treatment-induced changes correctly; knowledge and understanding of these is crucial for correct interpretation. Also, some patterns of response/progression cannot be correctly documented by RECIST, particularly in relation to organ-site (e.g. bone without associated soft-tissue lesion) and treatment type (e.g. focal therapies). These require specialist reader experience and communication with oncologists to determine the actual impact of the therapy and best evaluation strategy. In such situations, alternative imaging markers for tumour response may be used but the sources of variability of individual imaging techniques need to be known and accounted for. Communication between imaging experts and oncologists regarding the level of confidence in a biomarker is essential for the correct interpretation of a biomarker and its application to clinical decision-making. Though measurement automation is desirable and potentially reduces the variability of results, associated technical difficulties must be overcome, and human adjudications may be required.
Collapse
Affiliation(s)
- Laure Fournier
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Université de Paris, Assistance Publique–Hôpitaux de Paris (AP-HP), Hopital europeen Georges Pompidou, Department of Radiology, Paris Cardiovascular Research Center (PARCC) Unité Mixte de Recherche (UMRS) 970, Institut national de la santé et de la recherche médicale (INSERM), Paris, France
| | - Lioe-Fee de Geus-Oei
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Daniele Regge
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Surgical Sciences, University of Turin, Turin, Italy
- Radiology Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-Istituto Di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Turin, Italy
| | - Daniela-Elena Oprea-Lager
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Cancer Centre Amsterdam, Amsterdam University Medical Centers [Vrije Universiteit (VU) University], Amsterdam, Netherlands
| | - Melvin D’Anastasi
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Medical Imaging Department, Mater Dei Hospital, University of Malta, Msida, Malta
| | - Luc Bidaut
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- College of Science, University of Lincoln, Lincoln, United Kingdom
| | - Tobias Bäuerle
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Egesta Lopci
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine Unit, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) – Humanitas Research Hospital, Milan, Italy
| | - Giovanni Cappello
- Department of Surgical Sciences, University of Turin, Turin, Italy
- Radiology Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-Istituto Di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Turin, Italy
| | - Frederic Lecouvet
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Marius Mayerhoefer
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang G. Kunz
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Joost J. C. Verhoeff
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Damiano Caruso
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Marion Smits
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
- Brain Tumour Centre, Erasmus Medical Centre (MC) Cancer Institute, Rotterdam, Netherlands
| | - Ralf-Thorsten Hoffmann
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Institute and Policlinic for Diagnostic and Interventional Radiology, University Hospital, Carl-Gustav-Carus Technical University Dresden, Dresden, Germany
| | - Sofia Gourtsoyianni
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, Areteion Hospital, Athens, Greece
| | - Regina Beets-Tan
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- School For Oncology and Developmental Biology (GROW) School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Emanuele Neri
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Diagnostic and Interventional Radiology, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Nandita M. deSouza
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, United States
| | - Christophe M. Deroose
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Caroline Caramella
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Radiology Department, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph Centre International des Cancers Thoraciques, Université Paris-Saclay, Le Plessis-Robinson, France
| |
Collapse
|
17
|
Woodford RG, Zhou DDX, Kok PS, Lord SJ, Friedlander M, Marschner IC, Simes RJ, Lee CK. The validity of progression-free survival 2 as a surrogate trial end point for overall survival. Cancer 2022; 128:1449-1457. [PMID: 34985773 DOI: 10.1002/cncr.34085] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/01/2021] [Accepted: 10/11/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Overall survival (OS) is the gold-standard end point for oncology trials. However, the availability of multiple therapeutic options after progression and crossover to receive investigational agents confound and delay OS data maturation. Progression-free survival 2 (PFS-2), defined as the time from randomization to progression on first subsequent therapy, has been proposed as a surrogate for OS. Using a meta-analytic approach, the authors aimed to assess the association between OS and PFS-2 and compare this with progression-free survival 1 (PFS-1) and the objective response rate (ORR). METHODS An electronic literature search was performed to identify randomized trials of systemic therapies in advanced solid tumors that reported PFS-2 as a prespecified end point. Correlations between OS and PFS-2, OS and PFS-1, and OS and ORR as hazard ratios (HRs) or odds ratios (ORs) were assessed via linear regression weighted by trial size. RESULTS Thirty-eight trials were included, and they comprised 19,031 patients across 8 tumor types. PFS-2 displayed a moderate correlation with OS (r = 0.67; 95% confidence interval [CI], 0.08-0.69). Conversely, correlations of ORR (r = 0.12; 95% CI, 0.00-0.13) and PFS-1 (r = 0.21; 95% CI, 0.00-0.33) were poor. The findings for PFS-2 were consistent for subgroup analyses by treatment type (immunotherapy vs nonimmunotherapy: r = 0.67 vs 0.67), survival post progression (<12 vs ≥12 months: r = 0.86 vs 0.79), and percentage not receiving subsequent treatment (<50% vs ≥50%: r = 0.70 vs 0.63). CONCLUSIONS Across diverse tumors and therapies, the treatment effect on PFS-2 correlated moderately with the treatment effect on OS. PFS-2 performed consistently better than PFS-1 and ORR, regardless of postprogression treatment and postprogression survival. PFS-2 should be included as a key trial end point in future randomized trials of solid tumors.
Collapse
Affiliation(s)
- Rachel G Woodford
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia.,St George Cancer Care Centre, Sydney, New South Wales, Australia
| | - Deborah D-X Zhou
- Chris O'Brien Lifehouse, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Peey-Sei Kok
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Sally J Lord
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Michael Friedlander
- Nelune Cancer Centre, Prince of Wales Hospital and Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Ian C Marschner
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - R John Simes
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Chee Khoon Lee
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia.,St George Cancer Care Centre, Sydney, New South Wales, Australia
| |
Collapse
|
18
|
Zhao L, Liu L, Zhao H, Bao J, Dou Y, Yang Z, Lin Y, Sun Z, Meng L, Yan L, Liu A. Therapy response assessment of non-small cell lung cancer using dual-energy computed tomography iodine map. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2022; 30:111-122. [PMID: 34719473 DOI: 10.3233/xst-210989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To investigate feasibility of the quantitative parameters of dual-energy computed tomography (DECT) to assess therapy response in advanced non-small cell lung cancer (NSCLC) compared with the traditional enhanced CT parameters based on the Response Evaluation Criteria in Solid Tumors (RECIST) guidelines. METHODS Forty-five patients with unresectable locally advanced NSCLC who underwent DECT before and after chemotherapy or concurrent chemoradiotherapy (cCRT) were prospectively enrolled. By comparing baseline studies with follow-up, patients were divided into two groups according to RECIST guidelines as follows: disease control (DC, including partial response and stable disease) and progressive disease (PD). The diameter (D), attenuation, iodine concentration and normalized iodine concentration of arterial and venous phases (ICA, ICv, NICA, NICv) and the percentage of these changes pre- and post-therapy were measured and calculated. The Pearson correlation was used to analyze correlation between various quantitative parameters. The receiver operating characteristic (ROC) curves were used to evaluate accuracy of therapy response prediction. RESULTS The change percentages of Attenuation (Δ-Attenuation-A and Δ-Attenuation-V), IC (ΔICA and ΔICV) and NIC (ΔNICA and ΔNICV) pre- and post-therapy correlate with the change percentage of D (ΔD). Among these, ΔICA strongly correlates with ΔD (r = 0.793, P < 0.001). The areas under ROC curves generated using Δ-Attenuation-A, ΔICA, and ΔNICA are 0.796, 0.900, and 0.880 with the corresponding cutoff value of 9.096, -15.692, and -4.7569, respectively, which are significantly different (P < 0.001). CONCLUSIONS The quantitative parameters of DECT iodine map, especially iodine concentration, in arterial phase provides a new quantitative image marker to predict therapy response of patients diagnosed with advanced NSCLC.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Radiology, the Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Lijuan Liu
- Department of Radiology, the Affiliated Beijing Chuiyangliu Hospital of Tsinghua University, Beijing, China
| | - Haiyan Zhao
- Department of Oncology, the Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Jiaqi Bao
- Department of Oncology, Inner Mongolia People's Hospital, Inner Mongolia, China
| | - Yana Dou
- Department of Scientific Marketing, Siemens Healthineers AG, China
| | - Zhenxing Yang
- Department of Radiology, the Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Yang Lin
- Department of Scientific Marketing, Siemens Healthineers AG, China
| | - Zhenting Sun
- Department of Radiology, the Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Lingxin Meng
- Department of Radiology, the Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Li Yan
- Department of Respiratory, the Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Aishi Liu
- Department of Radiology, the Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| |
Collapse
|
19
|
Tyan K, Bae JE, Lorch JH, Margalit DN, Tishler RB, Huynh MA, Jo VY, Haddad RI, Chau NG, Hanna GJ, Schoenfeld JD. Oligometastatic adenoid cystic carcinoma: Correlating tumor burden and time to treatment with outcomes. Head Neck 2021; 44:722-734. [DOI: 10.1002/hed.26964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 11/12/2021] [Accepted: 12/10/2021] [Indexed: 12/25/2022] Open
Affiliation(s)
- Kevin Tyan
- Harvard Medical School Boston Massachusetts USA
- Department of Medical Oncology Dana‐Farber Cancer Institute Boston Massachusetts USA
- Department of Radiation Oncology Dana‐Farber Cancer Institute and Brigham & Women's Hospital Boston Massachusetts USA
| | - Ji Eun Bae
- Department of Medical Oncology Dana‐Farber Cancer Institute Boston Massachusetts USA
| | - Jochen H. Lorch
- Department of Medical Oncology Dana‐Farber Cancer Institute Boston Massachusetts USA
| | - Danielle N. Margalit
- Department of Radiation Oncology Dana‐Farber Cancer Institute and Brigham & Women's Hospital Boston Massachusetts USA
| | - Roy B. Tishler
- Department of Radiation Oncology Dana‐Farber Cancer Institute and Brigham & Women's Hospital Boston Massachusetts USA
| | - Mai Anh Huynh
- Department of Radiation Oncology Dana‐Farber Cancer Institute and Brigham & Women's Hospital Boston Massachusetts USA
| | - Vickie Y. Jo
- Department of Pathology Brigham & Women's Hospital Boston Massachusetts USA
| | - Robert I. Haddad
- Department of Medical Oncology Dana‐Farber Cancer Institute Boston Massachusetts USA
| | - Nicole G. Chau
- Department of Medical Oncology Dana‐Farber Cancer Institute Boston Massachusetts USA
- BC Cancer Vancouver Center Vancouver British Columbia Canada
| | - Glenn J. Hanna
- Department of Medical Oncology Dana‐Farber Cancer Institute Boston Massachusetts USA
| | - Jonathan D. Schoenfeld
- Department of Radiation Oncology Dana‐Farber Cancer Institute and Brigham & Women's Hospital Boston Massachusetts USA
| |
Collapse
|
20
|
O'Donohue TJ, Ibáñez G, Coutinho DF, Mauguen A, Siddiquee A, Rosales N, Calder P, Ndengu A, You D, Long M, Roberts SS, Kung AL, Dela Cruz FS. Translational Strategies for Repotrectinib in Neuroblastoma. Mol Cancer Ther 2021; 20:2189-2197. [PMID: 34482287 DOI: 10.1158/1535-7163.mct-21-0126] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/06/2021] [Accepted: 08/26/2021] [Indexed: 11/16/2022]
Abstract
Limited clinical data are available regarding the utility of multikinase inhibition in neuroblastoma. Repotrectinib (TPX-0005) is a multikinase inhibitor that targets ALK, TRK, JAK2/STAT, and Src/FAK, which have all been implicated in the pathogenesis of neuroblastoma. We evaluated the preclinical activity of repotrectinib monotherapy and in combination with chemotherapy as a potential therapeutic approach for relapsed/refractory neuroblastoma. In vitro sensitivity to repotrectinib, ensartinib, and cytotoxic chemotherapy was evaluated in neuroblastoma cell lines. In vivo antitumor effect of repotrectinib monotherapy, and in combination with chemotherapy, was evaluated using a genotypically diverse cohort of patient-derived xenograft (PDX) models of neuroblastoma. Repotrectinib had comparable cytotoxic activity across cell lines irrespective of ALK mutational status. Combination with chemotherapy demonstrated increased antiproliferative activity across several cell lines. Repotrectinib monotherapy had notable antitumor activity and prolonged event-free survival compared with vehicle and ensartinib in PDX models (P < 0.05). Repotrectinib plus chemotherapy was superior to chemotherapy alone in ALK-mutant and ALK wild-type PDX models. These results demonstrate that repotrectinib has antitumor activity in genotypically diverse neuroblastoma models, and that combination of a multikinase inhibitor with chemotherapy may be a promising treatment paradigm for translation to the clinic.
