1
|
Mondelo-Macía P, García-González J, León-Mateos L, Abalo A, Bravo S, Chantada Vazquez MDP, Muinelo-Romay L, López-López R, Díaz-Peña R, Dávila-Ibáñez AB. Identification of a Proteomic Signature for Predicting Immunotherapy Response in Patients With Metastatic Non-Small Cell Lung Cancer. Mol Cell Proteomics 2024; 23:100834. [PMID: 39216661 PMCID: PMC11474190 DOI: 10.1016/j.mcpro.2024.100834] [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: 05/26/2023] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Immunotherapy has improved survival rates in patients with cancer, but identifying those who will respond to treatment remains a challenge. Advances in proteomic technologies have enabled the identification and quantification of nearly all expressed proteins in a single experiment. Integrating mass spectrometry with high-throughput technologies has facilitated comprehensive analysis of the plasma proteome in cancer, facilitating early diagnosis and personalized treatment. In this context, our study aimed to investigate the predictive and prognostic value of plasma proteome analysis using the SWATH-MS (Sequential Window Acquisition of All Theoretical Mass Spectra) strategy in newly diagnosed patients with non-small cell lung cancer (NSCLC) receiving pembrolizumab therapy. We enrolled 64 newly diagnosed patients with advanced NSCLC treated with pembrolizumab. Blood samples were collected from all patients before and during therapy. A total of 171 blood samples were analyzed using the SWATH-MS strategy. Plasma protein expression in metastatic NSCLC patients prior to receiving pembrolizumab was analyzed. A first cohort (discovery cohort) was employed to identify a proteomic signature predicting immunotherapy response. Thus, 324 differentially expressed proteins between responder and non-responder patients were identified. In addition, we developed a predictive model and found a combination of seven proteins, including ATG9A, DCDC2, HPS5, FIL1L, LZTL1, PGTA, and SPTN2, with stronger predictive value than PD-L1 expression alone. Additionally, survival analyses showed an association between the levels of ATG9A, DCDC2, SPTN2 and HPS5 with progression-free survival (PFS) and/or overall survival (OS). Our findings highlight the potential of proteomic technologies to detect predictive biomarkers in blood samples from NSCLC patients, emphasizing the correlation between immunotherapy response and the idenfied protein set.
Collapse
Affiliation(s)
- Patricia Mondelo-Macía
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Jorge García-González
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain; Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Luis León-Mateos
- Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain; Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Alicia Abalo
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain
| | - Susana Bravo
- Proteomic Unit, Instituto de Investigaciones Sanitarias-IDIS, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - María Del Pilar Chantada Vazquez
- Proteomic Unit, Instituto de Investigaciones Sanitarias-IDIS, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Rafael López-López
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain; Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain; Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Roberto Díaz-Peña
- Fundación Pública Galega de Medicina Xenómica, SERGAS; Grupo de Medicina Xenomica-USC, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; Faculty of Health Sciences, Universidad Autónoma de Chile, Talca, Chile
| | - Ana B Dávila-Ibáñez
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain; Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
| |
Collapse
|
2
|
Khan F, Jones K, Lyon P. Immune checkpoint inhibition: a future guided by radiology. Br J Radiol 2023; 96:20220565. [PMID: 36752570 PMCID: PMC10321249 DOI: 10.1259/bjr.20220565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/04/2023] [Accepted: 01/29/2023] [Indexed: 02/09/2023] Open
Abstract
The limitation of the function of antitumour immune cells is a common hallmark of cancers that enables their survival. As such, the potential of immune checkpoint inhibition (ICI) acts as a paradigm shift in the treatment of a range of cancers but has not yet been fully capitalised. Combining minimally and non-invasive locoregional therapies offered by radiologists with ICI is now an active field of research with the aim of furthering therapeutic capabilities in medical oncology. In parallel to this impending advancement, the "imaging toolbox" available to radiologists is also growing, enabling more refined tumour characterisation as well as greater accuracy in evaluating responses to therapy. Options range from metabolite labelling to cellular localisation to immune checkpoint screening. It is foreseeable that these novel imaging techniques will be integrated into personalised treatment algorithms. This growth in the field must include updating the current standardised imaging criteria to ensure they are fit for purpose. Such criteria is crucial to both appropriately guide clinical decision-making regarding next steps of treatment, but also provide reliable prognosis. Quantitative approaches to these novel imaging techniques are also already being investigated to further optimise personalised therapeutic decision-making. The therapeutic potential of specific ICIs and locoregional therapies could be determined before administration thus limiting unnecessary side-effects whilst maintaining efficacy. Several radiological aspects of oncological care are advancing simultaneously. Therefore, it is essential that each development is assessed for clinical use and optimised to ensure the best treatment decisions are being offered to the patient. In this review, we discuss state of the art advances in novel functional imaging techniques in the field of immuno-oncology both pre-clinically and clinically.
