1
|
Tapias LF, Shen R, Cassivi SD, Reisenauer JS, Lunn BW, Lechtenberg BJ, Nichols FC, Wigle DA, Blackmon SH. Impact of FDG PET Standardized Uptake Value in Resected Clinical Stage IA Non-Small Cell Lung Cancer. Ann Thorac Surg 2024; 117:1017-1023. [PMID: 37080373 DOI: 10.1016/j.athoracsur.2023.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/28/2023] [Accepted: 04/10/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND A significant proportion of patients with clinical stage IA non-small cell lung cancer (NSCLC) experience will recurrence and decreased survival after surgery. This study examined the impact of preoperative primary tumor positron emission tomography (PET) scan maximum standardized uptake value (SUVmax) on oncologic outcomes after surgery. METHODS This was a retrospective review of 251 patients who underwent surgical treatment of clinical stage IA NSCLC at an academic medical center (2005-2014). Patients were classified according to PET SUVmax level (low vs high) for analysis of upstaging, tumor recurrence, and overall survival. RESULTS Median SUVmax values were higher in squamous cell carcinoma than in adenocarcinoma (median 3.3 vs 7.2; P < .0001). There were 109 (43.4%) patients in the SUVmax low group and 142 (56.6%) in the SUVmax high group. Patients with SUVmax high had larger tumors. SUVmax high was associated with higher rates of nodal upstaging (16.2% vs 4.6% in SUVmax low; P = .004), particularly in N1 nodes. SUVmax high was independently associated with nodal upstaging (adjusted odds ratio, 3.95; 95% CI, 1.36-11.46; P = .011). SUVmax high was associated with time to recurrence (hazard ratio, 1.62; 95% CI, 1.03-2.54; P = .036), but this association was lost on multivariable analysis (hazard ratio, 1.52; 95% CI, 0.91-2.54; P = .106). SUVmax was not associated with overall survival. CONCLUSIONS Preoperative PET SUVmax level is strongly associated with nodal upstaging, particularly in N1 nodes, in patients with clinical stage IA NSCLC who undergo resection. PET SUVmax should be regarded as a risk factor when considering candidacy for sublobar resections and in future trials involving patients with stage I NSCLC.
Collapse
Affiliation(s)
- Luis F Tapias
- Division of Thoracic Surgery, Mayo Clinic, Rochester, Minnesota.
| | - Robert Shen
- Division of Thoracic Surgery, Mayo Clinic, Rochester, Minnesota
| | | | | | - Brendan W Lunn
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Dennis A Wigle
- Division of Thoracic Surgery, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
2
|
Nakada T, Takahashi Y, Sakakura N, Iwata H, Ohtsuka T, Kuroda H. Prognostic Radiological Tools for Clinical Stage IA Pure Solid Lung Cancer. ACTA ACUST UNITED AC 2021; 28:3846-3856. [PMID: 34677246 PMCID: PMC8534325 DOI: 10.3390/curroncol28050328] [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: 08/19/2021] [Revised: 09/20/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022]
Abstract
In this study, we analyzed prognostic radiological tools and surgical outcomes for radiologically pure solid adenocarcinomas (AD) and squamous cell carcinoma (SQ) in clinical stage IA. We retrospectively investigated 130 patients who underwent surgical resections. We assessed the predictive risk factors for recurrence and pathological lymph node metastasis (LNM). There was no statistical difference in recurrence free survival (RFS) or cancer-specific survival (CSS) between AD and SQ groups (p = 0.642 and p = 0.403, respectively). In the whole cohort, tumor size on lung window and mediastinal settings, and tumor disappearance ratio using high-resolution computed tomography (HRCT) were not prognostic parameters (p = 0.127, 0.066, and 0.082, respectively). The maximal standardized uptake value (SUVmax) using positron emission tomography-CT was associated with recurrence (p = 0.016). According to the receiver operating characteristic curve, the cut-off value of SUVmax for recurrence was 4.6 (p = 0.016). The quantitative continuous variables using any radiological tools were not associated with LNM. However, tumor diameter on mediastinal setting ≥8 mm with SUVmax ≥2.4 could be a risk factor for LNM. Pure solid AD and SQ were equivalent for the RFS and CSS. SUVmax was useful to predict recurrence. The tumor diameter on a mediastinal setting and SUVmax were useful in predicting pathological LNM.