Collapse
Affiliation(s)
- Tara J O'Donohue
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Glorymar Ibáñez
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Armaan Siddiquee
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nestor Rosales
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul Calder
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andoyo Ndengu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daoqi You
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew Long
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Filemon S Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
21
|
Ramesh K, Gurbani SS, Mellon EA, Huang V, Goryawala M, Barker PB, Kleinberg L, Shu HKG, Shim H, Weinberg BD. The Longitudinal Imaging Tracker (BrICS-LIT):A Cloud Platform for Monitoring Treatment Response in Glioblastoma Patients. ACTA ACUST UNITED AC 2021; 6:93-100. [PMID: 32548285 PMCID: PMC7289246 DOI: 10.18383/j.tom.2020.00001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Glioblastoma is a common and aggressive form of brain cancer affecting up to 20,000 new patients in the US annually. Despite rigorous therapies, current median survival is only 15-20 months. Patients who complete initial treatment undergo follow-up imaging at routine intervals to assess for tumor recurrence. Imaging is a central part of brain tumor management, but MRI findings in patients with brain tumor can be challenging to interpret and are further confounded by interpretation variability. Disease-specific structured reporting attempts to reduce variability in imaging results by implementing well-defined imaging criteria and standardized language. The Brain Tumor Reporting and Data System (BT-RADS) is one such framework streamlined for clinical workflows and includes quantitative criteria for more objective evaluation of follow-up imaging. To facilitate accurate and objective monitoring of patients during the follow-up period, we developed a cloud platform, the Brain Imaging Collaborative Suite's Longitudinal Imaging Tracker (BrICS-LIT). BrICS-LIT uses semiautomated tumor segmentation algorithms of both T2-weighted FLAIR and contrast-enhanced T1-weighted MRI to assist clinicians in quantitative assessment of brain tumors. The LIT platform can ultimately guide clinical decision-making for patients with glioblastoma by providing quantitative metrics for BT-RADS scoring. Further, this platform has the potential to increase objectivity when measuring efficacy of novel therapies for patients with brain tumor during their follow-up. Therefore, LIT will be used to track patients in a dose-escalated clinical trial, where spectroscopic MRI has been used to guide radiation therapy (Clinicaltrials.gov NCT03137888), and compare patients to a control group that received standard of care.
Collapse
Affiliation(s)
- Karthik Ramesh
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA.,Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA
| | - Saumya S Gurbani
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA.,Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA
| | - Eric A Mellon
- Departments of Radiation Oncology, Sylvester Comprehensive Cancer Center; and
| | - Vicki Huang
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA.,Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA
| | | | | | | | - Hui-Kuo G Shu
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA.,Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA.,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - Brent D Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
22
|
Shafiei A, Bagheri M, Farhadi F, Apolo AB, Biassou NM, Folio LR, Jones EC, Summers RM. CT Evaluation of Lymph Nodes That Merge or Split during the Course of a Clinical Trial: Limitations of RECIST 1.1. Radiol Imaging Cancer 2021; 3:e200090. [PMID: 33874734 PMCID: PMC8189184 DOI: 10.1148/rycan.2021200090] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 02/11/2021] [Accepted: 02/16/2021] [Indexed: 06/12/2023]
Abstract
Purpose To compare Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 with volumetric measurement in the setting of target lymph nodes that split into two or more nodes or merge into one conglomerate node. Materials and Methods In this retrospective study, target lymph nodes were evaluated on CT scans from 166 patients with different types of cancer; 158 of the scans came from The Cancer Imaging Archive. Each target node was measured using RECIST 1.1 criteria before and after merging or splitting, followed by volumetric segmentation. To compare RECIST 1.1 with volume, a single-dimension hypothetical diameter (HD) was determined from the nodal volume. The nodes were divided into three groups: (a) one-target merged (one target node merged with other nodes); (b) two-target merged (two neighboring target nodes merged); and (c) split node (a conglomerate node cleaved into smaller fragments). Bland-Altman analysis and t test were applied to compare RECIST 1.1 with HD. On the basis of the RECIST 1.1 concept, we compared response category changes between RECIST 1.1 and HD. Results The data set consisted of 30 merged nodes (19 one-target merged and 11 two-target merged) and 20 split nodes (mean age for all 50 included patients, 50 years ± 7 [standard deviation]; 38 men). RECIST 1.1, volumetric, and HD measurements indicated an increase in size in all one-target merged nodes. While volume and HD indicated an increase in size for nodes in the two-target merged group, RECIST 1.1 showed a decrease in size in all two-target merged nodes. Although volume and HD demonstrated a decrease in size of all split nodes, RECIST 1.1 indicated an increase in size in 60% (12 of 20) of the nodes. Discrepancy of the response categories between RECIST 1.1 and HD was observed in 5% (one of 19) in one-target merged, 82% (nine of 11) in two-target merged, and 55% (11 of 20) in split nodes. Conclusion RECIST 1.1 does not optimally reflect size changes when lymph nodes merge or split. Keywords: CT, Lymphatic, Tumor Response Supplemental material is available for this article. © RSNA, 2021.
Collapse
|
23
|
Oberg AL, Heinzen EP, Hou X, Al Hilli MM, Hurley RM, Wahner Hendrickson AE, Goergen KM, Larson MC, Becker MA, Eckel-Passow JE, Maurer MJ, Kaufmann SH, Haluska P, Weroha SJ. Statistical analysis of comparative tumor growth repeated measures experiments in the ovarian cancer patient derived xenograft (PDX) setting. Sci Rep 2021; 11:8076. [PMID: 33850213 PMCID: PMC8044116 DOI: 10.1038/s41598-021-87470-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Repeated measures studies are frequently performed in patient-derived xenograft (PDX) models to evaluate drug activity or compare effectiveness of cancer treatment regimens. Linear mixed effects regression models were used to perform statistical modeling of tumor growth data. Biologically plausible structures for the covariation between repeated tumor burden measurements are explained. Graphical, tabular, and information criteria tools useful for choosing the mean model functional form and covariation structure are demonstrated in a Case Study of five PDX models comparing cancer treatments. Power calculations were performed via simulation. Linear mixed effects regression models applied to the natural log scale were shown to describe the observed data well. A straight growth function fit well for two PDX models. Three PDX models required quadratic or cubic polynomial (time squared or cubed) terms to describe delayed tumor regression or initial tumor growth followed by regression. Spatial(power), spatial(power) + RE, and RE covariance structures were found to be reasonable. Statistical power is shown as a function of sample size for different levels of variation. Linear mixed effects regression models provide a unified and flexible framework for analysis of PDX repeated measures data, use all available data, and allow estimation of tumor doubling time.
Collapse
Affiliation(s)
- Ann L Oberg
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Ethan P Heinzen
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Xiaonan Hou
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mariam M Al Hilli
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Division of Subspecialty Care for Women's Health, Gynecologic Oncology, Women's Health Institute, Cleveland Clinic, Cleveland, OH, 44126, USA
| | - Rachel M Hurley
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Andrea E Wahner Hendrickson
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Krista M Goergen
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Melissa C Larson
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Marc A Becker
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Blueprint Medicines, 45 Sidney St., Cambridge, MA, 02139, USA
| | - Jeanette E Eckel-Passow
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Matthew J Maurer
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Paul Haluska
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Bristol-Meyers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | - S John Weroha
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
24
|
Cho YJ, Kwon H, Kwon YJ, Kim SC, Kim DY, Namgoong JM. Effects of sirolimus in the treatment of unresectable infantile hemangioma and vascular malformations in children: A single-center experience. J Vasc Surg Venous Lymphat Disord 2021; 9:1488-1494. [PMID: 33836285 DOI: 10.1016/j.jvsv.2021.03.014] [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: 09/15/2020] [Accepted: 03/19/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Recently, sirolimus has emerged as a safe and effective treatment modality for unresectable vascular lesions. In the present study, we investigated the effectiveness and safety of sirolimus from our early experience with patients with unresectable vascular anomalies. METHODS The medical records and radiologic images of all patients with unresectable vascular anomalies treated with sirolimus at our center from January 2018 to November 2019 were retrospectively reviewed. All patients were administered oral doses of sirolimus 0.8 mg/m2 every 12 hours as the initial dose, followed by maintenance of a target serum concentration (5-10 ng/mL) with therapeutic drug monitoring. RESULTS Six patients with unresectable vascular anomalies were treated with sirolimus for ≥10 months. Their median age at the initiation of sirolimus treatment was 17 months (range, 8-67 months). The median duration of treatment was 13 months (range, 10-16 months). One patient had a good response, four had an intermediate response, and one had no response to sirolimus therapy. None of the patients had discontinued sirolimus therapy because of adverse effects. CONCLUSIONS Sirolimus can be used effectively and safely for patients with unresectable vascular anomalies. However, further prospective studies are warranted to evaluate the long-term effects of sirolimus and clarify the indications for early intervention.
