1
|
Arenare L, Di Liello R, De Placido P, Gridelli C, Morabito A, Pignata S, Nuzzo F, Avallone A, Maiello E, Gargiulo P, Schettino C, Gravina A, Gallo C, Chiodini P, Di Maio M, Perrone F, Piccirillo MC. Under-reporting of subjective symptoms and its prognostic value: a pooled analysis of 12 cancer clinical trials. ESMO Open 2024; 9:102941. [PMID: 38452437 PMCID: PMC10937229 DOI: 10.1016/j.esmoop.2024.102941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Oncologists tend to under-report subjective symptoms during cancer treatment. This study describes the under-reporting rate of selected symptoms and explores its association with overall survival (OS). A secondary aim is to test the association of patient-reported symptoms with OS. PATIENTS AND METHODS This is a post hoc analysis on data pooled from 12 randomized trials, promoted by the National Cancer Institute of Naples (Italy), enrolling patients between 2002 and 2019, with published primary analyses. Occurrence and grade of six side-effects (anorexia, nausea, vomiting, constipation, diarrhea and fatigue) reported by physicians were compared with corresponding symptoms reported by patients in quality-of-life (QoL) questionnaires. Under-reporting was defined as the rate of cases reported grade 0 by the physician while grade ≥1 by the patient. Prognostic value was tested in a multivariable model stratified by trial, including age, sex and performance status as confounders. A landmark threshold was defined for OS analyses. RESULTS 3792 patients with advanced lung, ovarian, pancreatic, breast or colorectal cancer were pooled; 2603 (68.6%) were eligible having at least one toxicity assessment and one QoL questionnaire, before the first planned disease restaging. Concordance between physicians' and patients' reporting was low with Cohen's k coefficients ranging from 0.03 (fatigue) to 0.33 (vomiting). Under-reporting ranged from 52.7% (nausea) to 80.5% (anorexia), and was not associated with OS. Patient-reported anorexia, vomiting and fatigue ('a little' or more) were significantly associated with shorter OS. CONCLUSIONS Under-reporting of treatment side-effects is frequent, but it does not affect OS. Patients' reported symptoms should be used for prognostic evaluation.
Collapse
Affiliation(s)
- L Arenare
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples
| | - R Di Liello
- Oncologia Medica, P.O. Ospedale del Mare-ASL Napoli 1 Centro, Naples
| | - P De Placido
- Department of Clinical Medicine and Surgery, Università Federico II, Naples
| | - C Gridelli
- Divisione di Oncologia Medica, A.O.R.N. San Giuseppe Moscati, Contrada Amoretta, Avellino
| | - A Morabito
- Oncologia Clinica Sperimentale Toraco-Polmonare, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples
| | - S Pignata
- Oncologia Clinica Sperimentale Uroginecologica Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples
| | - F Nuzzo
- Oncologia Clinica Sperimentale Di Senologia, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples
| | - A Avallone
- Oncologia Clinica Sperimentale Addominale, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples
| | - E Maiello
- Oncologia, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo
| | - P Gargiulo
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples
| | - C Schettino
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples
| | - A Gravina
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples
| | - C Gallo
- Statistica Medica, Università della Campania 'Luigi Vanvitelli', Naples
| | - P Chiodini
- Statistica Medica, Università della Campania 'Luigi Vanvitelli', Naples
| | - M Di Maio
- Department of Oncology, Università di Torino, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - F Perrone
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples
| | - M C Piccirillo
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples.
| |
Collapse
|
2
|
Wu J, Wang X, Li Z, Yi X, Hu D, Wang Q, Zhong T. Small extracellular vesicles promote the formation of the pre-metastatic niche through multiple mechanisms in colorectal cancer. Cell Cycle 2024; 23:131-149. [PMID: 38341861 PMCID: PMC11037293 DOI: 10.1080/15384101.2024.2311501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/24/2024] [Indexed: 02/13/2024] Open
Abstract
Colorectal cancer (CRC) ranks among the most prevalent global malignancies, posing significant threats to human life and health due to its high recurrence and metastatic potential. Small extracellular vesicles (sEVs) released by CRC play a pivotal role in the formation of the pre-metastatic niche (PMN) through various mechanisms, preparing the groundwork for accelerated metastatic invasion. This review systematically describes how sEVs promote CRC metastasis by upregulating inflammatory factors, promoting immunosuppression, enhancing angiogenesis and vascular permeability, promoting lymphangiogenesis and lymphatic network remodeling, determining organophilicity, promoting stromal cell activation and remodeling and inducing the epithelial-to-mesenchymal transition (EMT). Furthermore, we explore potential mechanisms by which sEVs contribute to PMN formation in CRC and propose novel insights for CRC diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Jiyang Wu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiaoxing Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zhengzhe Li
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiaomei Yi
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Die Hu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qi Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
3
|