Collapse
Affiliation(s)
- Takeo Nakada
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Aichi 464-8681, Japan; (Y.T.); (N.S.); (H.K.)
- Department of Surgery, Division of Thoracic Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan;
- Correspondence: ; Tel.: +81-52-762-6111
| | - Yusuke Takahashi
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Aichi 464-8681, Japan; (Y.T.); (N.S.); (H.K.)
| | - Noriaki Sakakura
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Aichi 464-8681, Japan; (Y.T.); (N.S.); (H.K.)
| | - Hiroshi Iwata
- East Nagoya Radiological Diagnosis Foundation, Aichi 464-0044, Japan;
| | - Takashi Ohtsuka
- Department of Surgery, Division of Thoracic Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan;
| | - Hiroaki Kuroda
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Aichi 464-8681, Japan; (Y.T.); (N.S.); (H.K.)
| |
Collapse
|
3
|
Cucchiara F, Petrini I, Romei C, Crucitta S, Lucchesi M, Valleggi S, Scavone C, Capuano A, De Liperi A, Chella A, Danesi R, Del Re M. Combining liquid biopsy and radiomics for personalized treatment of lung cancer patients. State of the art and new perspectives. Pharmacol Res 2021; 169:105643. [PMID: 33940185 DOI: 10.1016/j.phrs.2021.105643] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Lung cancer has become a paradigm for precision medicine in oncology, and liquid biopsy (LB) together with radiomics may have a great potential in this scenario. They are both minimally invasive, easy to perform, and can be repeated during patient's follow-up. Also, increasing evidence suggest that LB and radiomics may provide an efficient way to screen and diagnose tumors at an early stage, including the monitoring of any change in the tumor molecular profile. This could allow treatment optimization, improvement of patients' quality of life, and healthcare-related costs reduction. Latest reports on lung cancer patients suggest a combination of these two strategies, along with cutting-edge data analysis, to decode valuable information regarding tumor type, aggressiveness, progression, and response to treatment. The approach seems more compatible with clinical practice than the current standard, and provides new diagnostic companions being able to suggest the best treatment strategy compared to conventional methods. To implement radiomics and liquid biopsy directly into clinical practice, an artificial intelligence (AI)-based system could help to link patients' clinical data together with tumor molecular profiles and imaging characteristics. AI could also solve problems and limitations related to LB and radiomics methodologies. Further work is needed, including new health policies and the access to large amounts of high-quality and well-organized data, allowing a complementary and synergistic combination of LB and imaging, to provide an attractive choice e in the personalized treatment of lung cancer.
Collapse
Affiliation(s)
- Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Chiara Romei
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Maurizio Lucchesi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Simona Valleggi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa De Liperi
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Antonio Chella
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy.
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
4
|
Perillo A, Agbaje Olufemi MV, De Robbio J, Mancuso RM, Roscigno A, Tirozzi M, Scognamiglio IR. Liquid biopsy in NSCLC: a new challenge in radiation therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:156-173. [PMID: 36046142 PMCID: PMC9400754 DOI: 10.37349/etat.2021.00038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 02/23/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the most common cancer and the leading cause of cancer mortality worldwide. To date, tissue biopsy has been the gold standard for the diagnosis and the identification of specific molecular mutations, to guide choice of therapy. However, this procedure has several limitations. Liquid biopsy could represent a solution to the intrinsic limits of traditional biopsy. It can detect cancer markers such as circulating tumor DNA or RNA (ctDNA, ctRNA), and circulating tumor cells, in plasma, serum or other biological fluids. This procedure is minimally invasive, reproducible and can be used repeatedly. The main clinical applications of liquid biopsy in non-small cell lung cancer (NSCLC) patients are the early diagnosis, stratification of the risk of relapse, identification of mutations to guide application of targeted therapy and the evaluation of the minimum residual disease. In this review, the current role of liquid biopsy and associated markers in the management of NSCLC patients was analyzed, with emphasis on ctDNA and CTCs, and radiotherapy.