Collapse
Affiliation(s)
- Yu Jeong Cho
- Department of Pediatric Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyunhee Kwon
- Department of Pediatric Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Yong Jae Kwon
- Department of Pediatric Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Seong Chul Kim
- Department of Pediatric Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Dae Yeon Kim
- Department of Pediatric Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea.
| | - Jung-Man Namgoong
- Department of Pediatric Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Kuzmanović Elabjer B, Bušić M, Pleše A, Bjeloš M, Miletić D, Vukojević N. Ultrasound Biomicroscopy Documented Anterior Uveal Melanoma Regression after Ruthenium-106 Plaque Therapy. Ocul Oncol Pathol 2021; 7:224-232. [PMID: 34307336 DOI: 10.1159/000512030] [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: 08/01/2020] [Accepted: 10/03/2020] [Indexed: 11/19/2022] Open
Abstract
Introduction Ultrasound biomicroscopy (UBM) is the only widely used method for the evaluation of anterior uveal melanoma (AUM). Objective Documentation of regression of AUM treated with ruthenium-106 (Ru-106) plaque types CCB and CCC using UBM. Methods This single institution-based retrospective case series involved 10 Caucasian patients with AUM followed after brachytherapy with UBM from January 2014 until February 2019. The largest prominence of the tumor perpendicular to the sclera or the cornea (including scleral/corneal thickness) (D) and the largest basal dimension (B) were measured in millimeters with UBM for all patients prior to the brachytherapy and at 4-month interval follow-up. Tumor regression was calculated as a percentage of decrease in the initial D and B values. Results The study involved 10 patients with a mean age of 64.4 years (yr) (range 46-80 yr). D ranged from 1.82 to 5.5 mm (median 2.99 mm) and B from 2.32 to 12.38 mm (median 4.18 mm). The apical radiation dose in all patients was 100 Gy. The median follow-up was 42.02 months. Regression for D was 21.11 ± 13.66%, 31.09 ± 14.66%, and 34.92 ± 19.86% at 1st, 2nd, and 3rd year of the follow-up, respectively, while for B it was 21.58 ± 16.05%, 28.98 ± 17.71%, and 32.06 ± 18.96%, respectively. Tumor recurrence was documented in 2/10 patients. Conclusion The major regression of AUM, treated with Ru-106 plaque types CCB and CCC, was documented in the first 2 years after brachytherapy in our study group. In the following years, only minimal regression was documented that warns of the need for close monitoring and active search for local recurrences.
Collapse
Affiliation(s)
- Biljana Kuzmanović Elabjer
- Faculty of Dental Medicine and Health Care Osijek, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia.,Faculty of Medicine Osijek, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia.,University Eye Clinic - WHO Collaborating Center, University Hospital "Sveti Duh", Zagreb, Croatia
| | - Mladen Bušić
- Faculty of Dental Medicine and Health Care Osijek, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia.,Faculty of Medicine Osijek, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia.,University Eye Clinic - WHO Collaborating Center, University Hospital "Sveti Duh", Zagreb, Croatia
| | - Andrej Pleše
- Faculty of Dental Medicine and Health Care Osijek, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia.,University Eye Clinic - WHO Collaborating Center, University Hospital "Sveti Duh", Zagreb, Croatia
| | - Mirjana Bjeloš
- Faculty of Dental Medicine and Health Care Osijek, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia.,Faculty of Medicine Osijek, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia.,University Eye Clinic - WHO Collaborating Center, University Hospital "Sveti Duh", Zagreb, Croatia
| | - Daliborka Miletić
- Faculty of Dental Medicine and Health Care Osijek, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia.,Faculty of Medicine Osijek, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia.,University Eye Clinic - WHO Collaborating Center, University Hospital "Sveti Duh", Zagreb, Croatia
| | - Nenad Vukojević
- University Eye Clinic, University Hospital Centre Zagreb, Zagreb, Croatia
| |
Collapse
|
26
|
Pediatric Rhabdomyosarcomas: Three-Dimensional Radiological Assessments after Induction Chemotherapy Predict Survival Better than One-Dimensional and Two-Dimensional Measurements. Cancers (Basel) 2020; 12:cancers12123808. [PMID: 33348683 PMCID: PMC7766999 DOI: 10.3390/cancers12123808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 11/16/2022] Open
Abstract
Radiological response to neoadjuvant chemotherapy is currently used to assess the efficacy of treatment in pediatric patients with rhabdomyosarcoma (RMS), but the association between early tumor response on imaging and survival is still controversial. The aim of this study was to investigate the prognostic value of assessing radiological response after induction therapy in pediatric RMS, comparing four different methods. This retrospective, two-center study was conducted on 66 non-metastatic RMS patients. Two radiologists measured tumor size on pre- and post-treatment magnetic resonance (MR) or computed tomography (CT) images using four methods: considering maximal diameter with the 1D-RECIST (Response Evaluation Criteria in Solid Tumors); multiplying the two maximal diameters with the 2D-WHO (World Health Organization); multiplying the three maximal diameters with the 3D-EpSSG (European pediatric Soft tissue sarcoma Study Group); obtaining a software-assisted volume assessment with the 3D-Osirix. Each patient was classified as a responder or non-responder based on the proposed thresholds for each method. Tumor response was compared with survival using Kaplan-Meier plots, the log-rank test, and Cox's regression. Agreement between methods and observers (weighted-κ) was also calculated. The 5-year event-free survival (5yr-EFS) calculated with the Kaplan-Meier plots was significantly longer for responders than for non-responders with all the methods, but the 3D assessments differentiated between the two groups better than the 1D-RECIST or 2D-WHO (p1D-RECIST = 0.018, p2D-WHO = 0.007, p3D-EpSSG and p3D-Osirix < 0.0001). Comparing the 5yr-EFS of responders and non-responders also produced adjusted hazard ratios of 3.57 (p = 0.0158) for the 1D-RECIST, 5.05 for the 2D-WHO (p = 0.0042), 14.40 for the 3D-EpSSG (p < 0.0001) and 11.60 for the 3D-Osirix (p < 0.0001), indicating that the volumetric measurements were significantly more strongly associated with EFS. Inter-method agreement was excellent between the 3D-EpSSG and the 3D-Osirix (κ = 0.98), and moderate for the other comparisons (0.5 < κ < 0.8). The 1D-RECIST and the 2D-WHO tended to underestimate response to treatment. Inter-observer agreement was excellent with all methods (κ > 0.8) except for the 2D-WHO (κ = 0.7). In conclusion, early tumor response was confirmed as a significant prognostic factor in RMS, and the 3D-EpSSG and 3D-Osirix methods predicted response to treatment better than the 1D-RECIST or 2D-WHO measurements.
Collapse
|
27
|
Gatson NTN, Bross SP, Odia Y, Mongelluzzo GJ, Hu Y, Lockard L, Manikowski JJ, Mahadevan A, Kazmi SAJ, Lacroix M, Conger AR, Vadakara J, Nayak L, Chi TL, Mehta MP, Puduvalli VK. Early imaging marker of progressing glioblastoma: a window of opportunity. J Neurooncol 2020; 148:629-640. [PMID: 32602020 DOI: 10.1007/s11060-020-03565-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/17/2020] [Indexed: 12/28/2022]
Abstract
PURPOSE Therapeutic intervention at glioblastoma (GBM) progression, as defined by current assessment criteria, is arguably too late as second-line therapies fail to extend survival. Still, most GBM trials target recurrent disease. We propose integration of a novel imaging biomarker to more confidently and promptly define progression and propose a critical timepoint for earlier intervention to extend therapeutic exposure. METHODS A retrospective review of 609 GBM patients between 2006 and 2019 yielded 135 meeting resection, clinical, and imaging inclusion criteria. We qualitatively and quantitatively analyzed 2000+ sequential brain MRIs (initial diagnosis to first progression) for development of T2 FLAIR signal intensity (SI) within the resection cavity (RC) compared to the ventricles (V) for quantitative inter-image normalization. PFS and OS were evaluated using Kaplan-Meier curves stratified by SI. Specificity and sensitivity were determined using a 2 × 2 table and pathology confirmation at progression. Multivariate analysis evaluated SI effect on the hazard rate for death after adjusting for established prognostic covariates. Recursive partitioning determined successive quantifiers and cutoffs associated with outcomes. Neurological deficits correlated with SI. RESULTS Seventy-five percent of patients developed SI on average 3.4 months before RANO-assessed progression with 84% sensitivity. SI-positivity portended neurological decline and significantly poorer outcomes for PFS (median, 10 vs. 15 months) and OS (median, 20 vs. 29 months) compared to SI-negative. RC/V ratio ≥ 4 was the most significant prognostic indicator of death. CONCLUSION Implications of these data are far-reaching, potentially shifting paradigms for glioma treatment response assessment, altering timepoints for salvage therapeutic intervention, and reshaping glioma clinical trial design.