Avallone A, Piccirillo MC, Nasti G, Rosati G, Carlomagno C, Di Gennaro E, Romano C, Tatangelo F, Granata V, Cassata A, Silvestro L, De Stefano A, Aloj L, Vicario V, Nappi A, Leone A, Bilancia D, Arenare L, Petrillo A, Lastoria S, Gallo C, Botti G, Delrio P, Izzo F, Perrone F, Budillon A. Effect of Bevacizumab in Combination With Standard Oxaliplatin-Based Regimens in Patients With Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA Netw Open 2021; 4:e2118475. [PMID: 34309665 PMCID: PMC8314140 DOI: 10.1001/jamanetworkopen.2021.18475] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IMPORTANCE Although bevacizumab is a standard of care in combination treatments for metastatic colorectal cancer (mCRC), its clinical benefit has been limited. OBJECTIVE To determine whether sequential scheduling of bevacizumab administration in combination with chemotherapy improves treatment efficacy in patients with mCRC, in keeping with the tumor vascular normalization hypothesis. DESIGN, SETTING, AND PARTICIPANTS This open-label, randomized clinical phase 3 trial was conducted from May 8, 2012, to December 9, 2015, at 3 Italian centers. Patients aged 18 to 75 years with unresectable, previously untreated, or single line-treated mCRC were recruited. Follow-up was completed December 31, 2019, and data were analyzed from February 26 to July 24, 2020. INTERVENTIONS Patients received 12 biweekly cycles of standard oxaliplatin-based regimens (modified FOLFOX-6 [levo-folinic acid, fluorouracil, and oxaliplatin]/modified CAPOX [capecitabine and oxaliplatin]) plus bevacizumab administered either on the same day as chemotherapy (standard arm) or 4 days before chemotherapy (experimental arm). MAIN OUTCOMES AND MEASURES The primary end point was the objective response rate (ORR) measured with Response Evaluation Criteria in Solid Tumors, version 1.1. Secondary end points included progression-free survival, overall survival, safety, and quality of life (QOL). RESULTS Overall, 230 patients (136 men [59.1%]; median age, 62.3 [interquartile range, 53.3-67.6] years) were randomly assigned to the standard arm (n = 115) or the experimental arm (n = 115). The median duration of follow-up was 68.3 (95% CI, 61.0-70.0) months. No difference in ORR (57.4% [95% CI, 47.8%-66.6%] in the standard arm and 56.5% [95% CI, 47.0-65.7] in the experimental arm; P = .89) or progression-free survival (10.5 [95% CI, 9.1-12.3] months in the standard arm and 11.7 [95% CI, 9.9-12.9] months in the experimental arm; P = .15) was observed. However, the median overall survival was 29.8 (95% CI, 22.5-41.1) months in the experimental arm compared with 24.1 (95% CI, 18.6-29.8) months in the standard arm (adjusted hazard ratio, 0.73; 95% CI, 0.54-0.99; P = .04). Moreover, the experimental arm was associated with a significant reduction in the rate of severe diarrhea (6 [5.3%] vs 19 [16.5%]; P = .006) and nausea (2 [1.8%] vs 8 [7.0%]; P = .05) and improved physical functioning (mean [SD] change from baseline, 0.65 [1.96] vs -7.41 [2.95] at 24 weeks; P = .02), and constipation scores (mean [SD] change from baseline, -17.2 [3.73] vs -0.62 [4.44]; P = .003). CONCLUSIONS AND RELEVANCE In this randomized clinical trial, sequential administration of bevacizumab plus chemotherapy did not improve ORR, the primary end point. However, the overall survival advantage, fewer adverse effects, and better health-related QOL associated with sequential bevacizumab administration might provide the basis for exploring antiangiogenic combination treatments with innovative perspectives. TRIAL REGISTRATION EudraCT Identifier: 2011-004997-27; ClinicalTrials.gov Identifier: NCT01718873.
Collapse
Affiliation(s)
- Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Maria C. Piccirillo
- Clinical Trials Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Guglielmo Nasti
- Innovative Therapy for Abdominal Metastases, IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Gerardo Rosati
- Medical Oncology Unit, S. Carlo Hospital, Potenza, Italy
| | - Chiara Carlomagno
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Carmela Romano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Fabiana Tatangelo
- Pathology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Vincenza Granata
- Radiology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Antonino Cassata
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Lucrezia Silvestro
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Alfonso De Stefano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Luigi Aloj
- Nuclear Medicine Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
- currently affiliated with Department of Radiology, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Valeria Vicario
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Anna Nappi
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | | | - Laura Arenare
- Clinical Trials Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Antonella Petrillo
- Radiology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Secondo Lastoria
- Nuclear Medicine Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Ciro Gallo
- Università della Campania Luigi Vanvitelli, Napoli, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Paolo Delrio
- Colorectal Oncological Surgery, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Francesco Izzo
- Colorectal Oncological Surgery, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
- Hepatobiliary Surgery Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Franco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
4
|
Avallone A, Piccirillo MC, Di Gennaro E, Romano C, Calabrese F, Roca MS, Tatangelo F, Granata V, Cassata A, Cavalcanti E, Maurea N, Maiolino P, Silvestro L, De Stefano A, Giuliani F, Rosati G, Tamburini E, Aprea P, Vicario V, Nappi A, Vitagliano C, Casaretti R, Leone A, Petrillo A, Botti G, Delrio P, Izzo F, Perrone F, Budillon A. Randomized phase II study of valproic acid in combination with bevacizumab and oxaliplatin/fluoropyrimidine regimens in patients with RAS-mutated metastatic colorectal cancer: the REVOLUTION study protocol. Ther Adv Med Oncol 2020; 12:1758835920929589. [PMID: 32849914 PMCID: PMC7425244 DOI: 10.1177/1758835920929589] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/04/2020] [Indexed: 01/30/2023] Open
