1
|
Long-Mira E, Bontoux C, Rignol G, Hofman V, Lassalle S, Benzaquen J, Boutros J, Lalvée-Moret S, Zahaf K, Lespinet-Fabre V, Bordone O, Maistre S, Bonnetaud C, Cohen C, Berthet JP, Marquette CH, Vouret-Craviari V, Ilié M, Hofman P. Exploring the Expression of CD73 in Lung Adenocarcinoma with EGFR Genomic Alterations. Cancers (Basel) 2025; 17:1034. [PMID: 40149368 PMCID: PMC11941413 DOI: 10.3390/cancers17061034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Immune checkpoint inhibitors (ICIs) benefit some lung cancer patients, but their efficacy is limited in advanced lung adenocarcinoma (LUAD) with EGFR mutations (EGFRm), largely due to a non-immunogenic tumour microenvironment (TME). Furthermore, EGFRm LUAD patients often experience increased toxicity with ICIs. CD73, an ectonucleotidase involved in adenosine production, promotes tumour immune evasion and could represent a novel therapeutic target. This study investigates CD73 expression in LUAD with EGFR alterations and its clinico-pathological correlations. METHODS CD73 expression in tumour (CD73TC) and stromal (CD73SC) cells was assessed in 76 treatment-naive LUAD patients using immunohistochemistry (IHC) (D7F9A clone) alongside IHC PD-L1 (22C3 clone). EGFR alterations were identified by molecular sequencing and FISH. Event-free survival (EFS) was analysed based on CD73TC expression. RESULTS CD73TC expression was observed in 66% of cases, with high expression (Tumour Proportion Score > 50%) correlating with improved EFS (p = 0.045). CD73TC and PD-L1 expression were not significantly correlated (p = 0.44), although a weak inverse trend was observed. CD73SC expression was detected in 18% of cases, predominantly in early-stage (p = 0.037), PD-L1-negative (p = 0.030), and non-EGFR-amplified (p = 0.0018) tumours. No significant associations were found with disease stage, histological subtype, EGFR mutation type, and amplification. CONCLUSIONS CD73 expression in EGFRm LUAD is heterogeneous and associated with diverse TME profiles. These findings support the potential of CD73 as a predictive biomarker and therapeutic target, highlighting its clinical relevance in EGFRm LUAD.
Collapse
Affiliation(s)
- Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Guylène Rignol
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Jonathan Benzaquen
- Department of Thoracic Oncology, IHU RespirERA Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (J.B.); (J.B.); (C.-H.M.)
| | - Jacques Boutros
- Department of Thoracic Oncology, IHU RespirERA Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (J.B.); (J.B.); (C.-H.M.)
| | - Salomé Lalvée-Moret
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Katia Zahaf
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Olivier Bordone
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Sophia Maistre
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Christelle Bonnetaud
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
| | - Charlotte Cohen
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (C.C.); (J.-P.B.)
| | - Jean-Philippe Berthet
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (C.C.); (J.-P.B.)
| | - Charles-Hugo Marquette
- Department of Thoracic Oncology, IHU RespirERA Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06100 Nice, France; (J.B.); (J.B.); (C.-H.M.)
| | - Valerie Vouret-Craviari
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, Biobank Côte d’Azur BB-0033-00025, FHU OncoAge, Centre Hospitalier Universitaire de Nice, 06000 Nice, France; (E.L.-M.); (C.B.); (G.R.); (V.H.); (S.L.); (M.I.)
- Institute for Research on Cancer and Aging, Team 4, Inserm U1081, CNRS UMR 7413, Université Côte d’Azur, 06000 Nice, France;
| |
Collapse
|
2
|
Doyen J, Dompmartin A, Cruzel C, Stefan D, Arnault JP, Coutte A, Picard-Gauci A, Mansard S, Gleyzolle B, Fontas E, Long-Mira E, Mirabel X, Mortier L, Montaudié H. Nivolumab and hypofractionated radiotherapy in patients with advanced melanoma: A phase 2 trial. Eur J Cancer 2025; 217:115256. [PMID: 39864364 DOI: 10.1016/j.ejca.2025.115256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/22/2024] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Radiotherapy is thought to enhance anti-tumor immunity, particularly when delivered in a hypofractionated and multisite manner. Therefore, we investigated the effects of combining radiotherapy with nivolumab in patients with advanced melanoma. METHODS This was a multicenter, non-randomized, phase 2 trial that enrolled patients with treatment-naïve metastatic melanoma. They received nivolumab (240 mg / 2 weeks) plus radiotherapy (day 15, 6 Gy × 3). When feasible, one target from each organ was irradiated (no irradiation of all targets). The primary endpoint was 1-year overall survival (OS). RESULTS This trial included 64 patients between March 2017 and July 2019. The median follow-up was 23.5 (2.3-43.8) months. The median age was 68 (35-95) years, patients were mostly male (67 %) with an Eastern Cooperative Oncology Group Performance Status (ECOG-PS) score of 0 (72 %), stage IV-M1c disease (47 %), and were BRAF-wild-type (67 %). The 2-year OS and 1-year PFS rates were 65.2 % and 56 %, respectively (P = 0.22 and P = 0.03, vs. 58 % and 43 %, respectively, in the Checkmate 066 study). Thirty-seven (58 %) and twenty-seven (42 %) patients were irradiated at one and multiple targets, respectively. The ECOG-PS (1 vs. 0; HR = 3.5; P = 0.005) was an independent prognostic factor for OS. Irradiating more than one site and irradiating a smaller cumulative tumor volume tended to correlate with better outcome. Grade 3-4 treatment-related adverse events occurred in 21.9 % of the patients (no grade 5). CONCLUSIONS Combined immunotherapy and hypofractionated radiotherapy did not improve survival compared to historical cohorts. The radiotherapy schedule needs to be optimized in order to improve these results.