Collapse
Affiliation(s)
- Annarita Perillo
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Mohamed Vincenzo Agbaje Olufemi
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Jacopo De Robbio
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Rossella Margherita Mancuso
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Anna Roscigno
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Maddalena Tirozzi
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Ida Rosalia Scognamiglio
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| |
Collapse
|
5
|
Zhang F, Wu X, Zhu J, Huang Y, Song X, Jiang L. 18F-FDG PET/CT and circulating tumor cells in treatment-naive patients with non-small-cell lung cancer. Eur J Nucl Med Mol Imaging 2021; 48:3250-3259. [PMID: 33630146 DOI: 10.1007/s00259-021-05260-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/11/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE This study retrospectively investigated the clinical utility of 2-deoxy-18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) and circulating tumor cells (CTCs) in the diagnosis and prognosis of treatment-naive patients with non-small-cell lung cancer (NSCLC). METHODS The blood samples of treatment-naive patients with NSCLC were collected for CTCs detection, and the tumor metabolic parameters of 18F-FDG PET/CT, including maximum standard uptake value (SUVmax), metabolic tumor volume of primary lesion (MTV-P) and combination of primary lesion and metastases (MTV-C), and total lesion glycolysis of primary lesion (TLG-P) and combination of primary lesion and metastases (TLG-C), were analyzed. Age, sex, smoking, serum tumor markers, tumor size, location, TNM stage, and genetic mutations were also reviewed. Moreover, progression-free survival (PFS) and overall survival (OS) of these patients were analyzed. RESULTS A total of 309 patients with NSCLC (200 men, 109 women; mean age: 61 ± 9 years) were enrolled in this study, including 217 patients with adenocarcinoma and 92 with squamous cell carcinoma. Of the 309 cases, 11 were misdiagnosed with benign diseases by 18F-FDG PET/CT. CTCs positivity was detected in 234 cases. The sensitivity of 18F-FDG PET/CT and CTCs in NSCLC were 96.4% and 75.7%, respectively. SUVmax, MTV-P, TLG-P, MTV-C, TLG-C, tumor size, and serum CYFRA211 levels were significantly higher in CTCs positive group than negative group; and advanced TNM stage, squamous cell carcinoma, and EGFR wild type presented higher CTCs positivity. Multivariate logistic regression analysis revealed that SUVmax was significantly associated with CTCs positivity. Multivariate cox regression analysis showed that TLG-P, TLG-C, and CTCs were independent predictors of PFS in patients with NSCLC, and TLG-C and CTCs were independent predictors of OS. CONCLUSIONS 18F-FDG PET/CT was superior to CTCs in the diagnosis of treatment-naive patients with NSCLC. The levels of CTCs in the peripheral blood were associated with tumor glucose metabolism in NSCLC. Metabolic parameters of 18F-FDG PET/CT and CTCs could separately predict the outcomes of treatment-naive patients with NSCLC.
Collapse
Affiliation(s)
- Fengxian Zhang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai, 200433, China
| | - Xiaodong Wu
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai, 200433, China
| | - Junjie Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai, 200433, China
| | - Yan Huang
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai, 200433, China
| | - Xiao Song
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai, 200433, China.
| | - Lei Jiang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai, 200433, China.
| |
Collapse
|
6
|
Marcu LG. Imaging Biomarkers of Tumour Proliferation and Invasion for Personalised Lung Cancer Therapy. J Pers Med 2020; 10:jpm10040222. [PMID: 33198090 PMCID: PMC7711676 DOI: 10.3390/jpm10040222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/03/2020] [Accepted: 11/10/2020] [Indexed: 12/28/2022] Open
Abstract
Personalised treatment in oncology has seen great developments over the last decade, due to both technological advances and more in-depth knowledge of radiobiological processes occurring in tumours. Lung cancer therapy is no exception, as new molecular targets have been identified to further increase treatment specificity and sensitivity. Yet, tumour resistance to treatment is still one of the main reasons for treatment failure. This is due to a number of factors, among which tumour proliferation, the presence of cancer stem cells and the metastatic potential of the primary tumour are key features that require better controlling to further improve cancer management in general, and lung cancer treatment in particular. Imaging biomarkers play a key role in the identification of biological particularities within tumours and therefore are an important component of treatment personalisation in radiotherapy. Imaging techniques such as PET, SPECT, MRI that employ tumour-specific biomarkers already play a critical role in patient stratification towards individualized treatment. The aim of the current paper is to describe the radiobiological challenges of lung cancer treatment in relation to the latest imaging biomarkers that can aid in the identification of hostile cellular features for further treatment adaptation and tailoring to the individual patient’s needs.