Abstract
Background Despite effective treatments, metastatic colorectal cancer (mCRC) prognosis is still poor, mostly in RAS-mutated tumors, thus suggesting the need for novel combinatorial therapies. Epigenetic alterations play an important role in initiation and progression of cancers, including CRC. Histone-deacetylase inhibitors (HDACi) have shown activity in combination with chemotherapy in the treatment of solid tumors. Owing to its HDACi activity and its safe use for epileptic disorders, valproic acid (VPA) is a good candidate for anticancer therapy that we have largely explored preclinically translating our findings in currently ongoing clinical studies. We have shown in CRC models that HDACi, including VPA, induces synergistic antitumor effects in combination with fluoropyrimidines. Furthermore, unpublished results from our group demonstrated that VPA induces differentiation and sensitization of CRC stem cells to oxaliplatin. Moreover, preclinical and clinical data suggest that HDACi may prevent/reverse anti-angiogenic resistance. Methods/Design A randomized, open-label, two-arm, multicenter phase-II study will be performed to explore whether the addition of VPA to first line bevacizumab/oxaliplatin/fluoropyrimidine regimens (mFOLFOX-6/mOXXEL) might improve progression-free survival (PFS) in RAS-mutated mCRC patients. A sample size of 200 patients was calculated under the hypothesis that the addition of VPA to chemotherapy/bevacizumab can improve PFS from 9 to 12 months, with one-sided alpha of 0.20 and a power of 0.80. Secondary endpoints are overall survival, objective response rate, metastases resection rate, toxicity, and quality of life. Moreover, the study will explore several prognostic and predictive biomarkers on blood samples, primary tumors, and on resected metastases. Discussion The "Revolution" study aims to improve the treatment efficacy of RAS-mutated mCRC through an attractive strategy evaluating the combination of VPA with standard cancer treatment. Correlative studies could identify novel biomarkers and could add new insight in the mechanism of interaction between VPA, fluoropyrimidine, oxaliplatin, and bevacizumab. Trial Registration EudraCT: 2018-001414-15; ClinicalTrials.gov identifier: NCT04310176.
Collapse
Affiliation(s)
- Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Via M. Semmola, Napoli, 80131, Italy
| | | | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Carmela Romano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Filomena Calabrese
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Fabiana Tatangelo
- Pathology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Vincenza Granata
- Radiology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Antonio Cassata
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Ernesta Cavalcanti
- Laboratory Medicine Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Nicola Maurea
- Cardiology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Lucrezia Silvestro
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Alfonso De Stefano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | | | - Gerardo Rosati
- Medical Oncology Unit, S. Carlo Hospital, Potenza, Italy
| | - Emiliano Tamburini
- Dipartimento di Oncologia e Cure Palliative, Azienda Ospedaliera Cardinale G. Panico, Tricase-Lecce, Italy
| | - Pasquale Aprea
- Vascular Access Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Valeria Vicario
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Anna Nappi
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Carlo Vitagliano
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Rossana Casaretti
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Antonella Petrillo
- Radiology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Paolo Delrio
- Colorectal Oncological Surgery, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Francesco Izzo
- Hepatobiliary Surgery Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori- IRCCS-Fondazione G. Pascale, Via M. Semmola, Napoli, 80131, Italy
| |
Collapse
|
5
|
El Kaffas A, Hoogi A, Zhou J, Durot I, Wang H, Rosenberg J, Tseng A, Sagreiya H, Akhbardeh A, Rubin DL, Kamaya A, Hristov D, Willmann JK. Spatial Characterization of Tumor Perfusion Properties from 3D DCE-US Perfusion Maps are Early Predictors of Cancer Treatment Response. Sci Rep 2020; 10:6996. [PMID: 32332790 PMCID: PMC7181711 DOI: 10.1038/s41598-020-63810-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/26/2020] [Indexed: 02/08/2023] Open
Abstract
There is a need for noninvasive repeatable biomarkers to detect early cancer treatment response and spare non-responders unnecessary morbidities and costs. Here, we introduce three-dimensional (3D) dynamic contrast enhanced ultrasound (DCE-US) perfusion map characterization as inexpensive, bedside and longitudinal indicator of tumor perfusion for prediction of vascular changes and therapy response. More specifically, we developed computational tools to generate perfusion maps in 3D of tumor blood flow, and identified repeatable quantitative features to use in machine-learning models to capture subtle multi-parametric perfusion properties, including heterogeneity. Models were developed and trained in mice data and tested in a separate mouse cohort, as well as early validation clinical data consisting of patients receiving therapy for liver metastases. Models had excellent (ROC-AUC > 0.9) prediction of response in pre-clinical data, as well as proof-of-concept clinical data. Significant correlations with histological assessments of tumor vasculature were noted (Spearman R > 0.70) in pre-clinical data. Our approach can identify responders based on early perfusion changes, using perfusion properties correlated to gold-standard vascular properties.