Collapse
Affiliation(s)
- Jérôme Doyen
- Department of Radiation Oncology, Côte d'Azur University, Antoine Lacassagne Cancer Center, Nice, France.
| | - Anne Dompmartin
- Department of Dermatology, University Hospital Center of Caen, France
| | - Coralie Cruzel
- Department of Clinical Research and Innovation, Côte d'Azur University, University Hospital Center of Nice, France
| | - Dinu Stefan
- Department of Radiation Oncology, François-Baclesse Cancer Center, Caen, France
| | | | - Alexandre Coutte
- Department of Radiation Oncology, University Hospital Center of Amiens, France
| | - Alexandra Picard-Gauci
- Department of Dermatology, Côte d'Azur University, University Hospital Center of Nice, France
| | - Sandrine Mansard
- Department of Dermatology, University Hospital Center of Clermont-Ferrand, France
| | - Baptiste Gleyzolle
- Department of Radiation Oncology, Jean-Perrin Cancer Center, Clermont-Ferrand, France
| | - Eric Fontas
- Department of Clinical Research and Innovation, Côte d'Azur University, University Hospital Center of Nice, France
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, Biobank Côte d'Azur BB-0033-00025, Côte d'Azur University, University Hospital Center of Nice, France
| | - Xavier Mirabel
- Department of Radiation Oncology, Oscar-Lambret Cancer Center, Lille, France
| | - Laurent Mortier
- Department of Dermatology, University Hospital Center of Lille, France
| | - Henri Montaudié
- Department of Dermatology, Côte d'Azur University, University Hospital Center of Nice, France
| |
Collapse
|
3
|
Hofman P, Ourailidis I, Romanovsky E, Ilié M, Budczies J, Stenzinger A. Artificial intelligence for diagnosis and predictive biomarkers in Non-Small cell lung cancer Patients: New promises but also new hurdles for the pathologist. Lung Cancer 2025; 200:108110. [PMID: 39879785 DOI: 10.1016/j.lungcan.2025.108110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/09/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
The rapid development of artificial intelligence (AI) based tools in pathology laboratories has brought forward unlimited opportunities for pathologists. Promising AI applications used for accomplishing diagnostic, prognostic and predictive tasks are being developed at a high pace. This is notably true in thoracic oncology, given the significant and rapid therapeutic progress made recently for lung cancer patients. Advances have been based on drugs targeting molecular alterations, immunotherapies, and, more recently antibody-drug conjugates which are soon to be introduced. For over a decade, many proof-of-concept studies have explored the use of AI algorithms in thoracic oncology to improve lung cancer patient care. However, despite the enthusiasm in this domain, the set-up and use of AI algorithms in daily practice of thoracic pathologists has not been operative until now, due to several constraints. The purpose of this review is to describe the potential but also the current barriers of AI applications in routine thoracic pathology for non-small cell lung cancer patient care and to suggest practical solutions for rapid future implementation.
Collapse
Affiliation(s)
- Paul Hofman
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, FHU OncoAge, Biobank BB-0033-00025, IRCAN, Côte d'Azur University, 30 avenue de la voie romaine 06002 Nice cedex 01, France.
| | - Iordanis Ourailidis
- Institute of Pathology Heidelberg, University Hospital Heidelberg, In Neuenheimer Feld 224 69120 Heidelberg, Germany
| | - Eva Romanovsky
- Institute of Pathology Heidelberg, University Hospital Heidelberg, In Neuenheimer Feld 224 69120 Heidelberg, Germany
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, FHU OncoAge, Biobank BB-0033-00025, IRCAN, Côte d'Azur University, 30 avenue de la voie romaine 06002 Nice cedex 01, France
| | - Jan Budczies
- Institute of Pathology Heidelberg, University Hospital Heidelberg, In Neuenheimer Feld 224 69120 Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology Heidelberg, University Hospital Heidelberg, In Neuenheimer Feld 224 69120 Heidelberg, Germany
| |
Collapse
|
4
|
Pal R, Krishnamoorthy M, Matsui A, Kang H, Morita S, Taniguchi H, Kobayashi T, Morita A, Choi HS, Duda DG, Kumar AT. Fluorescence Lifetime Imaging Enables In Vivo Quantification of PD-L1 Expression and Intertumoral Heterogeneity. Cancer Res 2025; 85:618-632. [PMID: 39514403 PMCID: PMC11967911 DOI: 10.1158/0008-5472.can-24-0880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/26/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Patient selection for cancer immunotherapy requires precise, quantitative readouts of biomarker expression in intact tumors that can be reliably compared across multiple subjects over time. The current clinical standard biomarker for assessing immunotherapy response is PD-L1 expression, typically quantified using IHC. This method, however, only provides snapshots of PD-L1 expression status in microscopic regions of ex vivo specimens. Although various targeted probes have been investigated for in vivo imaging of PD-L1, nonspecific probe accumulation within the tumor microenvironment has hindered accurate quantification, limiting the utility for preclinical and clinical studies. Here, we demonstrated that in vivo time-domain fluorescence imaging of an anti-PD-L1 antibody tagged with the near-infrared fluorophore IRDye 800CW (αPDL1-800) can yield quantitative estimates of baseline tumor PD-L1 heterogeneity across untreated mice, as well as variations in PD-L1 expression in mice undergoing clinically relevant anti-PD-1 treatment. The fluorescence lifetime (FLT) of PD-L1-bound αPDL1-800 was significantly longer than the FLT of nonspecifically accumulated αPDL1-800 in the tumor microenvironment. This FLT contrast allowed quantification of PD-L1 expression across mice both in superficial breast tumors using planar FLT imaging and in deep-seated liver tumors (>5 mm depth) using the asymptotic time-domain algorithm for fluorescence tomography. These findings suggest that FLT imaging can accelerate the preclinical investigation and clinical translation of new immunotherapy treatments by enabling robust quantification of receptor expression across subjects. Significance: Fluorescence lifetime imaging can quantify PD-L1 expression across multiple mice undergoing anti-PD-1 treatment, providing a critically needed noninvasive imaging method to quantify immunotherapy targets in vivo.