Collapse
Affiliation(s)
- Loredana G. Marcu
- Faculty of Informatics and Science, University of Oradea, 410087 Oradea, Romania;
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| |
Collapse
|
7
|
Avella DM, Manjunath Y, Singh A, Deroche CB, Kimchi ET, Staveley-O'Carroll KF, Mitchem JB, Kwon E, Li G, Kaifi JT. 18F-FDG PET/CT total lesion glycolysis is associated with circulating tumor cell counts in patients with stage I to IIIA non-small cell lung cancer. Transl Lung Cancer Res 2020; 9:515-521. [PMID: 32676315 PMCID: PMC7354116 DOI: 10.21037/tlcr.2020.04.10] [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] [Indexed: 01/29/2023]
Abstract
Background In non-small cell lung cancer (NSCLC), 18F-fluoro-2-deoxy-D-glucose (18F-FDG) uptake determined by PET and presence of circulating tumor cells (CTCs) in the peripheral blood independently predict outcomes. For 18F-FDG PET/CT staging interpretation, standardized uptake values (SUVmax/avg) are routinely used in clinical reporting. The goal was to investigate whether 18F-FDG uptake measured by SUVmax/avg, but also measures of metabolic tumor volume (MTV) and total lesion glycolysis (TLG) (MTV × SUVavg), are associated with CTCs. Methods Prospectively, 7.5 mL blood was drawn from NSCLC patients at the time of staging 18F-FDG PET/CT and from healthy control subjects. CTCs were identified by immunofluorescent staining (CK8/18/19pos/EpCAMpos/CD45neg/DAPIpos nucleus). 18F-FDG PET/CTs were analyzed for SUVmax, SUVavg, MTV, and TLG. Results In 16 NSCLC patients with stage I–IIIA, MTV and TLG, in contrast to SUVmax and SUVavg, were positively associated with CTCs (linear regression analysis). No CTCs were detectable in 20 healthy control subjects. Conclusions This pilot study demonstrates that 18F-FDG PET/CT TLG correlates with CTCs in NSCLC patients without distant metastases. TLG might be a more appropriate marker for hematogenous micrometastatic potential than SUVs.
Collapse
Affiliation(s)
- Diego M Avella
- Department of Surgery, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Yariswamy Manjunath
- Department of Surgery, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Amolak Singh
- Department of Nuclear Medicine, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA
| | - Chelsea B Deroche
- Biostatistics and Research Design Unit, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA
| | - Eric T Kimchi
- Department of Surgery, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Kevin F Staveley-O'Carroll
- Department of Surgery, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Jonathan B Mitchem
- Department of Surgery, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Eric Kwon
- Department of Surgery, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA
| | - Guangfu Li
- Department of Surgery, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA
| | - Jussuf T Kaifi
- Department of Surgery, Health Management and Medical Informatics, University of Missouri, Columbia, MO, USA.,Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| |
Collapse
|
8
|
Circulating Tumor Cells and Metabolic Parameters in NSCLC Patients Treated with Checkpoint Inhibitors. Cancers (Basel) 2020; 12:cancers12020487. [PMID: 32092983 PMCID: PMC7072667 DOI: 10.3390/cancers12020487] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/04/2020] [Accepted: 02/17/2020] [Indexed: 12/19/2022] Open
Abstract
Circulating tumor cells (CTC) count and characterization have been associated with poor prognosis in recent studies. Our aim was to examine CTC count and its association with metabolic parameters and clinical outcomes in non-small cell lung carcinoma (NSCLC) patients treated with immune checkpoint inhibitors (ICI). For this prospective study, data from 35 patients (23 males, 12 females) were collected and analyzed. All patients underwent an 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG-PET/CT) scan and CTC detection through Isolation by Size of Tumor/Trophoblastic Cells (ISET) from peripheral blood samples obtained at baseline and 8 weeks after ICI initiation. Association of CTC count with clinical and metabolic characteristics was studied. Progression-free survival (PFS) and overall survival (OS) were analyzed using the Kaplan–Meier method and the log-rank test. Median follow-up was 13.2 months (range of 4.9–21.6). CTC were identified in 16 out of 35 patients (45.7%) at baseline and 10 out of 24 patients at 8 weeks (41.7%). Mean CTC numbers before and after 8 weeks were 15 ± 28 and 11 ± 19, respectively. Prior to ICI, the mean CTC number was significantly higher in treatment-naïve patients (34 ± 39 vs. 9 ± 21, p = 0.004). CTC count variation (ΔCTC) was significantly associated with tumor metabolic response set by European Organization for Research and Treatment of Cancer (EORTC) criteria (p = 0.033). At the first restaging, patients with a high tumor burden, that is, metabolic tumor volume (MTV) and total lesion glycolysis (TLG), had a higher CTC count (p = 0.009). The combination of mean CTC and median MTV at 8 weeks was associated with PFS (p < 0.001) and OS (p = 0.024). Multivariate analysis identified CTC count at 8 weeks as an independent predictor for PFS and OS, whereas ΔMTV and maximum standardized uptake value variation (ΔSUVmax) was predictive for PFS and OS, respectively. Our study confirmed that CTC number is modulated by previous treatments and correlates with metabolic response during ICI. Moreover, elevated CTC count, along with metabolic parameters, were found to be prognostic factors for PFS and OS.