Collapse
Affiliation(s)
- Ahmed El Kaffas
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Stanford, CA, USA. .,Department of Radiology, Integrative Biomedical Imaging Informatics at Stanford, School of Medicine, Stanford University, Stanford, CA, USA. .,Department of Radiology, Body Imaging, Stanford University, Stanford, CA, USA.
| | - Assaf Hoogi
- Department of Radiology, Integrative Biomedical Imaging Informatics at Stanford, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jianhua Zhou
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Stanford, CA, USA
| | - Isabelle Durot
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Stanford, CA, USA
| | - Huaijun Wang
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jarrett Rosenberg
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Stanford, CA, USA
| | - Albert Tseng
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Stanford, CA, USA
| | - Hersh Sagreiya
- Department of Radiology, Integrative Biomedical Imaging Informatics at Stanford, School of Medicine, Stanford University, Stanford, CA, USA
| | - Alireza Akhbardeh
- Department of Radiology, Integrative Biomedical Imaging Informatics at Stanford, School of Medicine, Stanford University, Stanford, CA, USA
| | - Daniel L Rubin
- Department of Radiology, Integrative Biomedical Imaging Informatics at Stanford, School of Medicine, Stanford University, Stanford, CA, USA
| | - Aya Kamaya
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Stanford, CA, USA.,Department of Radiology, Body Imaging, Stanford University, Stanford, CA, USA
| | - Dimitre Hristov
- Department of Radiation Oncology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jürgen K Willmann
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Stanford, CA, USA.,Department of Radiology, Body Imaging, Stanford University, Stanford, CA, USA
| |
Collapse
|
6
|
Zhang Q, Wang Q, Wang X, Li J, Shen L, Peng Z. Regorafenib, TAS-102, or fruquintinib for metastatic colorectal cancer: any difference in randomized trials? Int J Colorectal Dis 2020; 35:295-306. [PMID: 31848739 DOI: 10.1007/s00384-019-03477-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2019] [Indexed: 02/04/2023]
Abstract
PURPOSE Direct randomized comparisons of regorafenib, TAS-102, and fruquintinib for treating metastatic colorectal cancer (mCRC) are lacking. Here, we evaluated the efficacy and safety of three agents by a systematic review and a network meta-analysis. METHODS We included phase III randomized controlled trials in the PubMed, Embase, and Scopus Cochrane databases and ClinicalTrials.gov registry from initiation until January 2019. Data from randomized controlled trials including overall survival (OS), progression-free survival (PFS), and adverse events (AEs) were extracted. Direct meta-analysis and indirect meta-analysis using network meta-analysis were assessed. RESULTS Five trials comprising a total of 2586 patients were included. For efficacy analysis of OS, no statistically significant differences were observed between regorafenib and TAS-102 (HR 0.945, 95% CI [0.677, 1.320], P = 0.753), regorafenib and fruquintinib (HR 1.056, 95% CI [0.690, 1.621], P = 0.814), or TAS-102 and fruquintinib (HR 1.117, 95% CI [0.740, 1.685], P = 0.610). However, fruquintinib was superior in PFS compared with TAS-102 (HR 1.756, 95% CI [1.079, 2.857], P = 0.023). Regorafenib and TAS-102 appeared to have a similar effect on PFS (HR 0.907, 95% CI [0.611, 1.346], P = 0.641), as did regorafenib and fruquintinib (HR 1.592, 95% CI [0.968, 2.618], P = 0.067). None of the three agents were better in terms of all grade AEs or any grade of 3-5 AEs. However, subgroup analysis of AEs exhibited different toxicity profiles between the three drugs. CONCLUSIONS Indirect comparison suggested that the three agents had similar OS but that fruquintinib was superior in terms of PFS compared with that of TAS-102. These three agents had different toxicity profiles.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Qianqian Wang
- Department of Molecular Orthopaedics, Beijing Institute of Traumatology and Orthopaedics, Beijing, 100035, China
| | - Xicheng Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
7
|
Herrera M, Galindo-Pumariño C, García-Barberán V, Peña C. A Snapshot of The Tumor Microenvironment in Colorectal Cancer: The Liquid Biopsy. Int J Mol Sci 2019; 20:ijms20236016. [PMID: 31795332 PMCID: PMC6929174 DOI: 10.3390/ijms20236016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
The molecular profile of liquid biopsies is emerging as an alternative to tissue biopsies in the clinical management of malignant diseases. In colorectal cancer, significant liquid biopsy-based biomarkers have demonstrated an ability to discriminate between asymptomatic cancer patients and healthy controls. Furthermore, this non-invasive approach appears to provide relevant information regarding the stratification of tumors with different prognoses and the monitoring of treatment responses. This review focuses on the tumor microenvironment components which are detected in blood samples of colorectal cancer patients and might represent potential biomarkers. Exosomes released by tumor and stromal cells play a major role in the modulation of cancer progression in the primary tumor microenvironment and in the formation of an inflammatory pre-metastatic niche. Stromal cells-derived exosomes are involved in driving mechanisms that promote tumor growth, migration, metastasis, and drug resistance, therefore representing substantial signaling mediators in the tumor-stroma interaction. Besides, recent findings of specifically packaged exosome cargo in Cancer-Associated Fibroblasts of colorectal cancer patients identify novel exosomal biomarkers with potential clinical applicability. Furthermore, additional different signals emitted from the tumor microenvironment and also detectable in the blood, such as soluble factors and non-tumoral circulating cells, arise as novel promising biomarkers for cancer diagnosis, prognosis, and treatment response prediction. The therapeutic potential of these factors is still limited, and studies are in their infancy. However, innovative strategies aiming at the inhibition of tumor progression by systemic exosome depletion, exosome-mediated circulating tumor cell capturing, and exosome-drug delivery systems are currently being studied and may provide considerable advantages in the near future.