Collapse
Affiliation(s)
- Faraaz Khan
- Foundation Doctor, Buckinghamshire Hospitals NHS Trust, Amersham, Buckinghamshire, United Kingdom
| | - Keaton Jones
- Academic Clinical Lecturer Nuffield Department of Surgical Sciences University of Oxford, Wellington Square, Oxford, United Kingdom
| | - Paul Lyon
- Consultant Radiologist, Department of Radiology, Oxford University Hospitals, Headington, Oxford, United Kingdom
| |
Collapse
|
3
|
Riano I, Abuali I, Sharma A, Durant J, Dragnev KH. Role of Neoadjuvant Immune Checkpoint Inhibitors in Resectable Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2023; 16:233. [PMID: 37259381 PMCID: PMC9963056 DOI: 10.3390/ph16020233] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 08/23/2024] Open
Abstract
The neoadjuvant use of immune checkpoint inhibitors (ICI) in resectable non-small cell lung cancer (NSCLC) is being increasingly adopted, but questions about the most appropriate applications remain. Although patients with resectable NSCLC are often treated with surgery and adjuvant chemotherapy or targeted therapies +/- radiotherapy, they still have a high risk of recurrence and death. In recent years, immune checkpoint inhibitors (ICI) (anti-PD-1/PD-L1 and anti-CTLA-4) have provided a new and effective therapeutic strategy for the treatment of advanced NSCLC. Therefore, it is possible that ICIs for early-stage NSCLC may follow the pattern established in metastatic disease. Currently, there are several ongoing trials to determine the efficacy in the neoadjuvant setting for patients with local or regional disease. To date, only nivolumab in combination with chemotherapy has been approved by the U.S. FDA in the preoperative setting, but data continue to evolve rapidly, and treatment guidelines need to be determined. In this article, we review the current preclinical and clinical evidence on neoadjuvant ICIs alone and combination in the treatment of early-stage NSCLC.
Collapse
Affiliation(s)
- Ivy Riano
- Section of Medical Oncology, Dartmouth Cancer Center, Dartmouth Health, 1 Medical Center Drive, Lebanon, NH 03756, USA
- Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH 03755, USA
| | - Inas Abuali
- Division of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Aditya Sharma
- Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH 03755, USA
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Dartmouth Health, 1 Medical Drive, Lebanon, NH 03756, USA
| | - Jewelia Durant
- Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH 03755, USA
| | - Konstantin H. Dragnev
- Section of Medical Oncology, Dartmouth Cancer Center, Dartmouth Health, 1 Medical Center Drive, Lebanon, NH 03756, USA
- Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH 03755, USA
| |
Collapse
|
4
|
Wang M, Lu D, Bi M. Influence of concomitant gastric acid suppressants use on the survival of patients with non-small cell lung cancer treated with programmed death-1/ligand-1 inhibitors: A meta-analysis. Int Immunopharmacol 2022; 110:108955. [PMID: 35750017 DOI: 10.1016/j.intimp.2022.108955] [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: 02/17/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Influence of concomitant use of gastric acid suppressants (GAS) on survival of non-small cell lung cancer (NSCLC) patients receiving programmed death-1/ligand-1 (PD-1/PD-L1) inhibitors has rarely been comprehensively evaluated. A meta-analysis was performed to systematically evaluate the effect of concomitant GAS in NSCLC patients receiving PD-1/PD-L1inhibitors. METHODS Relevant observational studies were identified by search of Medline, Embase, and Web of Science databases from inception to May 26, 2022. A random-effect model which incorporates the possible between-study heterogeneity was used to combine the results. RESULTS Ten retrospective and one prospective cohort studies including 5892 patients were patients were included. Influence of concomitant proton pump inhibitors (PPIs) was evaluated in ten studies, and influence of GAS, including PPIs or histamine type-2 receptor antagonists were evaluated in one study. Pooled results showed that concomitant use of GAS was associated with worse progression-free survival (PFS, adjusted hazard ratio [HR]: 1.32, 95% confidence interval [CI]: 1.20 to 1.45, P < 0.001; I2 = 0%) and overall survival (OS, adjusted HR: 1.36, 95% CI: 1.26 to 1.48, P < 0.001; I2 = 0%) in NSCLC patients taking PD-1/PD-L1inhibitors. Subgroup analyses indicated that the association between concomitant use of GAS and poor survival in NSCLC patients taking PD-L1inhibitors was consistent in univariate and multivariate studies (P values for subgroup difference both > 0.05 for PFS and OS). CONCLUSIONS The meta-analysis by summarizing the up-to-date literatures showed that use of GAS, primarily PPIs, may be associated with poor survival outcomes in patients with NSCLC receiving PD-1/PD-L1inhibitors.
Collapse
Affiliation(s)
- Mingyu Wang
- Department of Oncology, the First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China; Department of Respiratory Disease, the First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Depeng Lu
- Department of Gastroenterology, the First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Minghong Bi
- Department of Oncology, the First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China.