Collapse
|
9
|
Carrió I, Flotats A. Liquid biopsies and molecular imaging: friends or foes? Clin Transl Imaging 2019. [DOI: 10.1007/s40336-019-00350-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
10
|
de Miguel-Pérez D, Bayarri-Lara CI, Ortega FG, Russo A, Moyano Rodriguez MJ, Alvarez-Cubero MJ, Maza Serrano E, Lorente JA, Rolfo C, Serrano MJ. Post-Surgery Circulating Tumor Cells and AXL Overexpression as New Poor Prognostic Biomarkers in Resected Lung Adenocarcinoma. Cancers (Basel) 2019; 11:cancers11111750. [PMID: 31703465 PMCID: PMC6896005 DOI: 10.3390/cancers11111750] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 01/05/2023] Open
Abstract
Background: The prognosis of early stage non-small cell lung cancer (NSCLC) is quite disappointing and the benefits of adjuvant therapy are relatively small. Thus, there is an urgent need to identify novel prognostic and predictive biomarkers. Lung adenocarcinoma has distinct clinical–pathological characteristics and novel therapeutic strategies are under active evaluation in the adjuvant setting. Here, we investigated the prognostic impact of circulating tumor cells (CTCs) and gene and miRNA tissue expression in resectable NSCLC. Patients and methods: We assessed the association between CTC subpopulations and the outcome of resected early stage lung adenocarcinoma (ADC) patients at three different time-points (CTC1-3) (before surgery, after one month, and after six months) in comparison to squamous cell carcinoma (SCC). Furthermore, gene and miRNA tissue expression, immunoprofiling, and epithelial-to-mesenchymal transition (EMT) markers were correlated with outcome. Results: ADC (n = 47) and SCC (n = 50) revealed different tissue expression profiles, resulting in the presence of different CTC subpopulations. In ADC, miR-155 correlated with AXL and IL6R expression, which were related to the presence of EMT CTC1 (p = 0.014 and p = 0.004). In the multivariate analysis, CTC2 was an independent prognostic factor for relapse-free survival, and CTC3 and AXL were independent prognostic for overall survival only in ADC. Neither the surgery nor the adjuvant treatment influenced the prognosis of these patients. Conclusions: Our study elucidate the prognostic impact of tissue AXL expression and the presence of CTCs after surgery in adenocarcinoma patients. Tissue AXL expression and CTC EMT activation could potentially represent biomarkers for the stratification of ADC patients that might benefit from new adjuvant therapies.
Collapse
Affiliation(s)
- Diego de Miguel-Pérez
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain; (D.d.M.-P.); (F.G.O.); (M.J.A.-C.); (E.M.S.); (J.A.L.)
- Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada. Av. de la Investigación, 11, 18071 Granada, Spain
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine. 22 S. Greene Street, Baltimore, MD 21201, USA;
| | - Clara Isabel Bayarri-Lara
- Department of Thoracic Surgery, Virgen de las Nieves University Hospital, Av. de las Fuerzas Armadas, 2, 18014 Granada, Spain; (C.I.B.-L.); (M.J.M.R.)
| | - Francisco Gabriel Ortega
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain; (D.d.M.-P.); (F.G.O.); (M.J.A.-C.); (E.M.S.); (J.A.L.)
| | - Alessandro Russo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine. 22 S. Greene Street, Baltimore, MD 21201, USA;
| | - María José Moyano Rodriguez
- Department of Thoracic Surgery, Virgen de las Nieves University Hospital, Av. de las Fuerzas Armadas, 2, 18014 Granada, Spain; (C.I.B.-L.); (M.J.M.R.)
| | - Maria Jesus Alvarez-Cubero
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain; (D.d.M.-P.); (F.G.O.); (M.J.A.-C.); (E.M.S.); (J.A.L.)
| | - Elizabeth Maza Serrano
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain; (D.d.M.-P.); (F.G.O.); (M.J.A.-C.); (E.M.S.); (J.A.L.)
- Integral Oncology Division, San Cecilio Clinical University Hospital, Calle Dr. Oloriz 16, 18012 Granada, Spain
| | - José Antonio Lorente
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain; (D.d.M.-P.); (F.G.O.); (M.J.A.-C.); (E.M.S.); (J.A.L.)
- Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada. Av. de la Investigación, 11, 18071 Granada, Spain
| | - Christian Rolfo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine. 22 S. Greene Street, Baltimore, MD 21201, USA;
- Correspondence: (C.R.); (M.J.S.); Tel.: +1-410-328-7224 (C.R.); +34-958-715-500 (M.J.S.)
| | - María José Serrano
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain; (D.d.M.-P.); (F.G.O.); (M.J.A.-C.); (E.M.S.); (J.A.L.)
- Integral Oncology Division, San Cecilio Clinical University Hospital, Calle Dr. Oloriz 16, 18012 Granada, Spain
- Correspondence: (C.R.); (M.J.S.); Tel.: +1-410-328-7224 (C.R.); +34-958-715-500 (M.J.S.)
| |
Collapse
|
11
|
Russo A, De Miguel Perez D, Gunasekaran M, Scilla K, Lapidus R, Cooper B, Mehra R, Adamo V, Malapelle U, Rolfo C. Liquid biopsy tracking of lung tumor evolutions over time. Expert Rev Mol Diagn 2019; 19:1099-1108. [PMID: 31608732 DOI: 10.1080/14737159.2020.1680287] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: The rise of the personalized era in lung cancer prompted the evaluation of novel diagnostic tools to overcome some of the limits of traditional tumor genotyping. Liquid biopsy refers to a multitude of minimally invasive techniques that can allow a real-time biomolecular characterization of the tumor through the analysis of human body fluids.Areas covered: Herein we provide a comprehensive overview of the role of liquid biopsy in lung cancer, mainly focusing on the most studied members of the liquid biopsy family, cell-free DNA (cfDNA) and circulating tumor cells (CTCs).Expert opinion: Among the different components of the large liquid biopsy family, cfDNA is the most studied and widely adopted source for tumor genotyping in lung cancer, already entered clinical practice for detection of both sensitizing and resistance EGFR mutations. However, the impressive technological advances made in the last few years are expanding its potential applications, allowing a more comprehensive plasma genotyping through next-generation sequencing and moving from advanced/metastatic disease to novel frontiers, such as early detection and minimal residual disease evaluation.