Collapse
Affiliation(s)
- Mercedes Herrera
- Department of Oncology-Pathology, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Cristina Galindo-Pumariño
- Medical Oncology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, 28034 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
| | - Vanesa García-Barberán
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
- Laboratorio de Oncología Molecular, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
- Correspondence: (V.G.-B.); (C.P.)
| | - Cristina Peña
- Medical Oncology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, 28034 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
- Correspondence: (V.G.-B.); (C.P.)
| |
Collapse
|
8
|
Granata V, Fusco R, Avallone A, Catalano O, Piccirillo M, Palaia R, Nasti G, Petrillo A, Izzo F. A radiologist's point of view in the presurgical and intraoperative setting of colorectal liver metastases. Future Oncol 2018; 14:2189-2206. [PMID: 30084273 DOI: 10.2217/fon-2018-0080] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multidisciplinary management of patients with metastatic colorectal cancer requires in each phase an adequate choice of the most appropriate imaging modality. The first challenging step is liver lesions detection and characterization, using several imaging modality ultrasound, computed tomography, magnetic resonance and positron emission tomography. The criteria to establish the metastases resectability have been modified. Not only the lesions number and site but also the functional volume remnant after surgery and the quality of the nontumoral liver must be taken into account. Radiologists should identify the liver functional volume remnant and during liver surgical procedures should collaborate with the surgeon to identify all lesions, including those that disappeared after the therapy, using intraoperative ultrasound with or without contrast medium.
Collapse
Affiliation(s)
- Vincenza Granata
- Radiology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Roberta Fusco
- Radiology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Antonio Avallone
- Abdominal Oncology Division, Istitutonazionale Tumori - IRCSS - Fondazione G Pascale, Napoli, Italia
| | - Orlando Catalano
- Radiology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Mauro Piccirillo
- Hepatobiliary Surgical Oncology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Raffaele Palaia
- Hepatobiliary Surgical Oncology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Guglielmo Nasti
- Abdominal Oncology Division, Istitutonazionale Tumori - IRCSS - Fondazione G Pascale, Napoli, Italia
| | - Antonella Petrillo
- Radiology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| | - Francesco Izzo
- Hepatobiliary Surgical Oncology Division, Istitutonazionale Tumori - IRCCS - Fondazione G Pascale, Napoli, Italia
| |
Collapse
|
9
|
Sturrock M, Miller IS, Kang G, Hannis Arba'ie N, O'Farrell AC, Barat A, Marston G, Coletta PL, Byrne AT, Prehn JH. Anti-angiogenic drug scheduling optimisation with application to colorectal cancer. Sci Rep 2018; 8:11182. [PMID: 30046049 PMCID: PMC6060139 DOI: 10.1038/s41598-018-29318-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/04/2018] [Indexed: 11/09/2022] Open
Abstract
Bevacizumab (bvz) is a first choice anti-angiogenic drug in oncology and is primarily administered in combination with chemotherapy. It has been hypothesized that anti-angiogenic drugs enhance efficacy of cytotoxic drugs by "normalizing" abnormal tumor vessels and improving drug penetration. Nevertheless, the clinical relevance of this phenomenon is still unclear with several studies over recent years suggesting an opposing relationship. Herein, we sought to develop a new computational tool to interrogate anti-angiogenic drug scheduling with particular application in the setting of colorectal cancer (CRC). Specifically, we have employed a mathematical model of vascular tumour growth which interrogates the impact of anti-angiogenic treatment and chemotherapeutic treatment on tumour volume. Model predictions were validated using CRC xenografts which underwent treatment with a clinically relevant combinatorial anti-angiogenic regimen. Bayesian model selection revealed the most appropriate term for capturing the effect of treatments on the tumour size, and provided insights into a switch-like dependence of FOLFOX delivery on the tumour vasculature. Our experimental data and mathematical model suggest that delivering chemotherapy prior to bvz may be optimal in the colorectal cancer setting.