Collapse
Affiliation(s)
- Na Tosha N Gatson
- Neuroscience Institute, Geisinger Health, Danville, PA, 17822, USA. .,Cancer Institute, Geisinger Health, Danville, PA, 17822, USA. .,Geisinger Commonwealth School of Medicine, Scranton, PA, 18509, USA. .,Geisinger Medical Center, Neuroscience Institute MC 14-03, 100 N. Academy Ave, Danville, PA, 17822, USA.
| | - Shane P Bross
- Neuroscience Institute, Geisinger Health, Danville, PA, 17822, USA
| | - Yazmin Odia
- Department of Neuro-Oncology, Miami Cancer Institute/Baptist Health South Florida, Miami, FL, 33176, USA
| | | | - Yirui Hu
- Department of Population Health Sciences, Geisinger Health, Danville, PA, 17822, USA
| | - Laura Lockard
- Geisinger Commonwealth School of Medicine, Scranton, PA, 18509, USA
| | | | - Anand Mahadevan
- Cancer Institute, Geisinger Health, Danville, PA, 17822, USA
| | - Syed A J Kazmi
- Department of Pathology, Geisinger Health, Danville, PA, 17822, USA
| | - Michel Lacroix
- Neuroscience Institute, Geisinger Health, Danville, PA, 17822, USA
| | - Andrew R Conger
- Neuroscience Institute, Geisinger Health, Danville, PA, 17822, USA.,Geisinger Commonwealth School of Medicine, Scranton, PA, 18509, USA
| | - Joseph Vadakara
- Cancer Institute, Geisinger Health, Danville, PA, 17822, USA
| | - Lakshmi Nayak
- Harvard Medical School, Center for Neuro-Oncology,, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - T Linda Chi
- Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute/Baptist Health South Florida, Miami, FL, 33176, USA
| | - Vinay K Puduvalli
- Division of Neuro-Oncology, The OH State University Comprehensive Cancer Center - James and OSU Neurological Institute, Columbus, OH, 43210, USA.,Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
28
|
Vera-Yunca D, Girard P, Parra-Guillen ZP, Munafo A, Trocóniz IF, Terranova N. Machine Learning Analysis of Individual Tumor Lesions in Four Metastatic Colorectal Cancer Clinical Studies: Linking Tumor Heterogeneity to Overall Survival. AAPS JOURNAL 2020; 22:58. [PMID: 32185612 PMCID: PMC7078147 DOI: 10.1208/s12248-020-0434-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/12/2020] [Indexed: 12/23/2022]
Abstract
Total tumor size (TS) metrics used in TS models in oncology do not consider tumor heterogeneity, which could help to better predict drug efficacy. We analyzed individual target lesions (iTLs) of patients with metastatic colorectal carcinoma (mCRC) to determine differences in TS dynamics by using the ClassIfication Clustering of Individual Lesions (CICIL) methodology. Results from subgroup analyses comparing genetic mutations and TS metrics were assessed and applied to survival analyses. Data from four mCRC clinical studies were analyzed (1781 patients, 6369 iTLs). CICIL was used to assess differences in lesion TS dynamics within a tissue (intra-class) or across different tissues (inter-class). First, lesions were automatically classified based on their location. Cross-correlation coefficients (CCs) determined if each pair of lesions followed similar or opposite dynamics. Finally, CCs were grouped by using the K-means clustering method. Heterogeneity in tumor dynamics was lower in the intra-class analysis than in the inter-class analysis for patients receiving cetuximab. More tumor heterogeneity was found in KRAS mutated patients compared to KRAS wild-type (KRASwt) patients and when using sum of longest diameters versus sum of products of diameters. Tumor heterogeneity quantified as the median patient's CC was found to be a predictor of overall survival (OS) (HR = 1.44, 95% CI 1.08-1.92), especially in KRASwt patients. Intra- and inter-tumor tissue heterogeneities were assessed with CICIL. Derived metrics of heterogeneity were found to be a predictor of OS time. Considering differences between lesions' TS dynamics could improve oncology models in favor of a better prediction of OS.
Collapse
Affiliation(s)
- Diego Vera-Yunca
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Pascal Girard
- Merck Institute for Pharmacometrics, Merck Serono S.A., Switzerland, a Subsidiary of Merck KGaA, Darmstadt, Germany
| | - Zinnia P Parra-Guillen
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Alain Munafo
- Merck Institute for Pharmacometrics, Merck Serono S.A., Switzerland, a Subsidiary of Merck KGaA, Darmstadt, Germany
| | - Iñaki F Trocóniz
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Nadia Terranova
- Merck Institute for Pharmacometrics, Merck Serono S.A., Switzerland, a Subsidiary of Merck KGaA, Darmstadt, Germany.
| |
Collapse
|
29
|
Aykan NF, Özatlı T. Objective response rate assessment in oncology: Current situation and future expectations. World J Clin Oncol 2020; 11:53-73. [PMID: 32133275 PMCID: PMC7046919 DOI: 10.5306/wjco.v11.i2.53] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 11/05/2019] [Accepted: 11/29/2019] [Indexed: 02/06/2023] Open
Abstract
The tumor objective response rate (ORR) is an important parameter to demonstrate the efficacy of a treatment in oncology. The ORR is valuable for clinical decision making in routine practice and a significant end-point for reporting the results of clinical trials. World Health Organization and Response Evaluation Criteria in Solid Tumors (RECIST) are anatomic response criteria developed mainly for cytotoxic chemotherapy. These criteria are based on the visual assessment of tumor size in morphological images provided by computed tomography (CT) or magnetic resonance imaging. Anatomic response criteria may not be optimal for biologic agents, some disease sites, and some regional therapies. Consequently, modifications of RECIST, Choi criteria and Morphologic response criteria were developed based on the concept of the evaluation of viable tumors. Despite its limitations, RECIST v1.1 is validated in prospective studies, is widely accepted by regulatory agencies and has recently shown good performance for targeted cancer agents. Finally, some alternatives of RECIST were developed as immune-specific response criteria for checkpoint inhibitors. Immune RECIST criteria are based essentially on defining true progressive disease after a confirmatory imaging. Some graphical methods may be useful to show longitudinal change in the tumor burden over time. Tumor tissue is a tridimensional heterogenous mass, and tumor shrinkage is not always symmetrical; thus, metabolic response assessments using positron emission tomography (PET) or PET/CT may reflect the viability of cancer cells or functional changes evolving after anticancer treatments. The metabolic response can show the benefit of a treatment earlier than anatomic shrinkage, possibly preventing delays in drug approval. Computer-assisted automated volumetric assessments, quantitative multimodality imaging in radiology, new tracers in nuclear medicine and finally artificial intelligence have great potential in future evaluations.
Collapse
Affiliation(s)
- Nuri Faruk Aykan
- Department of Medical Oncology, Istinye University Medical School, Bahcesehir Liv Hospital, Istanbul 34510, Turkey
| | - Tahsin Özatlı
- Department of Medical Oncology, Istinye University Medical School, Bahcesehir Liv Hospital, Istanbul 34510, Turkey
| |
Collapse
|
30
|
Thevandiran D, Nga V, Chang KTE, Ng LP, Seow WT, Low DCY, Yeo TT, Low SYY. Paediatric meningiomas in Singapore - Case series of a rare entity. J Clin Neurosci 2020; 73:62-66. [PMID: 32067824 DOI: 10.1016/j.jocn.2020.01.044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/05/2020] [Indexed: 01/19/2023]
Abstract
Paediatric meningiomas are extremely rare. These tumours constitute only 2 to 3% of all childhood brain tumours. Despite similarities in histological features between PMs and their adult counterparts, there are important distinctions between them. In this case series, the authors describe their experience in paediatric meningiomas in Singapore's 2 children's hospitals from 1998 to 2018. The primary aim of this retrospective study is to evaluate the clinical, radiological and pathological characteristics, and associated outcomes of paediatric patients diagnosed with meningioma managed in our local institutions. Following that, the study's findings are secondary aims are corroborated with published literature. A total of 10 patients (4 males and 6 females) were identified for this study within the period of 01 January 1998 to 31 December 2018. Their ages ranged from 1 year old to 18 years old (median age 10.5 years old). Two of the patients had NF1 and NF2 respectively. There were 9 intracranial and 1 intraspinal paediatric meningiomas. Seven patients achieved gross total resection and 3 patients had subtotal resection. Eight patients did not have tumour recurrence or increase in size of tumour remnant during the course of their follow-up. In congruency with the literature, up to 40% of our patients had higher grade meningiomas and 55.6% had large tumour volumes more than 30 cm3. Owing to the paucity of knowledge for this unusual tumour, the authors emphasize the need for closer surveillance and in-depth genomic studies to identify novel therapies for this challenging condition.
Collapse
Affiliation(s)
- Dave Thevandiran
- Neurosurgical Service, KK Women's and Children's Hospital, Singapore
| | - Vincent Nga
- Division of Neurosurgery, Department of Surgery, National University Hospital, Singapore
| | - Kenneth T E Chang
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore
| | - Lee Ping Ng
- Neurosurgical Service, KK Women's and Children's Hospital, Singapore
| | - Wan Tew Seow
- Neurosurgical Service, KK Women's and Children's Hospital, Singapore; Department of Neurosurgery, National Neuroscience Institute, Singapore; SingHealth Duke-NUS Neuroscience Academic Clinical Program, Singapore
| | - David C Y Low
- Neurosurgical Service, KK Women's and Children's Hospital, Singapore; Department of Neurosurgery, National Neuroscience Institute, Singapore; SingHealth Duke-NUS Neuroscience Academic Clinical Program, Singapore
| | - Tseng Tsai Yeo
- Division of Neurosurgery, Department of Surgery, National University Hospital, Singapore
| | - Sharon Y Y Low
- Neurosurgical Service, KK Women's and Children's Hospital, Singapore; Department of Neurosurgery, National Neuroscience Institute, Singapore; SingHealth Duke-NUS Neuroscience Academic Clinical Program, Singapore.