Collapse
Affiliation(s)
- Rahul Pal
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Murali Krishnamoorthy
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Aya Matsui
- Department of Vascular Physiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Homan Kang
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Satoru Morita
- Department of Radiation Oncology, E. L. Steele Laboratories for Tumor Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hajime Taniguchi
- Department of Radiation Oncology, E. L. Steele Laboratories for Tumor Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Tohoku Graduate School of Medicine, Sendai, Japan
| | - Tatsuya Kobayashi
- Department of Radiation Oncology, E. L. Steele Laboratories for Tumor Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Atsuyo Morita
- Department of Radiation Oncology, E. L. Steele Laboratories for Tumor Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dan G. Duda
- Department of Radiation Oncology, E. L. Steele Laboratories for Tumor Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anand T.N. Kumar
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
5
|
Wellhausen J, Röhl L, Berszin M, Krücken I, Zebralla V, Pirlich M, Stoehr M, Wiegand S, Dietz A, Wald T, Wichmann G. Suppression of MCP-1, IFN-γ and IL-6 production of HNSCC ex vivo by pembrolizumab added to docetaxel and cisplatin (TP) exceeding those of TP alone is linked to improved survival. Front Immunol 2025; 15:1473897. [PMID: 39882242 PMCID: PMC11774711 DOI: 10.3389/fimmu.2024.1473897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Background Adding pembrolizumab, an anti-PD-1 antibody approved for treatment of head and neck squamous cell carcinoma (HNSCC) to neoadjuvant (induction-) chemotherapy utilizing docetaxel and cisplatin (TP) followed by radiotherapy may improve outcome in larynx organ-preservation (LOP) that is investigated in the European Larynx-Organ preservation Study (ELOS). As biomarkers for response to TP and pembrolizumab +TP are missing but may include cytokines, this work aims on determining cytokines potentially linked to outcome as prognostic markers sufficient to predict and/or monitor response to successful LOP. Methods Collagenase IV digests were generated from 47 histopathological confirmed HNSCC tumor samples and seeded in 96-well plates containing pembrolizumab, docetaxel, cisplatin either solely or in binary or ternary combination. According to the FLAVINO protocol, supernatants were collected after 3 days, adherent cells fixed using ethanol, air-dried and pan-cytokeratin positive epithelial cells counted using fluorescence microscopy. The cytokines IL-6, IL-8, IFN-γ, IP-10, MCP-1, TNF-α, and VEGF in the supernatant were quantified by sandwich ELISA. Results The mode of interaction between pembrolizumab and TP was assessed and correlated to outcome (overall, disease-specific and progression-free survival of patients). Suppression of MCP-1, IFN-γ and IL-6 production by pembrolizumab + TP exceeding the suppressive effect of TP was detected in the majority of samples and linked to improved survival. Multivariate Cox proportional hazard regression modeling revealed MCP-1, IFN-γ and IL-6 as independent outcome predictors. Conclusions Comparing response to TP vs. pembrolizumab vs. TP + pembrolizumab may allow for identification of patients with superior outcome independent from treatment applied.
Collapse
Affiliation(s)
- Jana Wellhausen
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Louisa Röhl
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Michael Berszin
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Irene Krücken
- Institute of Pathology, University Hospital Leipzig, Leipzig, Germany
- The Comprehensive Cancer Center Central Germany, Leipzig University Hospital, Leipzig, Germany
| | - Veit Zebralla
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
- The Comprehensive Cancer Center Central Germany, Leipzig University Hospital, Leipzig, Germany
| | - Markus Pirlich
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
- The Comprehensive Cancer Center Central Germany, Leipzig University Hospital, Leipzig, Germany
| | - Matthaeus Stoehr
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
- The Comprehensive Cancer Center Central Germany, Leipzig University Hospital, Leipzig, Germany
| | - Susanne Wiegand
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
- The Comprehensive Cancer Center Central Germany, Leipzig University Hospital, Leipzig, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Andreas Dietz
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
- The Comprehensive Cancer Center Central Germany, Leipzig University Hospital, Leipzig, Germany
| | - Theresa Wald
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
- The Comprehensive Cancer Center Central Germany, Leipzig University Hospital, Leipzig, Germany
| | - Gunnar Wichmann
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
- The Comprehensive Cancer Center Central Germany, Leipzig University Hospital, Leipzig, Germany
| |
Collapse
|
6
|
Kwon YH, Park S, Jiang H, Gurudatt NG, Lee K, Jeong H, Nie C, Shin J, Hyun KA, Jung HI. High-resolution spiral microfluidic channel integrated electrochemical device for isolation and detection of extracellular vesicles without lipoprotein contamination. Biosens Bioelectron 2025; 267:116792. [PMID: 39307033 DOI: 10.1016/j.bios.2024.116792] [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: 04/22/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 11/08/2024]
Abstract
Recent studies have indicated significant correlation between the concentration of immune checkpoint markers borne by extracellular vesicles (EVs) and the efficacy of immunotherapy. This study introduces a high-resolution spiral microfluidic channel-integrated electrochemical device (HiMEc), which is designed to isolate and detect EVs carrying the immune checkpoint markers programmed death ligand 1 (PD-L1) and programmed death protein 1 (PD-1), devoid of plasma-abundant lipoprotein contamination. Antigen-antibody reactions were applied to immobilize the lipoproteins on bead surfaces within the plasma, establishing a size differential with EVs. A plasma sample was then introduced into the spiral microfluidic channel, which facilitated the acquisition of nanometer-sized EVs and the elimination of micrometer-sized lipoprotein-bead complexes, along with the isolation and quantification of EVs using HiMEc. PD-L1 and PD-1 expression on EVs was evaluated in 30 plasma samples (10 from healthy donors, 20 from lung cancer patients) using HiMEc and compared to the results obtained from standard tissue-based PD-L1 testing, noting that HiMEc could be utilized to select further potential candidates. The obtained results are expected to contribute positively to the clinical assessment of potential immunotherapy beneficiaries.
Collapse
Affiliation(s)
- Yong-Hyun Kwon
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sunyoung Park
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; The DABOM Inc., Seoul, Republic of Korea
| | - Hairi Jiang
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - N G Gurudatt
- Department of Electrical and Computer Engineering, North Carolina State University, Raleigh, NC, USA
| | - Kyungyeon Lee
- Department of Medical Engineering, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | | | - Cheng Nie
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Joonchul Shin
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyung-A Hyun
- Korea Electronics Technology Institute (KETI), 25, Saenari-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13509, Republic of Korea.
| | - Hyo-Il Jung
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; The DABOM Inc., Seoul, Republic of Korea.