Collapse
Affiliation(s)
- M Sturrock
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - I S Miller
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - G Kang
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - N Hannis Arba'ie
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - A C O'Farrell
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - A Barat
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - G Marston
- Liverpool Hope University, Hope Park, Liverpool, L16 9JD, UK
| | - P L Coletta
- School of Medicine, University of Leeds Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - A T Byrne
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.,Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - J H Prehn
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| |
Collapse
|
10
|
Unseld M, Drimmel M, Siebenhüner A, Gleiss A, Bianconi D, Kieler M, Scheithauer W, Winder T, Prager GW. Optimizing Treatment Sequence for Late-line Metastatic Colorectal Cancer Patients Using Trifluridine/Tipiracil and Regorafenib. Clin Colorectal Cancer 2018; 17:274-279. [PMID: 30042010 DOI: 10.1016/j.clcc.2018.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/08/2018] [Accepted: 05/31/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Treatment sequencing for patients with refractory metastatic colorectal cancer (mCRC) has been highly debated. The thymidine-based nucleoside trifluridine/tipiracil (TAS-102) and the multikinase inhibitor regorafenib have demonstrated clinical benefits in randomized phase III trials compared with placebo. However, limited data are available on the most optimal therapy sequence involving TAS-102 and regorafenib. PATIENTS AND METHODS In the present retrospective, observational, real-life study, clinical data on mCRC patients treated with TAS-102 or an alternative salvage treatment at the Medical University of Vienna and University Hospital Zurich were collected from January 2013 to December 2016. RESULTS A total of 85 patients whose disease had progressed during fluoropyrimidine-based therapy (FBT) with or without an antibody were included. The disease control rate in patients treated with TAS-102 after FBT-based treatment was 24% compared with 35% in patients treated with regorafenib after FBT-based treatment (adjusted odds ratio, 1.75; 95% confidence interval, 0.41-7.47; P = .449). The progression-free survival (PFS) and overall survival (OS) for patients treated with TAS-102 was 2.8 months (quartile, 2.0-4.8 months) and 15.9 months, respectively. When the data were analyzed according to the subgroups of patients with or without an FBT-free period, the TAS-102-treated patients with a previous FBT-free interval had a PFS of 3.1 months and OS of 17.7 months compared with a PFS of 2.2 months and OS of 8.1 months for patients who received TAS-102 immediately after FBT. CONCLUSION Our results have confirmed the efficacy of TAS-102 and regorafenib in the real-life setting. The treatment sequence analysis showed a tendency for longer PFS and OS for TAS-102-treated patients after an FBT-free interval. Prospective randomized data are needed to gain more information about the most beneficial therapy sequence in the salvage treatment of mCRC.
Collapse
Affiliation(s)
- Matthias Unseld
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Magdalena Drimmel
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Andreas Gleiss
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Daniela Bianconi
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Markus Kieler
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Werner Scheithauer
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Thomas Winder
- Department of Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Gerald W Prager
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
11
|
Mollard S, Ciccolini J, Imbs DC, El Cheikh R, Barbolosi D, Benzekry S. Model driven optimization of antiangiogenics + cytotoxics combination: application to breast cancer mice treated with bevacizumab + paclitaxel doublet leads to reduced tumor growth and fewer metastasis. Oncotarget 2018; 8:23087-23098. [PMID: 28416742 PMCID: PMC5410287 DOI: 10.18632/oncotarget.15484] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/07/2017] [Indexed: 11/25/2022] Open
Abstract
Bevacizumab is the first-in-class antiangiogenic drug and is almost always administrated in combination with cytotoxics. Reports have shown that bevacizumab could induce a transient phase of vascular normalization, thus ensuring a better drug delivery when cytotoxics administration is adjuvant. However, determining the best sequence remains challenging. We have developed a mathematical model describing the impact of antiangiogenics on tumor vasculature. A 3.4 days gap between bevacizumab and paclitaxel was first proposed by our model. To test its relevance, 84 mice were orthotopically xenografted with human MDA-231Luc+ refractory breast cancer cells. Two sets of experiments were performed, based upon different bevacizumab dosing (10 or 20 mg/kg) and inter-cycle intervals (7 or 10 days), comprising several combinations with paclitaxel. Results showed that scheduling bevacizumab 3 days before paclitaxel improved antitumor efficacy (48% reduction in tumor size compared with concomitant dosing, p < 0.05) and reduced metastatic spreading. Additionally, bevacizumab alone could lead to more aggressive metastatic disease with shorter survival in animals. Our model was able to fit the experimental data and provided insights on the underlying dynamics of the vasculature's ability to deliver the cytotoxic agent. Final simulations suggested a new, data-informed optimal gap of 2.2 days. Our experimental data suggest that current concomitant dosing between bevacizumab and paclitaxel could be a sub-optimal strategy at bedside. In addition, this proof of concept study suggests that mathematical modelling could help to identify the optimal interval among a variety of possible alternate treatment modalities, thus refining the way experimental or clinical studies are conducted.