| |
Collapse
|
31
|
Ellingson BM, Abrey LE, Garcia J, Chinot O, Wick W, Saran F, Nishikawa R, Henriksson R, Mason WP, Harris RJ, Leu K, Woodworth DC, Mehta A, Raymond C, Chakhoyan A, Pope WB, Cloughesy TF. Post-chemoradiation volumetric response predicts survival in newly diagnosed glioblastoma treated with radiation, temozolomide, and bevacizumab or placebo. Neuro Oncol 2019; 20:1525-1535. [PMID: 29897562 DOI: 10.1093/neuonc/noy064] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background In the current study we used contrast-enhanced T1 subtraction maps to test whether early changes in enhancing tumor volume are prognostic for overall survival (OS) in newly diagnosed glioblastoma (GBM) patients treated with chemoradiation with or without bevacizumab (BV). Methods Seven hundred ninety-eight patients (404 BV and 394 placebo) with newly diagnosed GBM in the AVAglio trial (NCT00943826) had baseline MRI scans available, while 337 BV-treated and 269 placebo-treated patients had >4 MRI scans for response evaluation. The volume of contrast-enhancing tumor was quantified and used for subsequent analyses. Results A decrease in tumor volume during chemoradiation was associated with a longer OS in the placebo group (hazard ratio [HR] = 1.578, P < 0.0001) but not BV-treated group (HR = 1.135, P = 0.4889). Results showed a higher OS in patients on the placebo arm with a sustained decrease in tumor volume using a post-chemoradiation baseline (HR = 1.692, P = 0.0005), and a trend toward longer OS was seen in BV-treated patients (HR = 1.264, P = 0.0724). Multivariable Cox regression confirmed that sustained response or stable disease was prognostic for OS (HR = 0.7509, P = 0.0127) when accounting for age (P = 0.0002), KPS (P = 0.1516), postsurgical tumor volume (P < 0.0001), O6-methylguanine-DNA methyltransferase status (P < 0.0001), and treatment type (P = 0.7637) using the post-chemoradiation baseline. Conclusions The post-chemoradiation timepoint is a better baseline for evaluating efficacy in newly diagnosed GBM. Early progression during the maintenance phase is consequential in predicting OS, supporting the use of progression-free survival rates as a meaningful surrogate for GBM.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Physics and Biology in Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, California, USA.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,UCLA Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | | | - Olivier Chinot
- Aix-Marseille University, AP-HM, Service de Neuro-Oncologie, CHU Timone, Marseille, France
| | - Wolfgang Wick
- Clinical Cooperation Unit Neuro-oncology, German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Frank Saran
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | - Roger Henriksson
- Regional Cancer Center Stockholm, Stockholm, Sweden and Umeå University, Umeå, Sweden
| | | | - Robert J Harris
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Physics and Biology in Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,MedQIA, LLC, Los Angeles, California, USA
| | - Kevin Leu
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, California, USA
| | - Davis C Woodworth
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Physics and Biology in Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Arnav Mehta
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Ararat Chakhoyan
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Whitney B Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Timothy F Cloughesy
- UCLA Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
32
|
Ellingson BM, Aftab DT, Schwab GM, Hessel C, Harris RJ, Woodworth DC, Leu K, Chakhoyan A, Raymond C, Drappatz J, de Groot J, Prados MD, Reardon DA, Schiff D, Chamberlain M, Mikkelsen T, Desjardins A, Holland J, Ping J, Weitzman R, Wen PY, Cloughesy TF. Volumetric response quantified using T1 subtraction predicts long-term survival benefit from cabozantinib monotherapy in recurrent glioblastoma. Neuro Oncol 2019; 20:1411-1418. [PMID: 29660005 DOI: 10.1093/neuonc/noy054] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background To overcome challenges with traditional response assessment in anti-angiogenic agents, the current study uses T1 subtraction maps to quantify volumetric radiographic response in monotherapy with cabozantinib, an orally bioavailable tyrosine kinase inhibitor with activity against vascular endothelial growth factor receptor 2 (VEGFR2), hepatocyte growth factor receptor (MET), and AXL, in an open-label, phase II trial in patients with recurrent glioblastoma (GBM) (NCT00704288). Methods A total of 108 patients with adequate imaging data and confirmed recurrent GBM were included in this retrospective study from a phase II multicenter trial of cabozantinib monotherapy (XL184-201) at either 100 mg (N = 87) or 140 mg (N = 21) per day. Contrast enhanced T1-weighted digital subtraction maps were used to define volume of contrast-enhancing tumor at baseline and subsequent follow-up time points. Volumetric radiographic response (>65% reduction in contrast-enhancing tumor volume from pretreatment baseline tumor volume sustained for more than 4 wk) was tested as an independent predictor of overall survival (OS). Results Volumetric response rate for all therapeutic doses was 38.9% (41.4% and 28.6% for 100 mg and 140 mg doses, respectively). A log-linear association between baseline tumor volume and OS (P = 0.0006) and a linear correlation between initial change in tumor volume and OS (P = 0.0256) were observed. A significant difference in OS was observed between responders (median OS = 20.6 mo) and nonresponders (median OS = 8.0 mo) (hazard ratio [HR] = 0.3050, P < 0.0001). Multivariable analyses showed that continuous measures of baseline tumor volume (HR = 1.0233, P < 0.0001) and volumetric response (HR = 0.2240, P < 0.0001) were independent predictors of OS. Conclusions T1 subtraction maps provide value in determining response in recurrent GBM treated with cabozantinib and correlated with survival benefit.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | | | | | | | - Robert J Harris
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Davis C Woodworth
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Kevin Leu
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Ararat Chakhoyan
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jan Drappatz
- Department of Neurology and Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - John de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael D Prados
- Department of Neurosurgery, University of California San Francisco (UCSF), San Francisco, California
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David Schiff
- Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia
| | - Marc Chamberlain
- Department of Neurology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Annick Desjardins
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | | | - Jerry Ping
- Exelixis, South San Francisco, California
| | | | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
33
|
Brain Tumor Surgery is Safe in Octogenarians and Nonagenarians: A Single-Surgeon 741 Patient Series. World Neurosurg 2019; 132:e185-e192. [PMID: 31505286 DOI: 10.1016/j.wneu.2019.08.219] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVES Elderly patients with surgically accessible brain tumors are often not offered clinically indicated brain tumor surgery (BTS) because of to assumptions of greater risk for perioperative morbidity and mortality. Because brain tumor incidence is highest in the geriatric population, and because the global population is aging, accurate understanding of BTS risk in elderly patients is critical. We aimed to compare safety of BTS in elderly patients with younger counterparts to better understand the risk-benefit profile of BTS for elderly patients. METHODS Retrospective cohort study of young (20-29 years), senior (60-79 years), and elderly (80+ years) patients who underwent BTS with a single neurosurgeon. Differences between pre- and postoperative modified Rankin score (ΔmRS), length of hospitalization (LOH), complication rate, and 30-day readmission rates (30DRR) were recorded. RESULTS A total of 741 patients (83 elderly, 570 senior, and 88 young) were identified. No significant difference in preoperative mRS between different age groups, χ2 = 0.269, P = 0.874. Elderly complication rate was 6.0%, not significantly different from young (4.5%, P = 0.667) or senior (7.2%, P = 0.696) complication rate. Elderly LOH was 1.93 ± SD 0.176 days; not significantly different from young (3.01 ± 0.384 days, P = 0.081) or senior (2.47 ± 0.144 days, P = 0.881). Statistical equivalence testing showed with 95% confidence that there was equivalence in ΔmRS among age groups. CONCLUSIONS Elderly patients did not have significantly different ΔmRS, LOH, 30DRR, or complication rates after BTS compared with younger counterparts. Therefore, in healthy patients, advanced age alone should not prevent patients from being offered BTS.
Collapse
|
34
|
Imber BS, Lin AL, Zhang Z, Keshavamurthy KN, Deipolyi AR, Beal K, Cohen MA, Tabar V, DeAngelis LM, Geer EB, Yang TJ, Young RJ. Comparison of Radiographic Approaches to Assess Treatment Response in Pituitary Adenomas: Is RECIST or RANO Good Enough? J Endocr Soc 2019; 3:1693-1706. [PMID: 31528829 PMCID: PMC6735764 DOI: 10.1210/js.2019-00130] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/25/2019] [Indexed: 12/29/2022] Open
Abstract
Context Pituitary adenomas (PA) are often irregularly shaped, particularly posttreatment. There are no standardized radiographic criteria for assessing treatment response, substantially complicating interpretation of prospective outcome data. Existing imaging frameworks for intracranial tumors assume perfectly spherical targets and may be suboptimal. Objective To compare a three-dimensional (3D) volumetric approach against accepted surrogate measurements to assess PA posttreatment response (PTR). Design Retrospective review of patients with available pre- and postradiotherapy (RT) imaging. A neuroradiologist determined tumor sizes in one dimensional (1D) per Response Evaluation in Solid Tumors (RECIST) criteria, two dimensional (2D) per Response Assessment in Neuro-Oncology (RANO) criteria, and 3D estimates assuming a perfect sphere or perfect ellipsoid. Each tumor was manually segmented for 3D volumetric measurements. The Hakon Wadell method was used to calculate sphericity. Setting Tertiary cancer center. Patients or Other Participants Patients (n = 34, median age = 50 years; 50% male) with PA and MRI scans before and after sellar RT. Interventions Patients received sellar RT for intact or surgically resected lesions. Main Outcome Measures Radiographic PTR, defined as percent tumor size change. Results Using 3D volumetrics, mean sphericity = 0.63 pre-RT and 0.60 post-RT. With all approaches, most patients had stable disease on post-RT scan. PTR for 1D, 2D, and 3D spherical measurements were moderately well correlated with 3D volumetrics (e.g., for 1D: 0.66, P < 0.0001) and were superior to 3D ellipsoid. Intraclass correlation coefficient demonstrated moderate to good reliability for 1D, 2D, and 3D sphere (P < 0.001); 3D ellipsoid was inferior (P = 0.009). 3D volumetrics identified more potential partially responding and progressive lesions. Conclusions Although PAs are irregularly shaped, 1D and 2D approaches are adequately correlated with volumetric assessment.
Collapse
Affiliation(s)
- Brandon S Imber
- Department of Radiation Oncology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Andrew L Lin
- Department of Neurology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Zhigang Zhang
- Department of Epidemiology & Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Krishna Nand Keshavamurthy
- Department of Radiology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Amy Robin Deipolyi
- Department of Radiology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Kathryn Beal
- Department of Radiation Oncology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Marc A Cohen
- Department of Surgery, Head & Neck Service, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Viviane Tabar
- Department of Neurosurgery, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Lisa M DeAngelis
- Department of Neurology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Eliza B Geer
- Department of Endocrinology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - T Jonathan Yang
- Department of Radiation Oncology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Robert J Young
- Department of Radiology, Multidisciplinary Skull Base and Pituitary Center at Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
35
|
Institutional Implementation of a Structured Reporting System: Our Experience with the Brain Tumor Reporting and Data System. Acad Radiol 2019; 26:974-980. [PMID: 30661977 DOI: 10.1016/j.acra.2018.12.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 11/24/2022]
Abstract
RATIONALE AND OBJECTIVES Analyze the impact of implementing a structured reporting system for primary brain tumors, the Brain Tumor Reporting and Data System, on attitudes toward radiology reports at a single institution. MATERIALS AND METHODS Following Institutional Review Board approval, an initial 22 question, 5 point (1-worst to 5-best), survey was sent to faculty members, house staff members, and nonphysician providers at our institution who participate in the direct care of brain tumor patients. Results were used to develop a structured reporting strategy for brain tumors which was implemented across an entire neuroradiology section in a staged approach. Nine months following structured reporting implementation, a follow-up 27 question survey was sent to the same group of providers. Keyword search of radiology reports was used to assess usage of Brain Tumor Reporting and Data System over time. RESULTS Fifty-three brain tumor care providers responded to the initial survey and 38 to the follow-up survey. After implementing BT-RADS, respondents reported improved attitudes across multiple areas including: report consistency (4.3 vs. 3.4; p < 0.001), report ambiguity (4.2 vs. 3.2, p < 0.001), radiologist/physician communication (4.5 vs. 3.8; p < 0.001), facilitation of patient management (4.2 vs. 3.6; p = 0.003), and confidence in reports (4.3 vs. 3.5; p < 0.001). Providers were more satisfied with the BT-RADS structured reporting system (4.3 vs. 3.7; p = 0.04). Use of the reporting template progressively increased with 81% of brain tumor reports dictated using the new template by 9 months. CONCLUSION Implementing a structured template for brain tumor imaging improves perception of radiology reports among radiologists and referring providers involved in the care of brain tumor patients.