| |
Collapse
|
7
|
Cao XC, Mao XL, Lu SS, Zhu W, Huang W, Yi H, Yuan L, Zhou JH, Xiao ZQ. A PD-L1-Targeted Probe Cy5.5-A11 for In Vivo Imaging of Multiple Tumors. ACS OMEGA 2024; 9:43826-43833. [PMID: 39494025 PMCID: PMC11525735 DOI: 10.1021/acsomega.4c06761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024]
Abstract
PD-L1 is an immune checkpoint molecule mediating cancer immune escape, and its expression level in the tumor has been used as a biomarker to predict response to immune checkpoint inhibitor (ICI) therapy. Our previous study reveals that an 11 amino acid-long ANXA1-derived peptide (named A11) binds and degrades the PD-L1 protein in multiple cancers and is a potential peptide for cancer diagnosis and treatment. Near-infrared fluorescence (NIF) optical imaging of tumors offers a noninvasive method for detecting cancer and monitoring therapeutic responses. In this study, an NIF dye Cy5.5 was conjugated with A11 peptide to develop a novel PD-L1-targeted probe for molecular imaging of tumors and monitor the dynamic changes in PD-L1 expression in tumors. In vitro imaging studies showed that intense fluorescence was observed in triple-negative breast cancer MDA-MB-231, nonsmall cell lung cancer H460, and melanoma A375 cells incubated with Cy5.5-A11, and the cellular uptake of Cy5.5-A11 was efficiently inhibited by coincubation with unlabeled A11 or knockdown of cellular PD-L1 by shRNA. In vivo imaging studies showed accumulation of Cy5.5-A11 in the MDA-MB-231, H460, and A375 xenografts with good contrast from 0.5 to 24 h after intravenous injection, indicating that Cy5.5-A11 possesses the strong ability for in vivo tumor imaging. Moreover, the fluorescent signal of A11-Cy5.5 in the xenografts was successfully blocked by coinjection of unlabeled A11 peptide or knockdown of cellular PD-L1 by shRNA, indicating the specificity of Cy5.5-A11 targeting PD-L1 in tumor imaging. Our data demonstrate that Cy5.5-A11 is a novel tool for tumor imaging of PD-L1, which has the potential for detecting cancer and predicting ICI therapeutic responses.
Collapse
Affiliation(s)
- Xiao-Cheng Cao
- Department
of Pathology, Xiangya Hospital, Central
South University, Changsha 410008, China
- Research
Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China
- The
Higher Educational Key Laboratory for Cancer Proteomics and Translational
Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
- National
Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xue-Li Mao
- Research
Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China
- The
Higher Educational Key Laboratory for Cancer Proteomics and Translational
Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shan-Shan Lu
- Research
Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China
- The
Higher Educational Key Laboratory for Cancer Proteomics and Translational
Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wei Zhu
- Department
of Pathology, Xiangya Hospital, Central
South University, Changsha 410008, China
- National
Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410011, China
| | - Wei Huang
- Research
Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China
- The
Higher Educational Key Laboratory for Cancer Proteomics and Translational
Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Hong Yi
- Research
Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China
- The
Higher Educational Key Laboratory for Cancer Proteomics and Translational
Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Li Yuan
- Department
of Nuclear Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Jian-Hua Zhou
- Department
of Pathology, Xiangya Hospital, Central
South University, Changsha 410008, China
- National
Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410011, China
| | - Zhi-Qiang Xiao
- Department
of Pathology, Xiangya Hospital, Central
South University, Changsha 410008, China
- Research
Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China
- The
Higher Educational Key Laboratory for Cancer Proteomics and Translational
Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, China
- National
Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
8
|
Tárnoki DL, Karlinger K, Ridge CA, Kiss FJ, Györke T, Grabczak EM, Tárnoki ÁD. Lung imaging methods: indications, strengths and limitations. Breathe (Sheff) 2024; 20:230127. [PMID: 39360028 PMCID: PMC11444493 DOI: 10.1183/20734735.0127-2023] [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: 01/15/2024] [Accepted: 06/24/2024] [Indexed: 10/04/2024] Open
Abstract
Imaging methods are fundamental tools to detect and diagnose lung diseases, monitor their treatment and detect possible complications. Each modality, starting from classical chest radiographs and computed tomography, as well as the ever more popular and easily available thoracic ultrasound, magnetic resonance imaging and nuclear medicine methods, and new techniques such as photon counting computed tomography, radiomics and application of artificial intelligence, has its strong and weak points, which we should be familiar with to properly choose between the methods and interpret their results. In this review, we present the indications, strengths and main limitations of methods for chest imaging.
Collapse
Affiliation(s)
- Dávid László Tárnoki
- Medical Imaging Centre, Semmelweis University, Budapest, Hungary
- Oncologic Imaging and Invasive Diagnostic Centre and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Kinga Karlinger
- Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Carole A Ridge
- Department of Radiology, Royal Brompton and Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Fanni Júlia Kiss
- Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Tamás Györke
- Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Elzbieta Magdalena Grabczak
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Ádám Domonkos Tárnoki
- Medical Imaging Centre, Semmelweis University, Budapest, Hungary
- Oncologic Imaging and Invasive Diagnostic Centre and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| |
Collapse
|
9
|
Mehta A, Priya V P L, Teja L, Pillai K. The Prevalence of Programmed Death Ligand-1 (PD-L1) Expression in Non-Small Cell Lung Cancer: An Experience From Tertiary Care Hospital. Cureus 2024; 16:e72291. [PMID: 39583517 PMCID: PMC11585287 DOI: 10.7759/cureus.72291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND The prevalence of programmed cell death ligand-1(PD-L1) expression in non-small cell lung cancer (NSCLC) within unselected populations remains a research topic, though PD-L1 expression is important in guiding treatment decisions. OBJECTIVES The study objective was to ascertain the prevalence of PD-L1 expression in patients of NSCLC and its association with clinicopathological characteristics and other gene mutations. METHODS Samples from patients with NSCLCs were analyzed in the study to determine the expression of PD-L1 through immunohistochemistry (IHC) using rabbit anti-human PDL-1/CD274 monoclonal antibody. Correlation analysis was done using Pearson's correlation coefficient (r). The study analyzed the association between PD-L1 expression and clinicopathological features. RESULTS A total of 245 consecutive patients (154 men with 62.9%) with NSCLCs were subjected to PD-L1 testing, and 30.6% (n=75) were identified to be positive. It was twice as prevalent in men than women, with 154 men and 91 females, respectively. The PD-L1 expression failed to demonstrate any statistical significance with age, gender, smoking status, type of materials, location of biopsy, Eastern Cooperative Oncology Group Performance Status, and Epidermal Growth Factor Receptor mutation. High PD-L1 expression with tumor proportion score (TPS ≥ 50) was observed in 38.8% of patients, and it was more prevalent in female patients 29 (38.7%), smokers 59 (78.7%), stage IV tumors 44 (58.7%), and stage III tumors 28 (37.3%). CONCLUSIONS Our study in Kerala showed PD-L1 expression in NSCLC patients, correlated with clinicopathologic factors. Males, COPD, ALK+, and advanced-stage tumors had higher expression. Females, smokers, poorly differentiated tumors, and stage IV tumors had high PD-L1.