Collapse
Affiliation(s)
- Severine Mollard
- SMARTc Unit, Inserm S_911 CRO2, Aix Marseille University, Marseille, France.,Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joseph Ciccolini
- SMARTc Unit, Inserm S_911 CRO2, Aix Marseille University, Marseille, France
| | | | - Raouf El Cheikh
- SMARTc Unit, Inserm S_911 CRO2, Aix Marseille University, Marseille, France
| | | | | |
Collapse
|
12
|
Cao B, Zhou X, Yang W, Ma J, Zhou W, Fan D, Hong L. The role of cell-free DNA in predicting colorectal cancer prognosis. Expert Rev Gastroenterol Hepatol 2018; 12:39-48. [PMID: 28838275 DOI: 10.1080/17474124.2017.1372191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Colorectal cancer is a cancer of the digestive system with poor prognosis. Cell-free DNA has received much attention with its unique predominance, especially in colorectal cancer. Areas covered: This study has summarized recent advancements and challenges regarding cell-free DNA in predicting CRC prognosis. Furthermore, the authors make predictions on the potential developments concerning cell-free DNA in future prognosis prediction techniques. Expert commentary: Cell-free DNA has the value of predicting CRC prognosis as an important biomarke. Further clinical trials should be performed to promote translating cell-free DNA into clinical applications.
Collapse
Affiliation(s)
- Bo Cao
- a The First Brigade of Student , Fourth Military Medical University , Xi'an , China
| | - Xin Zhou
- a The First Brigade of Student , Fourth Military Medical University , Xi'an , China
| | - Wanli Yang
- b State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Jiaojiao Ma
- b State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Wei Zhou
- b State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Daiming Fan
- b State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Liu Hong
- b State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| |
Collapse
|
13
|
Lee J, Rhee WJ, Chang JS, Chang SK, Koom WS. Evaluation of predictive factors of vertebral compression fracture after conventional palliative radiotherapy for spinal metastasis from colorectal cancer. J Neurosurg Spine 2017; 28:333-340. [PMID: 29271724 DOI: 10.3171/2017.6.spine17282] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Delayed consequences of spinal radiotherapy (RT), including vertebral compression fracture (VCF), are critical complications. However, the predisposing factors that contribute to VCF after conventional RT are unclear. The aim of this study was to assess the incidence of VCF and to determine the predictors of VCF following conventional spinal RT specific to colorectal cancer (CRC). METHODS The authors retrospectively reviewed 237 spinal segments (147 metastatic and 90 nonmetastatic) in 53 patients with CRC who underwent RT with a median total dose of 30 Gy in 10 fractions between January 2007 and December 2014. The primary end point was the development of a VCF following RT, either de novo VCF or the progression of a baseline VCF. VCFs were assessed using the spinal instability neoplastic score (SINS) criteria. RESULTS Among all 237 spinal segments, 22 VCFs (9.3%) were observed following RT, including 13 de novo and 9 progressive fractures, and the median time to VCF was 4 months. All VCFs developed in metastatic spines. Among 147 metastatic spinal segments, 22 fractures were observed, with a 12-month cumulative incidence of VCF of 14.8%. Results of multivariable analysis indicated sex (p = 0.023) and SINS class II/III (p < 0.001) as risk factors related to development of a VCF in metastatic spinal segments. Among the SINS criteria, a lytic tumor and the presence of a baseline VCF were identified as predictors of VCF in metastatic spinal segments. CONCLUSIONS In osteolytic or mixed lesions that were predominant in spinal metastases of CRC, the incidence of VCF was not negligible, even in patients treated with conventional spinal RT. This was especially evident in patients with spinal metastases with a SINS score ≥ 7. Presence of a baseline VCF after spinal RT is a predictor of VCF development and should be observed carefully.
Collapse
Affiliation(s)
- Jeongshim Lee
- 1Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul.,2Department of Radiation Oncology, Inha University Hospital, Incheon; and
| | - Woo Joong Rhee
- 1Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul
| | - Jee Suk Chang
- 1Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul
| | - Sei Kyung Chang
- 3Department of Radiation Oncology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Woong Sub Koom
- 1Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul
| |
Collapse
|
14
|
Iwamoto N, Shimada T. Recent advances in mass spectrometry-based approaches for proteomics and biologics: Great contribution for developing therapeutic antibodies. Pharmacol Ther 2017; 185:147-154. [PMID: 29274706 DOI: 10.1016/j.pharmthera.2017.12.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Since the turn of the century, mass spectrometry (MS) technologies have continued to improve dramatically, and advanced strategies that were impossible a decade ago are increasingly becoming available. The basic characteristics behind these advancements are MS resolution, quantitative accuracy, and information science for appropriate data processing. The spectral data from MS contain various types of information. The benefits of improving the resolution of MS data include accurate molecular structural-derived information, and as a result, we can obtain a refined biomolecular structure determination in a sequential and large-scale manner. Moreover, in MS data, not only accurate structural information but also the generated ion amount plays an important rule. This progress has greatly contributed a research field that captures biological events as a system by comprehensively tracing the various changes in biomolecular dynamics. The sequential changes of proteome expression in biological pathways are very essential, and the amounts of the changes often directly become the targets of drug discovery or indicators of clinical efficacy. To take this proteomic approach, it is necessary to separate the individual MS spectra derived from each biomolecule in the complexed biological samples. MS itself is not so infinite to perform the all peak separation, and we should consider improving the methods for sample processing and purification to make them suitable for injection into MS. The above-described characteristics can only be achieved using MS with any analytical instrument. Moreover, MS is expected to be applied and expand into many fields, not only basic life sciences but also forensic medicine, plant sciences, materials, and natural products. In this review, we focus on the technical fundamentals and future aspects of the strategies for accurate structural identification, structure-indicated quantitation, and on the challenges for pharmacokinetics of high-molecular-weight protein biopharmaceuticals.