Collapse
|
36
|
Krivoshapkin AL, Sergeev GS, Gaytan AS, Kalneus LE, Kurbatov VP, Abdullaev OA, Salim N, Bulanov DV, Simonovich AE. Automated Volumetric Analysis of Postoperative Magnetic Resonance Imaging Predicts Survival in Patients with Glioblastoma. World Neurosurg 2019; 126:e1510-e1517. [PMID: 30910753 DOI: 10.1016/j.wneu.2019.03.142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Glioblastomas (GBMs) are primary brain tumors that are very difficult to treat. Magnetic resonance imaging (MRI) is the reference tool for diagnosis, postoperative control, and follow-up of GBM. The MRI tumor contrast enhancement part serves as a target for surgery. However, there are controversial data about the influence of pre- and postoperative tumor volumetric MRI parameters on overall survival (OS). METHODS Data of 57 patients with GBM were analyzed retrospectively. All patients had maximum safe resection and standard adjuvant treatment. All patients underwent 1.5-T MRI with contrast in the first 24 hours postoperatively. The data of pre- and postoperative volumetric parameters were analyzed using the original software. RESULTS Correlation analysis between the postoperative volume of the tumor contrast enhancement part and the patient's OS revealed a significant level (on the Chaddock scale) of inverse correlation. Residual tumor volume associated with OS of >6 months was determined as <2.5 cm3. The mortality risk in the first 6 months after tumor resection is 3.4 times higher when the tumor remnant is >2.5 cm3 (risk ratio, 3.4; P = 0.0002). CONCLUSIONS The volume of MRI contrast-enhancing GBM remnants after surgery, automatically measured by the software, was a significant predictor for early postoperative progression and death.
Collapse
Affiliation(s)
- Alexey L Krivoshapkin
- Department of Neurosurgery, Novosibirsk State Medical University, Novosibirsk, Russia.
| | - Gleb S Sergeev
- Neurosurgical Department, European Medical Center, Moscow, Russia
| | - Alekey S Gaytan
- Neurosurgical Department, European Medical Center, Moscow, Russia
| | - Leonid E Kalneus
- Physics Department, Novosibirsk State University, Novosibirsk, Russia
| | | | - Orkhan A Abdullaev
- Department of Neurosurgery, Novosibirsk State Medical University, Novosibirsk, Russia
| | - Nidal Salim
- Radiotherapy Center, European Medical Center, Moscow, Russia
| | | | | |
Collapse
|
37
|
Kim H, Goo JM, Kim YT, Park CM. Clinical T Category of Non–Small Cell Lung Cancers: Prognostic Performance of Unidimensional versus Bidimensional Measurements at CT. Radiology 2019; 290:807-813. [DOI: 10.1148/radiol.2019182068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Hyungjin Kim
- From the Department of Radiology (H.K., J.M.G., C.M.P.) and Department of Thoracic and Cardiovascular Surgery (Y.T.K.), Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Korea (J.M.G., C.M.P.); and Cancer Research Institute, Seoul National University, Seoul, Korea (J.M.G., Y.T.K., C.M.P.)
| | - Jin Mo Goo
- From the Department of Radiology (H.K., J.M.G., C.M.P.) and Department of Thoracic and Cardiovascular Surgery (Y.T.K.), Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Korea (J.M.G., C.M.P.); and Cancer Research Institute, Seoul National University, Seoul, Korea (J.M.G., Y.T.K., C.M.P.)
| | - Young Tae Kim
- From the Department of Radiology (H.K., J.M.G., C.M.P.) and Department of Thoracic and Cardiovascular Surgery (Y.T.K.), Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Korea (J.M.G., C.M.P.); and Cancer Research Institute, Seoul National University, Seoul, Korea (J.M.G., Y.T.K., C.M.P.)
| | - Chang Min Park
- From the Department of Radiology (H.K., J.M.G., C.M.P.) and Department of Thoracic and Cardiovascular Surgery (Y.T.K.), Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Korea (J.M.G., C.M.P.); and Cancer Research Institute, Seoul National University, Seoul, Korea (J.M.G., Y.T.K., C.M.P.)
| |
Collapse
|
38
|
Borcoman E, Nandikolla A, Long G, Goel S, Le Tourneau C. Patterns of Response and Progression to Immunotherapy. Am Soc Clin Oncol Educ Book 2018; 38:169-178. [PMID: 30231380 DOI: 10.1200/edbk_200643] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Patterns of response and progression to immunotherapy may differ from those observed with drugs such as chemotherapy and molecularly targeted agents. Specifically, some patients experience a response after progression that is retrospectively named pseudoprogression. This phenomenon of pseudoprogression, first reported in patients with melanoma who were treated with ipilimumab, has led to the development of immune-specific related response criteria, such as irRC (immune-related response criteria), irRECIST (immune-related RECIST), and iRECIST (immunotherapy RECIST) that allow continued treatment beyond progression. However, the rate of pseudoprogression has never exceeded 10% of patients across tumor types. Conversely, rapid progressions after immunotherapy, called hyperprogressions, were reported by three different teams in 9% to 29% of patients treated with immunotherapy. Because of the absence of control arms in these studies, it remains to be determined whether these rapid progressions reflect a detrimental effect of immunotherapy in these patients. Finally, preliminary data suggest that immunotherapy might also affect response to subsequent standard therapies. In total, given the rarity of pseudoprogressions across tumor types and the recent description of hyperprogressions, classic RECIST remains a reasonable and rational method to assess response to immunotherapy. Continuation of treatment beyond progression should be proposed only in carefully selected patients whose clinical conditions have improved and who have not experienced severe toxicities. Although there is an urgent need to identify predictive biomarkers of efficacy to immunotherapy, there is an equally urgent need to identify predictive factors of progression or possibly hyperprogression.
Collapse
Affiliation(s)
- Edith Borcoman
- From the Department of Drug Development and Innovation, Institut Curie, Paris and Saint-Cloud, France; Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Melanoma Institute Australia, North Sydney, NSW, Australia; INSERM U900 Research Unit, Saint-Cloud, France; Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Amara Nandikolla
- From the Department of Drug Development and Innovation, Institut Curie, Paris and Saint-Cloud, France; Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Melanoma Institute Australia, North Sydney, NSW, Australia; INSERM U900 Research Unit, Saint-Cloud, France; Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Georgina Long
- From the Department of Drug Development and Innovation, Institut Curie, Paris and Saint-Cloud, France; Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Melanoma Institute Australia, North Sydney, NSW, Australia; INSERM U900 Research Unit, Saint-Cloud, France; Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Sanjay Goel
- From the Department of Drug Development and Innovation, Institut Curie, Paris and Saint-Cloud, France; Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Melanoma Institute Australia, North Sydney, NSW, Australia; INSERM U900 Research Unit, Saint-Cloud, France; Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Christophe Le Tourneau
- From the Department of Drug Development and Innovation, Institut Curie, Paris and Saint-Cloud, France; Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Melanoma Institute Australia, North Sydney, NSW, Australia; INSERM U900 Research Unit, Saint-Cloud, France; Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| |
Collapse
|
39
|
Wang HK, Xu WH, Ma CG, Zhou LP, Shi GH, Zhang HL, Ye DW. Tumor growth velocity: A modified tumor growth rate defining tumor progression during sorafenib treatment in patients with metastatic renal cell carcinoma. Int J Urol 2018; 26:75-82. [PMID: 30325072 DOI: 10.1111/iju.13807] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/22/2018] [Indexed: 01/16/2023]
Abstract
OBJECTIVES To investigate the role of tumor growth velocity in defining tumor progression in metastatic renal cell carcinoma patients treated with the vascular endothelial growth factor tyrosine kinase inhibitor, sorafenib. METHODS A modified calculation for tumor growth velocity was introduced to evaluate the tumor growth velocity, before and after sorafenib withdrawal. Known prognostic factors together with tumor growth velocity before drug withdrawal and tumor growth velocity after drug withdrawal were compared using a χ2 -test from a contingency table, and partial likelihood test from a Cox regression model for overall survival. RESULTS A total of 114 patients who reached progressive disease and withdrew from sorafenib were enrolled after a median follow-up period of 107.8 months. Tumor growth velocity before drug withdrawal was 7.347 ± 4.040, and tumor growth velocity after drug withdrawal was 11.647 ± 5.937 (P < 0.001). Higher tumor growth velocity before drug withdrawal was correlated with a higher risk Memorial Sloan Kettering Cancer Center score (P = 0.022), Karnofsky Performance Status <80 (P = 0.028), non-clear cell carcinoma (P = 0.037), higher tumor nucleus grade (P < 0.001) and best treatment response (P < 0.001). Patients with tumor growth velocity before drug withdrawal >5.0 had shorter overall survival (P < 0.001). On multivariate analysis, factors associated with overall survival were high/intermediate Memorial Sloan Kettering Cancer Center risk score (hazard ratio 2.119, P = 0.006), non-clear histological subtype (hazard ratio 1.900, P = 0.031), tumor growth velocity before drug withdrawal ≥5.0 (hazard ratio 2.758, P < 0.001) and progressive disease as best response (hazard ratio 2.069, P = 0.001). CONCLUSIONS Significantly faster tumor growth can be observed if sorafenib is discontinued in the case of disease progression. Thus, we suggest not to withdraw targeted agents until tumor growth velocity is >5.0.