| |
Collapse
|
5
|
Zhang L, Wang H, Tian J, Sui L, Chen X. Concomitant Statins and the Survival of Patients with Non-Small-Cell Lung Cancer Treated with Immune Checkpoint Inhibitors: A Meta-Analysis. Int J Clin Pract 2022; 2022:3429462. [PMID: 35855055 PMCID: PMC9276478 DOI: 10.1155/2022/3429462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/17/2022] [Accepted: 05/28/2022] [Indexed: 11/17/2022] Open
Abstract
Statins are suggested to improve cancer survival by possible anti-inflammatory effect. However, it remains unclear if concomitant use of statins could improve the efficacy of immune checkpoint inhibitors (ICIs) in patients with non-small-cell lung cancer (NSCLC). Accordingly, a meta-analysis was performed to systematically evaluate the effect of concomitant statins in NSCLC patients receiving ICIs. Relevant studies were obtained by literature search in PubMed, Embase, and Web of Science databases. A conservative random-effect model was used to combine the results. Eight cohorts including 2382 patients were included. The programmed death-1/ligand-1 inhibitors were used in seven studies; while the cytotoxic T-lymphocyte-associated protein 4 inhibitors were used in the other study. It was shown that concomitant use of statin did not significantly affect the progression-free survival (PFS, hazard ratio (HR): 0.86, 95% confidence interval (CI): 0.70 to 1.07, P=0.17; I 2 = 62%) or overall survival (OS, HR: 0.86, 95% CI: 0.74 to 1.01, P=0.07; I 2 = 29%) of NSCLC patients receiving ICIs. Subgroup analyses showed consistent results in studies with univariate or multivariate analytic models (P for subgroup analysis = 0.97 and 0.38 for the outcome of PFS and OS, respectively). In conclusion, concomitant use of statin seemed to have no significant influence on the survival of patients with NSCLC who were treated with ICIs.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Oncology, The Hospital of Shunyi District of Beijing, Beijing 101300, China
| | - Hong Wang
- Department of Oncology, The Hospital of Shunyi District of Beijing, Beijing 101300, China
| | - Jizheng Tian
- Department of Oncology, The Hospital of Shunyi District of Beijing, Beijing 101300, China
| | - Lili Sui
- Department of Oncology, The Hospital of Shunyi District of Beijing, Beijing 101300, China
| | - Xiaoyan Chen
- Department of Oncology, The Hospital of Shunyi District of Beijing, Beijing 101300, China
| |
Collapse
|
6
|
Mondelo‐Macía P, García‐González J, León‐Mateos L, Anido U, Aguín S, Abdulkader I, Sánchez‐Ares M, Abalo A, Rodríguez‐Casanova A, Díaz‐Lagares Á, Lago‐Lestón RM, Muinelo‐Romay L, López‐López R, Díaz‐Peña R. Clinical potential of circulating free DNA and circulating tumour cells in patients with metastatic non-small-cell lung cancer treated with pembrolizumab. Mol Oncol 2021; 15:2923-2940. [PMID: 34465006 PMCID: PMC8564635 DOI: 10.1002/1878-0261.13094] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors, such as pembrolizumab, are revolutionizing therapeutic strategies for different cancer types, including non-small-cell lung cancer (NSCLC). However, only a subset of patients benefits from this therapy, and new biomarkers are needed to select better candidates. In this study, we explored the value of liquid biopsy analyses, including circulating free DNA (cfDNA) and circulating tumour cells (CTCs), as a prognostic or predictive tool to guide pembrolizumab therapy. For this purpose, a total of 109 blood samples were collected from 50 patients with advanced NSCLC prior to treatment onset and at 6 and 12 weeks after the initiation of pembrolizumab. Plasma cfDNA was measured using hTERT quantitative PCR assay. The CTC levels at baseline were also analysed using two enrichment technologies (CellSearch® and Parsortix systems) to evaluate the efficacy of both approaches at detecting the presence of programmed cell death ligand 1 on CTCs. Notably, patients with high baseline hTERT cfDNA levels had significantly shorter progression-free survival (PFS) and overall survival (OS) than those with low baseline levels. Moreover, patients with unfavourable changes in the hTERT cfDNA levels from baseline to 12 weeks showed a higher risk of disease progression. Additionally, patients in whom CTCs were detected using the CellSearch® system had significantly shorter PFS and OS than patients who had no CTCs. Finally, multivariate regression analyses confirmed the value of the combination of CTCs and cfDNA levels as an early independent predictor of disease progression, identifying a subgroup of patients who were negative for CTCs, who presented low levels of cfDNA and who particularly benefited from the treatment.
Collapse
Affiliation(s)
- Patricia Mondelo‐Macía
- Liquid Biopsy Analysis UnitTranslational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
- Universidade de Santiago de Compostela (USC)Santiago de CompostelaSpain
| | - Jorge García‐González
- Department of Medical OncologyComplexo Hospitalario Universitario de Santiago de Compostela (SERGAS)Santiago de CompostelaSpain
- Translational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSantiago de CompostelaSpain
| | - Luis León‐Mateos
- Department of Medical OncologyComplexo Hospitalario Universitario de Santiago de Compostela (SERGAS)Santiago de CompostelaSpain
- Translational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSantiago de CompostelaSpain
| | - Urbano Anido
- Department of Medical OncologyComplexo Hospitalario Universitario de Santiago de Compostela (SERGAS)Santiago de CompostelaSpain
- Translational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Santiago Aguín
- Department of Medical OncologyComplexo Hospitalario Universitario de Santiago de Compostela (SERGAS)Santiago de CompostelaSpain
- Translational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Ihab Abdulkader
- Department of PathologyComplexo Hospital Universitario de Santiago de Compostela (SERGAS)Universidade de Santiago de CompostelaSantiago de CompostelaSpain
| | - María Sánchez‐Ares
- Department of PathologyComplexo Hospital Universitario de Santiago de Compostela (SERGAS)Universidade de Santiago de CompostelaSantiago de CompostelaSpain
| | - Alicia Abalo
- Liquid Biopsy Analysis UnitTranslational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Aitor Rodríguez‐Casanova
- Cancer EpigenomicsTranslational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
- Roche‐CHUS Joint UnitTranslational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Ángel Díaz‐Lagares
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSantiago de CompostelaSpain
- Cancer EpigenomicsTranslational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Ramón Manuel Lago‐Lestón
- Liquid Biopsy Analysis UnitTranslational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
| | - Laura Muinelo‐Romay
- Liquid Biopsy Analysis UnitTranslational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSantiago de CompostelaSpain
| | - Rafael López‐López
- Department of Medical OncologyComplexo Hospitalario Universitario de Santiago de Compostela (SERGAS)Santiago de CompostelaSpain
- Translational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSantiago de CompostelaSpain
| | - Roberto Díaz‐Peña
- Liquid Biopsy Analysis UnitTranslational Medical Oncology (Oncomet)Health Research Institute of Santiago (IDIS)Santiago de CompostelaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSantiago de CompostelaSpain
| |
Collapse
|
7
|
Castello A, Lopci E. The Role of PET/CT in the Era of Immune Checkpoint Inhibitors: State of Art. Curr Radiopharm 2020; 13:24-31. [PMID: 31749440 DOI: 10.2174/1874471012666191015100106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/09/2019] [Accepted: 07/17/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have achieved astonishing results and improved overall survival (OS) in several types of malignancies, including advanced melanoma. However, due to a peculiar type of anti-cancer activity provided by these drugs, the response patterns during ICI treatment are completely different from that with "old" chemotherapeutic agents. OBJECTIVE To provide an overview of the available literature and potentials of 18F-FDG PET/CT in advanced melanoma during the course of therapy with ICI in the context of treatment response evaluation. METHOD Morphologic criteria, expressed by Response Evaluation Criteria in Solid Tumors (RECIST), immune-related response criteria (irRC), irRECIST, and, more recently, immune-RECIST (iRECIST), along with response criteria based on the metabolic parameters with 18F-Fluorodeoxyglucose (18FFDG), have been explored. RESULTS To overcome the limits of traditional response criteria, new metabolic response criteria have been introduced on time and are being continuously updated, such as the PET/CT Criteria for the early prediction of Response to Immune checkpoint inhibitor Therapy (PECRIT), the PET Response Evaluation Criteria for Immunotherapy (PERCIMT), and "immunotherapy-modified" PET Response Criteria in Solid Tumors (imPERCIST). The introduction of new PET radiotracers, based on monoclonal antibodies combined with radioactive elements ("immune-PET"), are of great interest. CONCLUSION Although the role of 18F-FDG PET/CT in malignant melanoma has been widely validated for detecting distant metastases and recurrences, evidences in course of ICI are still scarce and larger multicenter clinical trials are needed.