Collapse
Affiliation(s)
- Alessandro Russo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Messina, Italy
| | - Diego De Miguel Perez
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS, Granada, Spain.,Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Granada, Spain
| | - Muthukumar Gunasekaran
- Division of Cardiovascular Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Katherine Scilla
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rena Lapidus
- Translational Core Laboratory, University of Maryland Greenebaum Cancer Center, Baltimore, MD, USA
| | - Brandon Cooper
- Translational Core Laboratory, University of Maryland Greenebaum Cancer Center, Baltimore, MD, USA
| | - Ranee Mehra
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vincenzo Adamo
- Medical Oncology Unit, A.O. Papardo & Department of Human Pathology, University of Messina, Messina, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Christian Rolfo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Pan L, Yan G, Chen W, Sun L, Wang J, Yang J. Distribution of circulating tumor cell phenotype in early cervical cancer. Cancer Manag Res 2019; 11:5531-5536. [PMID: 31354357 PMCID: PMC6588089 DOI: 10.2147/cmar.s198391] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/18/2019] [Indexed: 12/15/2022] Open
Abstract
Background and objective: Circulating tumor cells (CTCs) can be classified into three phenotypes based on epithelial-to-mesenchymal transition (EMT) markers, including epithelial CTCs, mesenchymal CTCs, and mixed phenotypic CTCs. This study is aimed to analyze the correlation between CTC phenotypes and the clinicopathological features of patients with early cervical cancer. Methods: Peripheral blood samples were obtained from 90 patients with early cervical cancer. CTCs were isolated and classified. The correlations of CTC counts and CTC phenotypes with clinicopathological features of patients were analyzed. Results: The positivity rate for CTCs in patients with stage I-IIA cervical cancer was 90%. An increased CTC number was observed in patients with FIGO stage II, pelvic lymph node metastasis, and lymphovascular involvement. There were 38.89% epithelial CTCs, 23.33% mesenchymal CTCs, and 14.44% mixed phenotypic CTCs, Mesenchymal CTCs were more common in patients with FIGO stage II, pelvic lymph node metastasis, lymphovascular involvement, and deep stromal invasion. Conclusion: CTCs with mesenchymal phenotypes are closely related to pelvic lymph node metastasis and lymphatic vascular invasion in stage I-IIA cervical cancer. Detection of circulating tumor cell phenotypes is helpful for the early diagnosis of cervical cancer micro-metastasis and for the assessment of disease status.
Collapse
Affiliation(s)
- Li Pan
- Radiotherapy Department, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| | - Gaoshu Yan
- Radiotherapy Department, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| | - Wenli Chen
- Radiotherapy Department, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| | - Lei Sun
- Radiotherapy Department, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| | - Jichuan Wang
- Radiotherapy Department, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| | - Jialin Yang
- Radiotherapy Department, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| |
Collapse
|
13
|
Saarenheimo J, Eigeliene N, Andersen H, Tiirola M, Jekunen A. The Value of Liquid Biopsies for Guiding Therapy Decisions in Non-small Cell Lung Cancer. Front Oncol 2019; 9:129. [PMID: 30891428 PMCID: PMC6411700 DOI: 10.3389/fonc.2019.00129] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 02/13/2019] [Indexed: 12/15/2022] Open
Abstract
Targeted therapies have allowed for an individualized treatment approach in non-small-cell lung cancer (NSCLC). The initial therapeutic decisions and success of targeted therapy depend on genetic identification of personal tumor profiles. Tissue biopsy is the gold standard for molecular analysis, but non-invasive or minimally invasive liquid biopsy methods are also now used in clinical practice, allowing for later monitoring and optimization of the cancer treatment. The inclusion of liquid biopsy in the management of NSCLC provides strong evidence on early treatment response, which becomes a basis for determining disease progression and the need for changes in treatment. Liquid biopsies can drive the decision making for treatment strategies to achieve better patient outcomes. Cell-free DNA and circulating tumor cells obtained from the blood are promising markers for determining patient status. They may improve cancer treatments, allow for better treatment control, enable early interventions, and change decision making from reactive actions toward more predictive early interventions. This review aimed to present current knowledge on and the usefulness of liquid biopsy studies in NSCLC from the perspective of how it has allowed individualized treatments according to gene profiling and how the method may alter the treatment decisions in the future.
Collapse
Affiliation(s)
- Jatta Saarenheimo
- Department of Pathology, Vasa Central Hospital, Vaasa, Finland.,Department of Biological and Environmental Science, Nano Science Center, University of Jyväskylä, Jyväskylä, Finland
| | - Natalja Eigeliene
- Department of Oncology, Vasa Central Hospital, Vaasa, Finland.,Department of Oncology and Radiotherapy, University of Turku, Turku, Finland
| | - Heidi Andersen
- Department of Pulmonology, Vasa Central Hospital, Vaasa, Finland
| | - Marja Tiirola
- Department of Biological and Environmental Science, Nano Science Center, University of Jyväskylä, Jyväskylä, Finland
| | - Antti Jekunen
- Department of Oncology, Vasa Central Hospital, Vaasa, Finland.,Department of Oncology and Radiotherapy, University of Turku, Turku, Finland
| |
Collapse
|