Collapse
Affiliation(s)
- Hong-Kai Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Hao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chun-Guang Ma
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang-Ping Zhou
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guo-Hai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hai-Liang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding-Wei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Smedley NF, Ellingson BM, Cloughesy TF, Hsu W. Longitudinal Patterns in Clinical and Imaging Measurements Predict Residual Survival in Glioblastoma Patients. Sci Rep 2018; 8:14429. [PMID: 30258190 PMCID: PMC6158293 DOI: 10.1038/s41598-018-32397-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 07/09/2018] [Indexed: 11/13/2022] Open
Abstract
The growing amount of longitudinal data for a large population of patients has necessitated the application of algorithms that can discover patterns to inform patient management. This study demonstrates how temporal patterns generated from a combination of clinical and imaging measurements improve residual survival prediction in glioblastoma patients. Temporal patterns were identified with sequential pattern mining using data from 304 patients. Along with patient covariates, the patterns were incorporated as features in logistic regression models to predict 2-, 6-, or 9-month residual survival at each visit. The modeling approach that included temporal patterns achieved test performances of 0.820, 0.785, and 0.783 area under the receiver operating characteristic curve for predicting 2-, 6-, and 9-month residual survival, respectively. This approach significantly outperformed models that used tumor volume alone (p < 0.001) or tumor volume combined with patient covariates (p < 0.001) in training. Temporal patterns involving an increase in tumor volume above 122 mm3/day, a decrease in KPS across multiple visits, moderate neurologic symptoms, and worsening overall neurologic function suggested lower residual survival. These patterns are readily interpretable and found to be consistent with known prognostic indicators, suggesting they can provide early indicators to clinicians of changes in patient state and inform management decisions.
Collapse
Affiliation(s)
- Nova F Smedley
- Department of Bioengineering, Henry Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, USA.,Medical Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Benjamin M Ellingson
- Department of Bioengineering, Henry Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, USA.,UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - William Hsu
- Department of Bioengineering, Henry Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, USA. .,Medical Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW To review the growth kinetics of small renal masses and available imaging modalities for mass characterization and surveillance, highlight current organizational recommendations for the active surveillance of small renal masses, and discuss the most recently reported oncological outcomes of patients as they relate to various surveillance imaging protocols and progression to delayed intervention. RECENT FINDINGS Overall, organizational guideline recommendations are broad and lack specifics regarding timing and modality for follow-up imaging of small renal masses. Additionally, despite general consensus in the literature about certain criteria to trigger delayed intervention, there exist no formal guidelines. Active surveillance of small renal masses is an acceptable management strategy for patients with prohibitive surgical risk; however, standardized imaging protocols for surveillance are lacking, as are randomized, prospective trials to evaluate the ideal follow-up protocol.
Collapse
|
42
|
Ma SX, Zhou T, Huang Y, Yang YP, Zhan JH, Zhang YX, Zhang ZH, Zhao YY, Fang WF, Ma YX, Chen LK, Zhao HY, Zhang L. The efficacy of first-line chemotherapy in recurrent or metastatic nasopharyngeal carcinoma: a systematic review and meta-analysis. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:201. [PMID: 30023364 DOI: 10.21037/atm.2018.05.14] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background The standard first-line chemotherapy for patients with recurrent or metastatic nasopharyngeal carcinoma (R/M NPC) has not been well established. We conducted a pooled meta-analysis to evaluate the efficacy of commonly used first-line chemotherapy in this disease. Methods Electronic databases including PubMed, Embase, and Corchrane library were searched for eligible literatures. Objective response rate (ORR), disease control rate (DCR), progression free survival (PFS), and overall survival (OS) were pooled with the 95% confidence interval (CI) using R software. Results Totally 973 patients were available for analysis from 14 phase II single arm clinical trials and 2 phase III randomized clinical trials. Four regimens were identified including 5-fluorouracil plus platinum (FP), gemcitabine plus platinum (GP), taxanes plus platinum (TP), and triplet combination regimen. Of these four regimens, triplet combination regimen demonstrated best short-term efficacy with a highest ORR (0.74; 95% CI, 0.62-0.87), DCR (0.91; 95% CI, 0.87-0.95), and 6-month PFS rate (0.83; 95% CI, 0.75-0.91), while 1-year OS rate (0.74; 95% CI, 0.61-0.87) was a little lower than TP regimen. Meanwhile, TP regimen showed best prognosis with a highest 1-year OS rate of 0.79 (95% CI, 0.65-0.92) and pretty good short-term efficacy with an ORR of 0.60 (95% CI, 0.48-0.72) and a DCR of 0.92 (95% CI, 0.86-0.98) comparable with triplet combination therapy. FP regimen had the lowest ORR (0.52; 95% CI, 0.38-0.65) and 1-year OS rate (0.63; 95% CI, 0.57-0.69). Efficacy of GP regimen fell between FP and TP regimens with an ORR of 0.54 (95% CI, 0.38-0.65), a DCR of 0.85 (95% CI, 0.71-0.93), a 6-month PFS rate of 0.69 (95% CI, 0.60-0.78) and a 1-year OS rate of 0.71 (95% CI, 0.61-0.80). Conclusions Among four commonly used first-line chemotherapy regimens for R/M NPC, triplet combination regimen showed best short-term efficacy but failed to improve prognosis. TP regimen demonstrated fairly good short-term efficacy and best long-term efficacy, followed by GP regimen, while FP regimen was the lowest.
Collapse
Affiliation(s)
- Shu-Xiang Ma
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Ting Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Yun-Peng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Jian-Hua Zhan
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Ya-Xiong Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Zhong-Han Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Yuan-Yuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Wen-Feng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Yu-Xiang Ma
- Department of Cancer Research, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Li-Kun Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Hong-Yun Zhao
- Department of Cancer Research, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
43
|
Piombo V, Jochmann K, Hoffmann D, Wuelling M, Vortkamp A. Signaling systems affecting the severity of multiple osteochondromas. Bone 2018; 111:71-81. [PMID: 29545125 DOI: 10.1016/j.bone.2018.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/12/2018] [Accepted: 03/09/2018] [Indexed: 01/01/2023]
Abstract
Multiple osteochondromas (MO) syndrome is a dominant autosomal bone disorder characterized by the formation of cartilage-capped bony outgrowths that develop at the juxtaposition of the growth plate of endochondral bones. MO has been linked to mutations in either EXT1 or EXT2, two glycosyltransferases required for the synthesis of heparan sulfate (HS). The establishment of mouse mutants demonstrated that a clonal, homozygous loss of Ext1 in a wild type background leads to the development of osteochondromas. Here we investigate mechanisms that might contribute to the variation in the severity of the disease observed in human patients. Our results show that residual amounts of HS are sufficient to prevent the development of osteochondromas strongly supporting that loss of heterozygosity is required for osteochondroma formation. Furthermore, we demonstrate that different signaling pathways affect size and frequency of the osteochondromas thereby modulating the severity of the disease. Reduced Fgfr3 signaling, which regulates proliferation and differentiation of chondrocytes, increases osteochondroma number, while activated Fgfr3 signaling reduces osteochondroma size. Both, activation and reduction of Wnt/β-catenin signaling decrease osteochondroma size and frequency by interfering with the chondrogenic fate of the mutant cells. Reduced Ihh signaling does not change the development of the osteochondromas, while elevated Ihh signaling increases the cellularity and inhibits chondrocyte differentiation in a subset of osteochondromas and might thus predispose osteochondromas to the transformation into chondrosarcomas.
Collapse
Affiliation(s)
- Virginia Piombo
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Katja Jochmann
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Daniel Hoffmann
- Research Group Bioinformatics, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Manuela Wuelling
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Andrea Vortkamp
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
44
|
Unidimensional measurement may be superior to assess primary tumor response after neoadjuvant chemotherapy for nasopharyngeal carcinoma. Oncotarget 2018; 8:46937-46945. [PMID: 28159937 PMCID: PMC5564534 DOI: 10.18632/oncotarget.14941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 12/28/2016] [Indexed: 12/02/2022] Open
Abstract
Application of current response evaluation criteria in solid tumors (RECIST 1.1) for assessment of irregularly shaped nasopharyngeal carcinoma (NPC) is a gray area with much ambiguity. Our aim was to compare unidimensional measurements (UDM) and bidimensional measurements (BDM) on magnetic resonance images in alternative planes for measurement of tumor response after neoadjuvant chemotherapy (NACT) in patients with locally advanced NPC. 59 patients with untreated non-metastatic NPC were prospectively enrolled. The size or change in size of the primary tumor and retropharyngeal nodes was assessed by UDM and BDM on axial and coronal planes before and after 2 cycles of NACT. Tumor volume was considered as the reference standard. Correlation between volume and diameter was analyzed using a general linear model. The degree of agreement and discordance of response classification based on different measures were evaluated with κ statistic and McNemar's test, respectively. Both axial UDM (RECIST 1.1) and axial BDM (WHO) showed a significant association with volumetric standard. However, the agreement of axial UDM with VM was better than that of axial BDM (κ value: 0.514 to 0.372). In addition, when increasing coronal planes to evaluate tumor response with UDM and BDM, an inferior agreement between coronal BDM and VM was still observed. Notably, coronal UDM showed the best consistency with volume (κ = 0.585). Hence, axial UDM showed better correlation with volumetric measurements than axial BDM. Since coronal UDM showed high correlation to VM, we suggest further research to assess its use for response assessment of NPC after NACT.
Collapse
|
45
|
Pusceddu S, Prinzi N, Raimondi A, Corti F, Buzzoni R, Di Bartolomeo M, Seregni E, Maccauro M, Coppa J, Milione M, Mazzaferro V, de Braud F. Entering the third decade of experience with octreotide LAR in neuroendocrine tumors: A review of current knowledge. TUMORI JOURNAL 2018; 105:113-120. [DOI: 10.1177/0300891618765362] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gastroenteropancreatic neuroendocrine tumors (NETs) are a relatively rare group of heterogeneous neoplasms. The most significant advance in therapy of NETs has been the advent of the somatostatin analog octreotide, which represents a cornerstone in their management and dramatically changed the therapeutic landscape. Octreotide long-acting release (LAR) was developed to overcome some of the limitations of octreotide. Several clinical studies, including PROMID and RADIANT-2, have validated the clinical benefits of octreotide LAR in NETs, with tumor shrinkage in about 10% of patients and tumor stabilization in roughly half of cases. While the use of octreotide LAR is well-consolidated in NETs, some open questions remain. These include the use of high-dose octreotide LAR, as there is evidence that higher dose may provide longer disease control, and nonstandard treatment schedules, with administration every 21 days instead of 28 days, as well as their use in combination with targeted agents or peptide receptor radiotherapy in clinical practice. After 3 decades of clinical experience with octreotide LAR, the drug has a well-established safety profile. It is well-tolerated and treatment discontinuations due to adverse events are uncommon. One exception is cholelithiasis, which may increase with longer duration of treatment. According to the literature data, octreotide LAR is currently recommended in both functioning and nonfunctioning advanced NETs. This review summarizes the available clinical data with octreotide LAR and also provides future perspectives on its possible uses in patients with NETs.