Collapse
Affiliation(s)
- Angelo Castello
- Nuclear Medicine, Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Egesta Lopci
- Nuclear Medicine, Humanitas Clinical and Research Hospital, Rozzano, Italy
| |
Collapse
|
8
|
Keam S, Gill S, Ebert MA, Nowak AK, Cook AM. Enhancing the efficacy of immunotherapy using radiotherapy. Clin Transl Immunology 2020; 9:e1169. [PMID: 32994997 PMCID: PMC7507442 DOI: 10.1002/cti2.1169] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/04/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022] Open
Abstract
Recent clinical breakthroughs in cancer immunotherapy, especially with immune checkpoint blockade, offer great hope for cancer sufferers - and have greatly changed the landscape of cancer treatment. However, whilst many patients achieve clinical responses, others experience minimal benefit or do not respond to immune checkpoint blockade at all. Researchers are therefore exploring multimodal approaches by combining immune checkpoint blockade with conventional cancer therapies to enhance the efficacy of treatment. A growing body of evidence from both preclinical studies and clinical observations indicates that radiotherapy could be a powerful driver to augment the efficacy of immune checkpoint blockade, because of its ability to activate the antitumor immune response and potentially overcome resistance. In this review, we describe how radiotherapy induces DNA damage and apoptosis, generates immunogenic cell death and alters the characteristics of key immune cells in the tumor microenvironment. We also discuss recent preclinical work and clinical trials combining radiotherapy and immune checkpoint blockade in thoracic and other cancers. Finally, we discuss the scheduling of immune checkpoint blockade and radiotherapy, biomarkers predicting responses to combination therapy, and how these novel data may be translated into the clinic.
Collapse
Affiliation(s)
- Synat Keam
- National Centre for Asbestos Related DiseasesPerthWAAustralia
- School of MedicineThe University of Western AustraliaPerthWAAustralia
| | - Suki Gill
- Department of Radiation OncologySir Charles Gairdner HospitalPerthWAAustralia
| | - Martin A Ebert
- Department of Radiation OncologySir Charles Gairdner HospitalPerthWAAustralia
- School of Physics, Mathematics and ComputingThe University of Western AustraliaPerthWAAustralia
| | - Anna K Nowak
- National Centre for Asbestos Related DiseasesPerthWAAustralia
- School of MedicineThe University of Western AustraliaPerthWAAustralia
- Department of Medical OncologySir Charles Gairdner HospitalNedlands, PerthWAAustralia
| | - Alistair M Cook
- National Centre for Asbestos Related DiseasesPerthWAAustralia
- School of MedicineThe University of Western AustraliaPerthWAAustralia
| |
Collapse
|
9
|
Wang L, Hu Y, Wang S, Shen J, Wang X. Biomarkers of immunotherapy in non-small cell lung cancer. Oncol Lett 2020; 20:139. [PMID: 32934707 PMCID: PMC7471728 DOI: 10.3892/ol.2020.11999] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 06/24/2020] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy has markedly improved the survival rate of patients with non-small cell lung cancer (NSCLC) and has introduced a new era in lung cancer treatment. However, not all patients with lung cancer benefit from checkpoint blockade, and some suffer from notable immunotoxicities. Thus, it is crucial to identify potential biomarkers suitable for screening the population that may benefit from immunotherapy. Based on the current clinical trials, the aim of the present study was to review the biomarkers for immune checkpoint inhibition, as well as other effective, invalid and hyperprogression markers that may have the potential to better predict responders to immunotherapy among patients with NSCLC. All these biomarkers may be incorporated into the predictive utility of bio-score systems and decision-making algorithms, to better guide the application of immunotherapy in the clinical setting.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yue Hu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Shengchao Wang
- Department of Gynecological Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Jiali Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiaochen Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Breast Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
10
|
Maung TZ, Ergin HE, Javed M, Inga EE, Khan S. Immune Checkpoint Inhibitors in Lung Cancer: Role of Biomarkers and Combination Therapies. Cureus 2020; 12:e8095. [PMID: 32542150 PMCID: PMC7292688 DOI: 10.7759/cureus.8095] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide, with a poor prognosis. Despite aggressive treatment, progression-free survival (PFS) and overall survival are limited. Recently, various kinds of immune checkpoint inhibitors (ICIs) have emerged for several cancers, targeting PD1, PDL1, and CTLA-4. ICIs have made a significant breakthrough in cancer and revolutionized the management of cancer including lung cancer. However, there are a lot of controversies regarding which group of patients is most suitable to be treated with ICIs in terms of monotherapy, combination, and predictive biomarkers. We reviewed various kinds of studies, such as meta-analysis, randomized control trials, multi-center cohort studies, and case-control studies from PubMed written in English from the last five years. ICIs have significant benefits in the overall survival compared with traditional chemotherapy. Patients with a higher level of PDL1 expression and high tumor mutational burden (TMB) have a higher response rate, and those with EGFR-/ALK- were better than those with EGFR+/ALK+. The patient who responded to immunotherapy completely can still maintain the efficacy after two years of treatment. Neoadjuvant immunotherapy in patients with resectable non-small cell lung cancer resulted in a 45% major pathology response (MPR) and 40% downstaging. Combined therapy (ICIs + chemotherapy) was better than chemotherapy alone, irrespective of PD-L1 expression. A combination of ICIs such as CTLA-4 and PD-1/PD-L1 improved PFS as well. Radiochemotherapy ahead of ICIs is promising as well. However, ICIs combined with EGFR/ALK-TKI (tyrosine kinase inhibitor) are not suggested for the time being. PDL1 expression, TMB, and EGFR/ALK mutations are promising predictive biomarkers. Gut microbiota, galectin-3, and intensity of CD8 cell infiltration are other potential predictive biomarkers. These are very important in the future management of lung cancers as they can prevent unnecessary toxicities and cost of treatment.