Collapse
Affiliation(s)
- Asmita Mehta
- Respiratory Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, IND
| | - Lakshmi Priya V P
- Respiratory Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, IND
| | - Lakshmi Teja
- Respiratory Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, IND
| | - Krishna Pillai
- Respiratory Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, IND
| |
Collapse
|
10
|
Gambella A, Senetta R, Falco EC, Ricci AA, Mangherini L, Tampieri C, Fissore J, Orlando G, Manetta T, Mengozzi G, Mistrangelo M, Bertero L, Cassoni P. Prognostic and predictive role of YKL-40 in anal squamous cell carcinoma: a serological and tissue-based analysis in a multicentric cohort. Front Med (Lausanne) 2024; 11:1372195. [PMID: 39045410 PMCID: PMC11263350 DOI: 10.3389/fmed.2024.1372195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/03/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Anal squamous cell carcinoma (ASC) is a rare gastrointestinal malignancy showing an increased incidence over the past decades. YKL-40 is an immune modulator and pro-angiogenetic factor that showed a promising prognostic and predictive potential in several malignancies, but limited data are available for ASC. This study aims to provide an extensive evaluation of the prognostic and predictive role of YKL-40 in a multicenter cohort of ASC patients. Methods We retrospectively retrieved 72 consecutive cases of ASC diagnosed between February 2011 and March 2021. Both serum and tissue protein expression of YKL-40 were assessed, the latter in ASC tumor cells and peritumor immune cells. Results Increased YKL-40 serum levels at the time of diagnosis were associated with older age (p = 0.035), presence of cardiovascular/metabolic comorbidities (p = 0.007), and death for any cause (p = 0.011). In addition, high serum levels of YKL-40 were associated with a poor prognosis (HR: 2.82, 95% CI: 1.01-7.84; p = 0.047). Protein expression of YKL-40 in ASC tumor cells was significantly associated with low tumor grade (p = 0.031), while the increased expression in peritumor immune cells was associated with a worse response of patients to chemoradiotherapy (p = 0.007). However, YKL-40 protein expression in ASC tumor cells or peritumor immune cells did not significantly impact patient overall survival. Discussion In conclusion, YKL-40 resulted a relevant prognostic (serum level) and predictive (tissue protein expression in peritumor immune cells) biomarker and can considerably improve ASC patient clinical management.
Collapse
Affiliation(s)
- Alessandro Gambella
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Rebecca Senetta
- Pathology Unit, Department of Oncology, University of Turin, Turin, Italy
| | | | - Alessia Andrea Ricci
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Luca Mangherini
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Cristian Tampieri
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Jessica Fissore
- Pathology Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Giulia Orlando
- Pathology Unit, Department of Oncology, University of Turin, Turin, Italy
| | - Tilde Manetta
- Department of Laboratory Medicine, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Giulio Mengozzi
- Department of Laboratory Medicine, Città della Salute e della Scienza University Hospital, Turin, Italy
| | | | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Paola Cassoni
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
11
|
Liu N, Yang X, Gao C, Wang J, Zeng Y, Zhang L, Yin Q, Zhang T, Zhou H, Li K, Du J, Zhou S, Zhao X, Zhu H, Yang Z, Liu Z. Noninvasively Deciphering the Immunosuppressive Tumor Microenvironment Using Galectin-1 PET to Inform Immunotherapy Responses. J Nucl Med 2024; 65:728-734. [PMID: 38514084 DOI: 10.2967/jnumed.123.266888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Immune checkpoint blockade (ICB) has achieved groundbreaking results in clinical cancer therapy; however, only a subset of patients experience durable benefits. The aim of this study was to explore strategies for predicting tumor responses to optimize the intervention approach using ICB therapy. Methods: We used a bilateral mouse model for proteomics analysis to identify new imaging biomarkers for tumor responses to ICB therapy. A PET radiotracer was synthesized by radiolabeling the identified biomarker-targeting antibody with 124I. The radiotracer was then tested for PET prediction of tumor responses to ICB therapy. Results: We identified galectin-1 (Gal-1), a member of the carbohydrate-binding lectin family, as a potential negative biomarker for ICB efficacy. We established that Gal-1 inhibition promotes a sensitive immune phenotype within the tumor microenvironment (TME) for ICB therapy. To assess the pre-ICB treatment status of the TME, a Gal-1-targeted PET radiotracer, 124I-αGal-1, was developed. PET imaging with 124I-αGal-1 showed the pretreatment immunosuppressive status of the TME before the initiation of therapy, thus enabling the prediction of ICB resistance in advance. Moreover, the use of hydrogel scaffolds loaded with a Gal-1 inhibitor, thiodigalactoside, demonstrated that a single dose of thiodigalactoside-hydrogel significantly potentiated ICB and adoptive cell transfer immunotherapies by remodeling the immunosuppressive TME. Conclusion: Our study underscores the potential of Gal-1-targeted PET imaging as a valuable strategy for early-stage monitoring of tumor responses to ICB therapy. Additionally, Gal-1 inhibition effectively counteracts the immunosuppressive TME, resulting in enhanced immunotherapy efficacy.