Collapse
Affiliation(s)
- Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Alessandra Raimondi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Roberto Buzzoni
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Ettore Seregni
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Marco Maccauro
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Jorgelina Coppa
- Department of Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Massimo Milione
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Vincenzo Mazzaferro
- Department of Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
- University of Milan, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
- University of Milan, Milan, Italy
| |
Collapse
|
46
|
Terranova N, Girard P, Ioannou K, Klinkhardt U, Munafo A. Assessing Similarity Among Individual Tumor Size Lesion Dynamics: The CICIL Methodology. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2018; 7:228-236. [PMID: 29388396 PMCID: PMC5915614 DOI: 10.1002/psp4.12284] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/28/2017] [Accepted: 01/17/2018] [Indexed: 02/06/2023]
Abstract
Mathematical models of tumor dynamics generally omit information on individual target lesions (iTLs), and consider the most important variable to be the sum of tumor sizes (TS). However, differences in lesion dynamics might be predictive of tumor progression. To exploit this information, we have developed a novel and flexible approach for the non‐parametric analysis of iTLs, which integrates knowledge from signal processing and machine learning. We called this new methodology ClassIfication Clustering of Individual Lesions (CICIL). We used CICIL to assess similarities among the TS dynamics of 3,223 iTLs measured in 1,056 patients with metastatic colorectal cancer treated with cetuximab combined with irinotecan, in two phase II studies. We mainly observed similar dynamics among lesions within the same tumor site classification. In contrast, lesions in anatomic locations with different features showed different dynamics in about 35% of patients. The CICIL methodology has also been implemented in a user‐friendly and efficient Java‐based framework.
Collapse
Affiliation(s)
- Nadia Terranova
- Merck Institute for Pharmacometrics, Merck Serono S.A., Switzerland, a Subsidiary of Merck KGaA, Darmstadt, Germany
| | - Pascal Girard
- Merck Institute for Pharmacometrics, Merck Serono S.A., Switzerland, a Subsidiary of Merck KGaA, Darmstadt, Germany
| | - Konstantinos Ioannou
- Merck Institute for Pharmacometrics, Merck Serono S.A., Switzerland, a Subsidiary of Merck KGaA, Darmstadt, Germany
| | | | - Alain Munafo
- Merck Institute for Pharmacometrics, Merck Serono S.A., Switzerland, a Subsidiary of Merck KGaA, Darmstadt, Germany
| |
Collapse
|
47
|
Somarouthu B, Lee SI, Urban T, Sadow CA, Harris GJ, Kambadakone A. Immune-related tumour response assessment criteria: a comprehensive review. Br J Radiol 2018; 91:20170457. [PMID: 29172675 DOI: 10.1259/bjr.20170457] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Growing emphasis on precision medicine in oncology has led to increasing use of targeted therapies that encompass a spectrum of drug classes including angiogenesis inhibitors, immune modulators, signal transduction inhibitors, DNA damage modulators, hormonal agents etc. Immune therapeutic drugs constitute a unique group among the novel therapeutic agents that are transforming cancer treatment, and their use is rising. The imaging manifestations in patients on immune therapies appear to be distinct from those typically seen with conventional cytotoxic therapies. Patients on immune therapies may demonstrate a delayed response, transient tumour enlargement followed by shrinkage, stable size, or initial appearance of new lesions followed by stability or response. These newer patterns of response to treatment have rendered conventional criteria such as World Health Organization and response evaluation criteria in solid tumours suboptimal in monitoring changes in tumour burden. As a consequence, newer imaging response criteria such as immune-related response evaluation criteria in solid tumours and immune-related response criteria are being implemented in many trials to effectively monitor patients on immune therapies. In this review, we discuss the traditional and new imaging response criteria for evaluation of solid tumours, review the outcomes of various articles which compared traditional criteria with the new immune-related criteria and discuss pseudo-progression and immune-related adverse events.
Collapse
Affiliation(s)
| | - Susanna I Lee
- 2 Department of Radiology, Massachusetts General Hospital , Boston, MA , USA
| | - Trinity Urban
- 1 Tumor Imaging Metrics Core, Dana-Farber/Harvard Cancer Centre , Boston, MA , USA
| | - Cheryl A Sadow
- 3 Department of Radiology, Brigham and Women's Hospital , Boston, MA , USA
| | - Gordon J Harris
- 1 Tumor Imaging Metrics Core, Dana-Farber/Harvard Cancer Centre , Boston, MA , USA
| | - Avinash Kambadakone
- 2 Department of Radiology, Massachusetts General Hospital , Boston, MA , USA
| |
Collapse
|
48
|
Lissoni P, Rovelli F, Malugani F, Brivio F, Fumagalli L, Gardani GS. Changes in Circulating VEGF Levels in Relation to Clinical Response during Chemotherapy for Metastatic Cancer. Int J Biol Markers 2018; 18:152-5. [PMID: 12841685 DOI: 10.1177/172460080301800209] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abnormally high blood levels of vascular endothelial growth factor (VEGF) appear to be associated with a poor prognosis in advanced cancer, probably as a consequence of its angiogenic and immunosuppressive effects. The prognostic significance of changes in VEGF secretion during cancer chemotherapy is still unknown. This study aimed to investigate the relation between VEGF variations and therapeutic results during chemotherapy in advanced malignancies. The study included 90 metastatic cancer patients, 59 with non-small cell lung cancer and 31 with colorectal carcinoma. Chemotherapy consisted of cisplatin plus etoposide for NSCLC and camptothecin for colorectal cancer. Abnormally high (>2 SD with respect to values in healthy controls) pretreatment VEGF levels were found in 38/90 (42%) patients. The percentage of non-progressive disease in response to chemotherapy was significantly higher in patients with normal levels of VEGF prior to therapy than in those with elevated pretreatment values of VEGF (10/32 vs 4/27; p<0.05). Moreover, the percentage of VEGF level normalization during chemotherapy was significantly higher in patients with objective tumor response or stable disease than in progressing patients (10/18 vs 0/20; p<0.001). Finally, among patients with tumor response or disease stabilization, the one-year survival rate was significantly higher in patients with chemotherapy-induced normalization of VEGF than in those with persistently high VEGF blood levels (9/10 vs 3/8; p<0.05). These results suggest that changes in VEGF levels during chemotherapy may represent a useful biomarker to predict the effect of chemotherapy in terms of tumor response and survival in patients with metastatic solid neoplasms.
Collapse
Affiliation(s)
- P Lissoni
- Division of Radiation Oncology, University of Milan-Bicocca, San Gerardo Hospital, Monza, Italy
| | | | | | | | | | | |
Collapse
|
49
|
Jardim DL, de Melo Gagliato D, Giles FJ, Kurzrock R. Analysis of Drug Development Paradigms for Immune Checkpoint Inhibitors. Clin Cancer Res 2017; 24:1785-1794. [PMID: 29212781 DOI: 10.1158/1078-0432.ccr-17-1970] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/15/2017] [Accepted: 11/30/2017] [Indexed: 12/15/2022]
Abstract
Immune checkpoint inhibitors have unique toxicities and response kinetics compared with cytotoxic and gene-targeted anticancer agents. We investigated the impact of innovative/accelerated immunotherapy drug development/approval models on the accuracy of safety and efficacy assessments by searching the FDA website. Initial phase I trials for each agent were reviewed and safety and efficacy data compared with that found in later trials leading to regulatory approvals of the same agents. As of June 2017, the FDA approved six checkpoint inhibitors for a variety of cancer types. All checkpoint inhibitors received a priority review status and access to at least two additional FDA special access programs, more often breakthrough therapy designation and accelerated approval. Median clinical development time (investigational new drug application to approval) was 60.77 months [avelumab had the shortest timeline (52.33 months)]. Response rates during early phase I trials (median = 16%) are higher than for phase I trials of other agents (with the exception of gene-targeted agents tested with a biomarker). Doses approved were usually not identical to doses recommended on phase I trials. Approximately 50% of types of immune-related and 43% of types of clinically relevant toxicities from later trials were identified in early-phase trials. Even so, treatment-related mortality remains exceedingly low in later studies (0.33% of patients). In conclusion, efficacy and safety of immune checkpoint inhibitors appear to be reasonably predicted from the dose-finding portion of phase I trials, indicating that the fast-track development of these agents is safe and justified. Clin Cancer Res; 24(8); 1785-94. ©2017 AACR.
Collapse
Affiliation(s)
- Denis L Jardim
- Department of Clinical Oncology, Hospital Sirio Libanes, Sao Paulo, Brazil.
| | | | - Francis J Giles
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California, San Diego, California
| |
Collapse
|
50
|
Karlsson AK, Saleh SN. Checkpoint inhibitors for malignant melanoma: a systematic review and meta-analysis. Clin Cosmet Investig Dermatol 2017; 10:325-339. [PMID: 28883738 PMCID: PMC5580705 DOI: 10.2147/ccid.s120877] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background and objectives Rates of malignant melanoma are continuing to increase, and until recently effective treatments were lacking. However, since 2011 three immunotherapeutic agents, known as checkpoint inhibitors, have been approved. This review aims to establish whether these three drugs – ipilimumab, nivolumab, and pembrolizumab – offer greater efficacy and tolerability compared to control interventions (placebo, immunotherapy, or chemotherapy) in patients with stage III or IV unresectable cutaneous melanoma. Materials and methods A search on four major medical and scientific databases yielded 7,553 records, of which seven met the inclusion criteria, with a total study population of 3,628. Only prospective Phase II or III randomized controlled trials on checkpoint inhibitors for patients with unresectable cutaneous melanoma that reported data on survival (overall or progression-free), tumor response, or adverse events were included. Three meta-analyses were carried out. Results The hazard ratio for progression or death was 0.54 (95% confidence interval [CI]: 0.44–0.67), and the odds ratio for best overall response rate was 4.48 (95% CI: 2.77–7.24), both in favor of checkpoint inhibitors. However, control treatments were associated with an insignificantly lower rate of discontinuation of treatment due to adverse effects or treatment-related adverse events (odds ratio =1.63 [95% CI: 0.55–4.88]). Conclusion This study finds that checkpoint inhibitors are more effective than control interventions, both in terms of survival and tumor response, and yet no less tolerable. PD1 therapies (nivolumab and pembrolizumab) appear to offer greater efficacy than CTLA4 therapy (ipilimumab). The combination of nivolumab and ipilimumab was, however, the most effective, but significantly less tolerable than monotherapy. The lack of published clinical data does, however, limit this study. Further research is needed in two areas in particular: 1) to determine the optimal use of checkpoint inhibitors, specifically in terms of combination therapy, and 2) to identify reliable biomarkers to predictive responders and guide treatment assignment.
Collapse
Affiliation(s)
| | - Sohag N Saleh
- Faculty of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| |
Collapse
|