Collapse
Affiliation(s)
- Tun Zan Maung
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Huseyin Ekin Ergin
- Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Mehwish Javed
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Evelyn E Inga
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA.,Internal Medicine, LaSante Health Center, Brooklyn, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
11
|
Castello A, Toschi L, Rossi S, Mazziotti E, Lopci E. The immune-metabolic-prognostic index and clinical outcomes in patients with non-small cell lung carcinoma under checkpoint inhibitors. J Cancer Res Clin Oncol 2020; 146:1235-1243. [PMID: 32048008 DOI: 10.1007/s00432-020-03150-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/05/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE This prospective study evaluated whether peripheral blood biomarkers and metabolic parameters on F-18 fludeoxyglucose positron emission tomography/computed tomography (F-18 FDG PET/CT) could be associated with clinical outcome in non-small cell lung carcinoma (NSCLC) patients treated with immune checkpoint inhibitors (ICI). METHODS Data from 33 patients with NSCLC and treated with ICI were collected. Complete blood cell counts before and at the first restaging were measured. All patients underwent F-18 FDG PET/CT at baseline, while 25 patients at the first restaging. Progression-free survival (PFS) and overall survival (OS) were determined and compared using the Kaplan-Meier and the log-rank test. The median follow-up was 11.3 months (range 1-17 months). RESULTS Multivariate analyses demonstrated that low neutrophil-to-lymphocyte ratio (NLR < 4.9) and low total lesion glycolysis (TLG < 541.5 ml) at the first restaging were significantly associated with PFS (both p = 0.019) and OS (p = 0.001 and p = 0.048, respectively). An immune-metabolic-prognostic index (IMPI), based on post-NLR and post-TLG was developed, categorizing 3 groups: high risk, 2 factors; intermediate risk, 1 factor; low risk, 0 factors. Median PFS for low, intermediate and high risk was 7.8 months (95% CI 4.6-11.0), 5.6 months (95% CI 3.8-7.4), and 1.8 months (95% CI 1.6-2.0) (p < 0.001) respectively. Likewise, median OS was 15.2 months (95% CI 10.9-19.6), 13.2 months (95% CI 5.9-20.3), and 2.8 months (95% CI 1.4-4.2) (p < 0.001), respectively. CONCLUSION IMPI at the first restaging, combining both inflammatory and metabolic biomarkers, was correlated with PFS and OS. IMPI can be a potentially valuable tool for identifying NSCLC patients who are likely to benefit from ICI.
Collapse
Affiliation(s)
- Angelo Castello
- Department of Nuclear Medicine, Humanitas Clinical and Research Center-IRCCS, CAP, Via Manzoni 56, 20089, Rozzano, MI, Italy
| | - Luca Toschi
- Department of Oncology and Hematology, Humanitas Clinical and Research Center-IRCCS, Rozzano, MI, Italy
| | - Sabrina Rossi
- Department of Oncology and Hematology, Humanitas Clinical and Research Center-IRCCS, Rozzano, MI, Italy
| | - Emanuela Mazziotti
- Department of Nuclear Medicine, Humanitas Clinical and Research Center-IRCCS, CAP, Via Manzoni 56, 20089, Rozzano, MI, Italy
| | - Egesta Lopci
- Department of Nuclear Medicine, Humanitas Clinical and Research Center-IRCCS, CAP, Via Manzoni 56, 20089, Rozzano, MI, Italy.