Collapse
Affiliation(s)
- Ning Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiujie Yang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Chao Gao
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jianze Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yuwen Zeng
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Linyu Zhang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Qi Yin
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ting Zhang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Haoyi Zhou
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Kui Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jinhong Du
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Shixin Zhou
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xuyang Zhao
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhaofei Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China;
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, China
- Department of Nuclear Medicine, Peking University Third Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; and
- Peking University-Yunnan Baiyao International Medical Research Center, Beijing, China
| |
Collapse
|
12
|
Aggarwal A, Sharma S, Brar Z, Kumar V, Kumar A, Katara R, Mohanty SK. Comprehensive molecular analysis of driver mutations in non-small cell lung carcinomas and its correlation with PD-L1 expression, An Indian perspective. Pathol Res Pract 2024; 253:155013. [PMID: 38096714 DOI: 10.1016/j.prp.2023.155013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND The understanding of molecular mechanisms involved in non-small cell lung carcinoma (NSCLC) has revolutionized significantly in the recent years. These have helped to develop personalized management strategies by identifying specific molecular alterations such as mutations in EGFR, ROS1, BRAF, ERBB2, MET, ALK, and KRAS genes. These mutations are targetable ensuring a better clinical outcome. Next-generation sequencing (NGS) methodology is the recommended technique for the identification of driver mutations in the five hot-spot genes (EGFR, ALK, ROS1, MET, and BRAF) involved in the NSCLC. NGS has numerous advantages including multiplexing, tissue conservation, identification of rare and novel variants, and reduced cost over the sequential single gene testing. Herein, we sought to demonstrate the mutational profile in NSCLC and their clinicopathologic correlation in a contemporary cohort of Indian NSCLC patients. Additionally, we studied the correlation of oncogenic driver mutations with PD-L1 status in these patients. MATERIALS AND METHODS Five fifty-two stage IV NSCLC patients (adenocarcinoma=490; squamous cell carcinoma=51; adenosquamous carcinoma=5; large cell carcinoma=2; sarcomatoid carcinoma=3; spindle cell carcinoma=1) underwent broad molecular profiling by a custom-made, targeted DNA- and RNA-based five hot-spot genes lung cancer panel (EGFR, ALK, ROS1, BRAF, and MET), compatible with the NGS Ion S5 system. The mutations were correlated with the clinicopathologic characteristics. Additionally, PD-L1 expression status, available on 252 tumors, was correlated with the oncogenic drivers. RESULTS Validation of the 5 gene panel yielded the following results: a) specificity of 99.74%; b) sensitivity of 100% for single nucleotide variants (SNVs) (>5% variant allele frequency, VAF), indels (>10% VAF) and fusions; c) 100% intra- and inter-run reproducibility; d) 88% inter-laboratory agreement. Validated panel was then used to analyze clinical samples. Sixty percentage tumors harbored either one (54.71%) or multiple (3.26%) mutations. EGFR and BRAF V600E mutations, ALK and ROS1 rearrangements, and MET exon 14 skipping mutation were observed in 38.41% (n = 212) and 2.72% (n = 15) patients, 12.14% (n = 67) and 3.62% (n = 20) patients, and 1.09% (n = 6) patients, respectively. EGFR exon 19 deletion accounted for 52.83% of all mutations, followed by L858R (35.85%), T790M (5.19%), exon 20 insertions (6.6%), and other rare mutations (G719X, L861Q, S768I) (9.91%). Concurrent EGFR with ALK, EGFR with ROS1, EGFR with MET, and EGFR with BRAF were observed in 10, 4, 1, and 3 patients, respectively. PD-L1 was expressed in 134 patients (53.2%). Exon 19 deletion was more prevalent in PD-L1 negative tumors whereas exon 21 substitution (L858R) was seen more in PD-L1 positive tumors. CONCLUSIONS This is one of the largest cohorts of NSCLC for comprehensive targeted mutational profiling and correlation with the PD-L1 expression. The mutations are more prevalent in non-smoker females for all genes, except ALK (non-smoker males). MET and BRAF mutations are more common in elderly population whereas EGFR mutations, and ALK and ROS1 genes rearrangements are more prevalent in younger population. The most common histopathologic subtype/feature associated with various mutations was as follows: acinar with EGFR, solid with ALK, macronucleoli with ROS1, signet ring with MET, and micropapillary with BRAF.
Collapse
Affiliation(s)
- Aditi Aggarwal
- Department of Molecular Pathology, CORE Diagnostics, Gurugram, Haryana, India
| | - Shivani Sharma
- Department of Molecular Pathology, CORE Diagnostics, Gurugram, Haryana, India
| | - Zoya Brar
- Department of Molecular Pathology, CORE Diagnostics, Gurugram, Haryana, India
| | - Vipin Kumar
- Department of Molecular Pathology, CORE Diagnostics, Gurugram, Haryana, India
| | - Akash Kumar
- Department of Molecular Pathology, CORE Diagnostics, Gurugram, Haryana, India
| | - Rahul Katara
- Department of Molecular Pathology, CORE Diagnostics, Gurugram, Haryana, India
| | - Sambit K Mohanty
- Department of Molecular Pathology, CORE Diagnostics, Gurugram, Haryana, India.
| |
Collapse
|
13
|
Song XQ, Li RJ, Zhang S. Prediction of prognosis of patients with hepatocellular carcinoma based on immune-related score. Asian J Surg 2024; 47:310-319. [PMID: 37673742 DOI: 10.1016/j.asjsur.2023.08.175] [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: 03/01/2023] [Revised: 07/02/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Immune-related scores are currently used for prognostic evaluation and as an immunotherapy reference in various cancers. However, the relationship between immune-related score and hepatocellular carcinoma (HCC) prognosis has not yet been investigated. This study aimed to explore the clinical application value of immune-related score for predicting HCC prognosis-related indicators including disease-free survival (DFS) and overall survival (OS), and to construct a clinical nomogram prediction model related to verification. METHODS This study included 284 HCC patients who were selected from the Cancer Genome Atlas (TCGA) database and linked to the immune-related score downloaded from the public platform. A Cox proportional hazards regression model was used to estimate the adjusted risk ratio, and a nomogram was constructed based on multivariate analysis results and clinical significance. The model was internally verified by bootstrap. The performance of the prediction model was evaluated using the C-index and calibration curves. RESULTS Patients were divided into three subgroups according to the immune-related score level. Compared with patients in the low immune-related score group, the DFS of patients in the medium and high immune-related score groups was significantly prolonged (HR: 0.53, 95% CI: 0.32-0.87; HR: 0.37, 95% CI: 0.21-0.63, respectively). The OS of patients in the medium and high immune-related score groups was also significantly prolonged (HR: 0.43, 95% CI: 0.20-0.95, p = 0.038; HR: 0.29, 95% CI: 0.14-0.58, p < 0.001, respectively). The C-indexes for predicting DFS and OS were 0.687 (95% CI: 0.665-0.700) and 0.743 (95% CI: 0.709-0.776), respectively. The calibration curves of 3-year and 5-year DFS and OS showed that the results predicted by the nomogram were in good agreement with the actual observations. CONCLUSIONS Moderate/high-grade immune-related score was significantly associated with better DFS and OS in HCC patients. In addition, a nomogram for prognosis estimation can help clinicians predict the survival status of patients.