Collapse
Affiliation(s)
- Noriko Iwamoto
- Leading Technology of Bioanalysis and Protein Chemistry, SHIMADZU Corporation, Kyoto, Japan
| | - Takashi Shimada
- Leading Technology of Bioanalysis and Protein Chemistry, SHIMADZU Corporation, Kyoto, Japan.
| |
Collapse
|
15
|
Granata V, Fusco R, Catalano O, Avallone A, Palaia R, Botti G, Tatangelo F, Granata F, Cascella M, Izzo F, Petrillo A. Diagnostic accuracy of magnetic resonance, computed tomography and contrast enhanced ultrasound in radiological multimodality assessment of peribiliary liver metastases. PLoS One 2017; 12:e0179951. [PMID: 28632786 PMCID: PMC5478136 DOI: 10.1371/journal.pone.0179951] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022] Open
Abstract
PURPOSE We compared diagnostic performance of Magnetic Resonance (MR), Computed Tomography (CT) and Ultrasound (US) with (CEUS) and without contrast medium to identify peribiliary metastasis. METHODS We identified 35 subjects with histological proven peribiliary metastases who underwent CEUS, CT and MR study. Four radiologists evaluated the presence of peribiliary lesions, using a 4-point confidence scale. Echogenicity, density and T1-Weigthed (T1-W), T2-W and Diffusion Weighted Imaging (DWI) signal intensity as well as the enhancement pattern during contrast studies on CEUS, CT and MR so as hepatobiliary-phase on MRI was assessed. RESULTS All lesions were detected by MR. CT detected 8 lesions, while US/CEUS detected one lesion. According to the site of the lesion, respect to the bile duct and hepatic parenchyma: 19 (54.3%) were periductal, 15 (42.8%) were intra-periductal and 1 (2.8%) was periductal-intrahepatic. According to the confidence scale MRI had the best diagnostic performance to assess the lesion. CT obtained lower diagnostic performance. There was no significant difference in MR signal intensity and contrast enhancement among all metastases (p>0.05). There was no significant difference in CT density and contrast enhancement among all metastases (p>0.05). CONCLUSIONS MRI is the method of choice for biliary tract tumors but it does not allow a correct differential diagnosis among different histological types of metastasis. The presence of biliary tree dilatation without hepatic lesions on CT and US/CEUS study may be an indirect sign of peribiliary metastases and for this reason the patient should be evaluated by MRI.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| | - Roberta Fusco
- Division of Radiology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| | - Orlando Catalano
- Division of Radiology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| | - Antonio Avallone
- Division of Abdominal Oncology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| | - Raffaele Palaia
- Division of Hepatobiliary Surgical Oncology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| | - Gerardo Botti
- Division of Diagnostic Pathology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| | - Fabiana Tatangelo
- Division of Diagnostic Pathology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| | - Francesco Granata
- Departement of Civil and Mechanical Engineering, University of Cassino and Southern Lazio, Cassino, Italy
| | - Marco Cascella
- Division of Anesthesia, Endoscopy and Cardiology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| | - Francesco Izzo
- Division of Hepatobiliary Surgical Oncology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, “Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale”, Naples, Italy
| |
Collapse
|
16
|
Chebib R, Verlingue L, Cozic N, Faron M, Burtin P, Boige V, Hollebecque A, Malka D. Angiogenesis inhibition in the second-line treatment of metastatic colorectal cancer: A systematic review and pooled analysis. Semin Oncol 2017; 44:114-128. [PMID: 28923209 DOI: 10.1053/j.seminoncol.2017.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/18/2017] [Accepted: 07/31/2017] [Indexed: 12/26/2022]
Abstract
The last two decades have seen intensive efforts devoted to the development of compounds that target angiogenesis for the treatment of metastatic colorectal cancer (mCRC). In this review, we describe supporting evidence and ongoing development of angiogenesis inhibitors in the second-line treatment of mCRC, and summarize relevant randomized trials to help therapeutic decision-making in daily practice.
Collapse
Affiliation(s)
- Ralph Chebib
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Paris, France
| | - Loic Verlingue
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Paris, France
| | - Nathalie Cozic
- Service de Biostatistique et d'Epidémiologie, Gustave Roussy, Université Paris-Saclay, Paris, France; INSERM U1018, CESP, Gustave Roussy, Université Paris-Saclay, Paris, France; Ligue Nationale Contre le Cancer Meta-Analysis Platform, Gustave Roussy, Université Paris-Saclay, Paris France
| | - Matthieu Faron
- Service de Biostatistique et d'Epidémiologie, Gustave Roussy, Université Paris-Saclay, Paris, France; INSERM U1018, CESP, Gustave Roussy, Université Paris-Saclay, Paris, France; Département de Chirurgie Digestive, Gustave Roussy, Université Paris-Saclay, Paris, France
| | - Pascal Burtin
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Paris, France
| | - Valérie Boige
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Paris, France
| | - Antoine Hollebecque
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Paris, France; Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Villejuif, France
| | - David Malka
- Département de Médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Paris, France.
| |
Collapse
|