| |
Collapse
|
12
|
Decazes P, Bohn P. Immunotherapy by Immune Checkpoint Inhibitors and Nuclear Medicine Imaging: Current and Future Applications. Cancers (Basel) 2020; 12:E371. [PMID: 32041105 PMCID: PMC7072145 DOI: 10.3390/cancers12020371] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/21/2020] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy by using immune checkpoint inhibitors is a revolutionary development in oncology. Medical imaging is also impacted by this new therapy, particularly nuclear medicine imaging (also called radionuclide imaging), which uses radioactive tracers to visualize metabolic functions. Our aim was to review the current applications of nuclear medicine imaging in immunotherapy, along with their limitations, and the perspectives offered by this imaging modality. Method: Articles describing the use of radionuclide imaging in immunotherapy were researched using PubMed by April 2019 and analyzed. Results: More than 5000 articles were analyzed, and nearly 100 of them were retained. Radionuclide imaging, notably 18F-FDG PET/CT, already has a major role in many cancers for pre-therapeutic and therapeutic evaluation, diagnoses of adverse effects, called immune-related adverse events (IrAE), and end-of-treatment evaluations. However, these current applications can be hindered by immunotherapy, notably due to atypical response patterns such as pseudoprogression, which is defined as an increase in the size of lesions, or the visualization of new lesions, followed by a response, and hyperprogression, which is an accelerated tumor growth rate after starting treatment. To overcome these difficulties, new opportunities are offered, particularly therapeutic evaluation criteria adapted to immunotherapy and immuno-PET allowing us to predict responses to immunotherapy. Moreover, some new technological solutions are also promising, such as radiomic analyses and body composition on associated anatomical images. However, more research has to be done, notably for the diagnosis of hyperprogression and pseudoprogression. Conclusion: Immunotherapy, by its major impact on cancer and by the new patterns generated on images, is revolutionary in the field of medical images. Nuclear medicine imaging is already established and will be able to help meet new challenges through its plasticity.
Collapse
Affiliation(s)
- Pierre Decazes
- Department of Nuclear Medicine, Henri Becquerel Cancer Center, 76000 Rouen, France;
- LITIS-QuantIF-EA (Equipe d’Accueil) 4108, IRIB, Faculty of Medicine, University of Rouen, 76000 Rouen, France
| | - Pierre Bohn
- Department of Nuclear Medicine, Henri Becquerel Cancer Center, 76000 Rouen, France;
- LITIS-QuantIF-EA (Equipe d’Accueil) 4108, IRIB, Faculty of Medicine, University of Rouen, 76000 Rouen, France
| |
Collapse
|
13
|
Telo S, Calderoni L, Vichi S, Zagni F, Castellucci P, Fanti S. Alternative and New Radiopharmaceutical Agents for Lung Cancer. Curr Radiopharm 2020; 13:185-194. [PMID: 31868150 PMCID: PMC8206190 DOI: 10.2174/1874471013666191223151402] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/27/2019] [Accepted: 11/11/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND FDG PET/CT imaging has an established role in lung cancer (LC) management. Whilst it is a sensitive technique, FDG PET/CT has a limited specificity in the differentiation between LC and benign conditions and is not capable of defining LC heterogeneity since FDG uptake varies between histotypes. OBJECTIVE To get an overview of new radiopharmaceuticals for the study of cancer biology features beyond glucose metabolism in LC. METHODS A comprehensive literature review of PubMed/Medline was performed using a combination of the following keywords: "positron emission tomography", "lung neoplasms", "non-FDG", "radiopharmaceuticals", "tracers". RESULTS Evidences suggest that proliferation markers, such as 18F-Fluorothymidine and 11CMethionine, improve LC staging and are useful in evaluating treatment response and progression free survival. 68Ga-DOTA-peptides are already routinely used in pulmonary neuroendocrine neoplasms (NENs) management and should be firstly performed in suspected NENs. 18F-Fluoromisonidazole and other radiopharmaceuticals show a promising impact on staging, prognosis assessment and therapy response in LC patients, by visualizing hypoxia and perfusion. Radiolabeled RGD-peptides, targeting angiogenesis, may have a role in LC staging, treatment outcome and therapy. PET radiopharmaceuticals tracing a specific oncogene/signal pathway, such as EGFR or ALK, are gaining interest especially for therapeutic implications. Other PET tracers, like 68Ga-PSMA-peptides or radiolabeled FAPIs, need more development in LC, though, they are promising for therapy purposes. CONCLUSION To date, the employment of most of the described tracers is limited to the experimental field, however, research development may offer innovative opportunities to improve LC staging, characterization, stratification and response assessment in an era of increased personalized therapy.
Collapse
Affiliation(s)
- Silvi Telo
- Address correspondence to this author at the Department of Metropolitan Nuclear Medicine, University of Bologna, Bologna, Italy; Tel/Fax: +390512143959; E-mail:
| | | | | | | | | | | |
Collapse
|
14
|
Castello A, Rossi S, Lopci E. 18F-FDG PET/CT in Restaging and Evaluation of Response to Therapy in Lung Cancer: State of the Art. Curr Radiopharm 2019; 13:228-237. [PMID: 31886757 PMCID: PMC8493792 DOI: 10.2174/1874471013666191230144821] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 01/25/2019] [Accepted: 11/11/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Metabolic information provided by 18F-FDG PET/CT are useful for initial staging, therapy planning, response evaluation, and to a lesser extent for the follow-up of non-small cell lung cancer (NSCLC). To date, there are no established clinical guidelines in treatment response and early detection of recurrence. OBJECTIVE To provide an overview of 18F-FDG PET/CT in NSCLC and in particular, to discuss its utility in treatment response evaluation and restaging of lung cancer. METHODS A comprehensive search was used based on PubMed results. From all studies published in English those that explored the role of 18F-FDG PET/CT in the treatment response scenario were selected. RESULTS Several studies have demonstrated that modifications in metabolic activity, expressed by changes in SUV both in the primary tumor as well as in regional lymph nodes, are associated with tumor response and survival. Beside SUV, other metabolic parameters (i.e. MTV, TLG, and percentage changes) are emerging to be helpful for predicting clinical outcomes. CONCLUSION 18F-FDG parameters appear to be promising factors for evaluating treatment response and for detecting recurrences, although larger prospective trials are needed to confirm these evidences and to determine optimal cut-off values.