Collapse
Affiliation(s)
- Xian-Qing Song
- General Surgery Department, Baoan Central Hospital, The Fifth Affiliated Hospital of Shen Zhen University, Xixiang Street, Bao 'an District, Shenzhen, 518000, Guangdong, PR China.
| | - Rong-Jiang Li
- General Surgery Department, Baoan Central Hospital, The Fifth Affiliated Hospital of Shen Zhen University, Xixiang Street, Bao 'an District, Shenzhen, 518000, Guangdong, PR China.
| | - Sen Zhang
- Department of Colorectal Surgery, First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, Guangxi, 530000, PR China.
| |
Collapse
|
14
|
Pal R, K M, Matsui A, Kang H, Morita S, Taniguchi H, Kobayashi T, Morita A, Choi HS, Duda DG, Kumar ATN. In vivo quantification of programmed death-ligand-1 expression heterogeneity in tumors using fluorescence lifetime imaging. RESEARCH SQUARE 2023:rs.3.rs-3222037. [PMID: 37961361 PMCID: PMC10635296 DOI: 10.21203/rs.3.rs-3222037/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Cancer patient selection for immunotherapy is often based on programmed death-ligand-1 (PD-L1) expression as a biomarker. PD-L1 expression is currently quantified using immunohistochemistry, which can only provide snapshots of PD-L1 expression status in microscopic regions of ex vivo specimens. In vivo imaging using targeted agents can capture dynamic variations of PD-L1 expression in entire tumors within and across multiple subjects. Towards this goal, several PD-L1 targeted molecular imaging probes have been evaluated in murine models and humans. However, clinical translation of these probes has been limited due to a significant non-specific accumulation of the imaging probes and the inability of conventional imaging modalities to provide quantitative readouts that can be compared across multiple subjects. Here we report that in vivo time-domain (TD) fluorescence imaging can provide quantitative estimates of baseline tumor PD-L1 heterogeneity across untreated mice and variations in PD-L1 expression across mice undergoing clinically relevant anti-PD1 treatment. This approach relies on a significantly longer fluorescence lifetime (FLT) of PD-L1 specific anti-PD-L1 antibody tagged to IRDye 800CW (αPDL1-800) compared to nonspecific αPDL1-800. Leveraging this unique FLT contrast, we show that PD-L1 expression can be quantified across mice both in superficial breast tumors using planar FLT imaging, and in deep-seated liver tumors (>5 mm depth) using the asymptotic TD algorithm for fluorescence tomography. Our results suggest that FLT contrast can accelerate the preclinical investigation and clinical translation of novel molecular imaging probes by providing robust quantitative readouts of receptor expression that can be readily compared across subjects.
Collapse
Affiliation(s)
- Rahul Pal
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Murali K
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aya Matsui
- Department of Vascular Physiology, Graduate School of Medical Science, Kanazawa University, Japan
| | - Homan Kang
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Satoru Morita
- E. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hajime Taniguchi
- E. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Tohoku Graduate School of Medicine, Sendai, Japan
| | - Tatsuya Kobayashi
- E. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Atsuyo Morita
- E. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dan G Duda
- E. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anand T N Kumar
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Parra ER, Ilié M, Wistuba II, Hofman P. Quantitative multiplexed imaging technologies for single-cell analysis to assess predictive markers for immunotherapy in thoracic immuno-oncology: promises and challenges. Br J Cancer 2023; 129:1417-1431. [PMID: 37391504 PMCID: PMC10628288 DOI: 10.1038/s41416-023-02318-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/05/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023] Open
Abstract
The past decade has witnessed a revolution in cancer treatment by the shift from conventional drugs (chemotherapies) towards targeted molecular therapies and immune-based therapies, in particular the immune-checkpoint inhibitors (ICIs). These immunotherapies selectively release the host immune system against the tumour and have shown unprecedented durable remission for patients with cancers that were thought incurable such as advanced non-small cell lung cancer (aNSCLC). The prediction of therapy response is based since the first anti-PD-1/PD-L1 molecules FDA and EMA approvals on the level of PD-L1 tumour cells expression evaluated by immunohistochemistry, and recently more or less on tumour mutation burden in the USA. However, not all aNSCLC patients benefit from immunotherapy equally, since only around 30% of them received ICIs and among them 30% have an initial response to these treatments. Conversely, a few aNSCLC patients could have an efficacy ICIs response despite low PD-L1 tumour cells expression. In this context, there is an urgent need to look for additional robust predictive markers for ICIs efficacy in thoracic oncology. Understanding of the mechanisms that enable cancer cells to adapt to and eventually overcome therapy and identifying such mechanisms can help circumvent resistance and improve treatment. However, more than a unique universal marker, the evaluation of several molecules in the tumour at the same time, particularly by using multiplex immunostaining is a promising open room to optimise the selection of patients who benefit from ICIs. Therefore, urgent further efforts are needed to optimise to individualise immunotherapy based on both patient-specific and tumour-specific characteristics. This review aims to rethink the role of multiplex immunostaining in immuno-thoracic oncology, with the current advantages and limitations in the near-daily practice use.
Collapse
Affiliation(s)
- Edwin Roger Parra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Biobank Côte d'Azur BB-0033-00025, FHU OncoAge, IHU RespirERA, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Biobank Côte d'Azur BB-0033-00025, FHU OncoAge, IHU RespirERA, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France.
| |
Collapse
|
16
|
Hofman P. Implementation of the clinical practice of liquid biopsies for thoracic oncology the experience of the RespirERA university hospital institute (Nice, France). THE JOURNAL OF LIQUID BIOPSY 2023; 1:100004. [PMID: 40027288 PMCID: PMC11863941 DOI: 10.1016/j.jlb.2023.100004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 03/05/2025]
Abstract
According to international guidelines, it is mandatory to evaluate predictive biomarkers of targeted therapies and the response to immune check point inhibitors for patients with non-squamous non-small cell lung cancer (NS-NSCLC). For this purpose, a tissue sample is nowadays the gold standard, but biofluids, particularly peripheral blood, can be a complementary and sometimes an alternative approach to assess the status of different druggable genomic alterations of advanced NS-NSCLC. A liquid biopsy (LB) is an attractive approah for better treatment decision-making by thoracic oncologists for NSCLC patients in daily practice at both initial diagnosis and tumor progression. We describe the experience of a clinical and molecular pathology laboratory (LPCE, Nice, France) developing the use of in-house LB in thoracic oncology. Moreover, we report the changes in clinical care, the advantages, but also the possible constraints associated with implantation of LB in routine clinical practice.