Collapse
Affiliation(s)
- Angelo Castello
- Nuclear Medicine, Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Sabrina Rossi
- Medical Oncology, Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Egesta Lopci
- Nuclear Medicine, Humanitas Clinical and Research Hospital, Rozzano, Italy
| |
Collapse
|
15
|
Assessing the interactions between radiotherapy and antitumour immunity. Nat Rev Clin Oncol 2019; 16:729-745. [PMID: 31243334 DOI: 10.1038/s41571-019-0238-9] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2019] [Indexed: 12/17/2022]
Abstract
Immunotherapy, specifically the introduction of immune checkpoint inhibitors, has transformed the treatment of cancer, enabling long-term tumour control even in individuals with advanced-stage disease. Unfortunately, only a small subset of patients show a response to currently available immunotherapies. Despite a growing consensus that combining immune checkpoint inhibitors with radiotherapy can increase response rates, this approach might be limited by the development of persistent radiation-induced immunosuppression. The ultimate goal of combining immunotherapy with radiotherapy is to induce a shift from an ineffective, pre-existing immune response to a long-lasting, therapy-induced immune response at all sites of disease. To achieve this goal and enable the adaptation and monitoring of individualized treatment approaches, assessment of the dynamic changes in the immune system at the patient level is essential. In this Review, we summarize the available clinical data, including forthcoming methods to assess the immune response to radiotherapy at the patient level, ranging from serum biomarkers to imaging techniques that enable investigation of immune cell dynamics in patients. Furthermore, we discuss modelling approaches that have been developed to predict the interaction of immunotherapy with radiotherapy, and highlight how they could be combined with biomarkers of antitumour immunity to optimize radiotherapy regimens and maximize their synergy with immunotherapy.
Collapse
|
16
|
Grizzi F, Castello A, Qehajaj D, Toschi L, Rossi S, Pistillo D, Paleari V, Veronesi G, Novellis P, Monterisi S, Mineri R, Rahal D, Lopci E. Independent expression of circulating and tissue levels of PD-L1: correlation of clusters with tumor metabolism and outcome in patients with non-small cell lung cancer. Cancer Immunol Immunother 2019; 68:1537-1545. [PMID: 31482306 PMCID: PMC11028209 DOI: 10.1007/s00262-019-02387-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 08/27/2019] [Indexed: 12/29/2022]
Abstract
PURPOSE To evaluate the clinical-pathological and prognostic significance of the circulating PD-L1 level in patients with surgically treated NSCLC, by combining data for PD-L1 expression with other immune-related markers and tumor metabolism. METHODS Overall, 40 patients with resected NSCLC (stage Ia-IIIa) who had preoperative blood storage and underwent staging PET/CT were enrolled for the study. In all cases, we determined plasma levels of PD-L1 (pg/ml), immune-reactive areas (IRA %) covered by CD3, CD68, CD20, CD8, PD-1, and PD-L1 in the tumor specimen, and metabolic parameters on PET, i.e., SUVmax, SUVpeak, metabolic tumor volume (MTV), and total lesion glycolysis (TLG). Variables were statistically analyzed to establish their association with disease-free survival (DFS). RESULTS The circulating levels of PD-L1 in the bloodstream could be determined in 38/40 (95%) samples. The mean and median expression levels were 34.86 pg/ml and 24.83 pg/ml, respectively. We did not find any statistically significant correlation between circulating PD-L1 and tissue expression of PD-L1/PD-1. Some mild degree of positive correlation was determined between tissue PD-L1 and SUVmax (ρ = 0.390; p = 0.0148). Hierarchical clustering combining circulating, tissue, and metabolic parameters identified clusters with high metabolic tumor burden or high expression of plasma PD-L1 levels (Z score ≥ 2) as having a poor DFS (p = 0.033). The multivariate analysis detected stage and metabolism (i.e., SUVmax and SUVpeak) as independent prognostic factors for DFS. CONCLUSION Plasma levels of PD-L1 are independent of the expression of PD-1/PD-L1 in NSCLC tumor tissue and, when combined with other clinical-pathological parameters, allow for the identification of clusters with different outcomes.
Collapse
Affiliation(s)
- Fabio Grizzi
- Immunology and Inflammation, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, Italy
| | - Angelo Castello
- Nuclear Medicine Department, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, Italy
| | - Dorina Qehajaj
- Immunology and Inflammation, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, Italy
| | - Luca Toschi
- Medical Oncology, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, MI, Italy
| | - Sabrina Rossi
- Medical Oncology, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, MI, Italy
| | - Daniela Pistillo
- Biobank, Humanitas Cancer Center, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Valentina Paleari
- Biobank, Humanitas Cancer Center, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Giulia Veronesi
- Thoracic Surgery, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, Italy
| | - Pierluigi Novellis
- Thoracic Surgery, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, Italy
| | - Simona Monterisi
- Thoracic Surgery, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, Italy
| | - Rossana Mineri
- Molecular Biology Section, Clinical Investigation Laboratory, Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, 20089, Rozzano, Italy
| | - Daoud Rahal
- Pathology, Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, 20089, Rozzano, Italy
| | - Egesta Lopci
- Nuclear Medicine Department, Humanitas Clinical and Research Hospital, IRCCS, Via Manzoni 56, 20089, Rozzano, Italy.
| |
Collapse
|
17
|
Cui J, Yin Z, Liu G, Chen X, Gao X, Lu H, Li W, Lin D. Activating transcription factor 1 promoted migration and invasion in lung cancer cells through regulating EGFR and MMP-2. Mol Carcinog 2019; 58:1919-1924. [PMID: 31420907 DOI: 10.1002/mc.23086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/26/2019] [Accepted: 07/04/2019] [Indexed: 12/28/2022]
Abstract
Lung cancer is among the most frequently occurring cancers and the leading cause of cancer-related deaths worldwide. Nonsmall cell lung cancer is accountable for 85% to 90% of all lung cancer cases and develops distant metastases with high mortality. In this work, we elucidated the role of activating transcription factor 1 (ATF1) in migration and invasion of lung cancer cells. We found that the migration and invasion were inhibited with ATF1 silencing in lung cancer cells. By contrast, ATF1 overexpression led to promotion in migration and invasion. The alteration in ATF1 expression induced a change in the epidermal growth factor receptor (EGFR) and matrix metalloproteinases (MMP)-2 expression level in the same tendency. Thus, we provided a potential new candidate for therapies against lung cancer, showing the possible mechanism underlying the invasion and migration of lung cancer cells.
Collapse
Affiliation(s)
- Jinggang Cui
- Department of Respiratory Medicine, Suzhou Municipal Hospital, Suzhou Hospital Affiliated Nanjing Medical University, Suzhou, Jiangsu, China
| | - Zhaofang Yin
- Department of General Medicine, Suzhou Municipal Hospital, Suzhou Hospital Affiliated Nanjing Medical University, Suzhou, Jiangsu, China
| | - Guohua Liu
- Department of Respiratory Medicine, Suzhou Municipal Hospital, Suzhou Hospital Affiliated Nanjing Medical University, Suzhou, Jiangsu, China
| | - Xiaojun Chen
- Department of Respiratory Medicine, Suzhou Municipal Hospital, Suzhou Hospital Affiliated Nanjing Medical University, Suzhou, Jiangsu, China
| | - Xiaolai Gao
- Department of Respiratory Medicine, Suzhou Municipal Hospital, Suzhou Hospital Affiliated Nanjing Medical University, Suzhou, Jiangsu, China
| | - Huiling Lu
- Department of Respiratory Medicine, Suzhou Municipal Hospital, Suzhou Hospital Affiliated Nanjing Medical University, Suzhou, Jiangsu, China
| | - Wei Li
- Department of Respiratory Medicine, Suzhou Municipal Hospital, Suzhou Hospital Affiliated Nanjing Medical University, Suzhou, Jiangsu, China
| | - Dang Lin
- Department of Respiratory Medicine, Suzhou Municipal Hospital, Suzhou Hospital Affiliated Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
18
|
Alama A, Coco S, Genova C, Rossi G, Fontana V, Tagliamento M, Giovanna Dal Bello M, Rosa A, Boccardo S, Rijavec E, Biello F, Longo L, Cavalieri Z, Bruzzo C, Grossi F. Prognostic Relevance of Circulating Tumor Cells and Circulating Cell-Free DNA Association in Metastatic Non-Small Cell Lung Cancer Treated with Nivolumab. J Clin Med 2019; 8:jcm8071011. [PMID: 31295929 PMCID: PMC6679117 DOI: 10.3390/jcm8071011] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/18/2022] Open
Abstract
The treatment of advanced non-small cell lung cancer (NSCLC) has been revolutionized by immune checkpoint inhibitors (ICIs). The identification of prognostic and predictive factors in ICIs-treated patients is presently challenging. Circulating tumor cells (CTCs) and cell-free DNA (cfDNA) were evaluated in 89 previously treated NSCLC patients receiving nivolumab. Blood samples were collected before therapy and at the first and second radiological response assessments. CTCs were isolated by a filtration-based method. cfDNA was extracted from plasma and estimated by quantitative PCR. Patients with baseline CTC number and cfDNA below their median values (2 and 836.5 ng from 3 mL of blood and plasma, respectively) survived significantly longer than those with higher values (p = 0.05 and p = 0.04, respectively). The two biomarkers were then used separately and jointly as time-dependent covariates in a regression model confirming their prognostic role. Additionally, a four-fold risk of death for the subgroup presenting both circulating biomarkers above the median values was observed (p < 0.001). No significant differences were found between circulating biomarkers and best response. However, progressing patients with concomitant lower CTCs and cfDNA performed clinically well (p = 0.007), suggesting that jointed CTCs and cfDNA might help discriminate a low-risk population which might benefit from continuing ICIs beyond progression.
Collapse
Affiliation(s)
- Angela Alama
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy.
| | - Simona Coco
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Carlo Genova
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Giovanni Rossi
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Vincenzo Fontana
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Marco Tagliamento
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Maria Giovanna Dal Bello
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Alessandra Rosa
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Simona Boccardo
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Erika Rijavec
- Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza, 28, 20122 Milan, Italy
| | - Federica Biello
- AOU Maggiore della Carità, Corso Mazzini 18, 28100 Novara, Italy
| | - Luca Longo
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Zita Cavalieri
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Cristina Bruzzo
- Lung Cancer Unit, Division of Medical Oncology II, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Francesco Grossi
- Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza, 28, 20122 Milan, Italy
| |
Collapse
|
19
|
Castello A, Lopci E. Response assessment of bone metastatic disease: seeing the forest for the trees RECIST, PERCIST, iRECIST, and PCWG-2. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2019; 63:150-158. [PMID: 31286751 DOI: 10.23736/s1824-4785.19.03193-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tumor response is often used as a surrogate marker for survival practically in all clinical trials. Therefore, robust and valid response criteria during the course of trials are fundamental for the assessment of response to therapy. This aspect, however, becomes particularly challenging when it comes to bone metastases. In the era of targeted therapies and immune-checkpoint inhibitors (ICI), response assessment by morphologic-based criteria cannot detect the real tumor response and, consequently, fail to demonstrate the actual clinical benefit. This review will focus on some of the most common morphologic and metabolic response criteria and their application for bone lesions, highlighting relative strengths and weaknesses as well as potential future methods in the era of target therapies and immunotherapy with ICI.
Collapse
Affiliation(s)
- Angelo Castello
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital, IRCCS, Rozzano, Milan, Italy
| | - Egesta Lopci
- Nuclear Medicine Unit, Humanitas Clinical and Research Hospital, IRCCS, Rozzano, Milan, Italy -
| |
Collapse
|