Collapse
Affiliation(s)
- Paul Hofman
- Côte d’Azur University, IHU RespirERA, FHU OncoAge, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, Nice, France
- Côte d’Azur University, IRCAN, Inserm U1081, CNRS 7284, France
| |
Collapse
|
17
|
Li T, Wang X, Niu M, Wang M, Zhou J, Wu K, Yi M. Bispecific antibody targeting TGF-β and PD-L1 for synergistic cancer immunotherapy. Front Immunol 2023; 14:1196970. [PMID: 37520520 PMCID: PMC10373067 DOI: 10.3389/fimmu.2023.1196970] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
The PD-1/PD-L1 signaling pathway plays a crucial role in cancer immune evasion, and the use of anti-PD-1/PD-L1 antibodies represents a significant milestone in cancer immunotherapy. However, the low response rate observed in unselected patients and the development of therapeutic resistance remain major obstacles to their clinical application. Accumulating studies showed that overexpressed TGF-β is another immunosuppressive factor apart from traditional immune checkpoints. Actually, the effects of PD-1 and TGF-β pathways are independent and interactive, which work together contributing to the immune evasion of cancer cell. It has been verified that blocking TGF-β and PD-L1 simultaneously could enhance the efficacy of PD-L1 monoclonal antibody and overcome its treatment resistance. Based on the bispecific antibody or fusion protein technology, multiple bispecific and bifunctional antibodies have been developed. In the preclinical and clinical studies, these updated antibodies exhibited potent anti-tumor activity, superior to anti-PD-1/PD-L1 monotherapies. In the review, we summarized the advances of bispecific antibodies targeting TGF-β and PD-L1 in cancer immunotherapy. We believe these next-generation immune checkpoint inhibitors would substantially alter the cancer treatment paradigm, especially in anti-PD-1/PD-L1-resistant patients.
Collapse
Affiliation(s)
- Tianye Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Xinrun Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Mengke Niu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mingli Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Hofman P. Matched tissue and liquid biopsies for advanced non-small cell lung cancer patients A potentially indispensable complementary approach. Transl Oncol 2023; 35:101735. [PMID: 37413719 PMCID: PMC10366644 DOI: 10.1016/j.tranon.2023.101735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 05/17/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023] Open
Abstract
The introduction of liquid biopsies (LB) has brought forth a number of therapeutic opportunities into the domain of thoracic oncology. Many of which have been adopted for care of patients presenting with advanced non-squamous non-small cell lung cancer (aNS-NSCLC). For example, one of the most frequent indications to perform a LB in these patients, at least in Europe, is for patients treated with tyrosine kinase inhibitors (TKIs) targeting EGFR and ALK genomic alterations when the tumor progresses. A tissue biopsy (TB) must then be taken, ideally from a site of a tumor that progresses, in particular if the LB does not permit detection of a mechanism of resistance to TKI. A LB from a patient with aNS-NSCLC is recommended before first-line therapy if no tissue and/or cytological material is accessible or if the extracted nucleic acid is insufficient in amount and/or of poor quality. At present a LB and a TB are rarely performed simultaneously before treatment and/or on tumor progression. This complementary/matched testing approach is still controversial but needs to be better evaluated to determine the true benefit to care of patients. This review provides an update on the complementarity of the LB and TB method for care of patients presenting with aNS-NSCLC.
Collapse
Affiliation(s)
- Paul Hofman
- Laboratory of Clinical and Experimental Pathology de Pathologie, University Côte d'Azur, FHU OncoAge, Biobank BB-0033-00025, IHU RespireRA, 30 Avenue de la Voie Romaine, 01, Nice 06002 CEDEX, France.
| |
Collapse
|
19
|
Chen H, Ge M, Zhang F, Xing Y, Yu S, Chen C, Zhang H, Wang X, Gao X, Chen F, Chen P, Zhang D, Zhan Q, Zhu Y. Correlation between immunotherapy biomarker PD-L1 expression and genetic alteration in patients with non-small cell lung cancer. Genomics 2023; 115:110648. [PMID: 37217086 DOI: 10.1016/j.ygeno.2023.110648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/19/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
Programmed death-ligand 1 (PD-L1) has been widely used in immunotherapy evaluation of patients with non-small cell lung cancer (NSCLC). However, the effect is not particularly ideal, and the association between PD-L1 and genetic alterations requires more exploration. Here, we performed targeted next-generation sequencing and PD-L1 immunohistochemistry (IHC) testing for PD-L1 expression on both tumor cells (TCs) and tumor-infiltrating immune cells (ICs) in 1549 patients. Our studies showed that surgical method of resection was positively correlated with IC+, and a low tumor mutation burden (TMB) was negatively correlated with TC+. Furthermore, we found that EGFR was mutually exclusive with both ALK and STK11. In addition, the features between PD-L1 expression status and genomic alterations were characterized. These results suggest that clinical characteristics and molecular phenotypes are associated with PD-L1 expression signatures, which may provide novel insights for improving the efficiency of immune checkpoint inhibitors (ICIs) in immunotherapy.
Collapse
Affiliation(s)
- Hefeng Chen
- Department of Pulmonary and Critical Care Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Mengxi Ge
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | - Xing Gao
- 3D Medicines Inc., Shanghai, China
| | | | | | | | - Qiong Zhan
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Youcai Zhu
- Department of Thoracic Disease Center, Zhejiang Rongjun Hospital, Jiaxing, China.
| |
Collapse
|
20
|
Ilié M, Hofman V, Bontoux C, Goffinet S, Benzaquen J, Heeke S, Boutros J, Lassalle S, Long-Mira E, Zahaf K, Lalvée S, Lespinet-Fabre V, Bordone O, Tanga V, Gómez-Caro A, Cohen C, Berthet JP, Marquette CH, Hofman P. Lack of correlation between MET and PD-L1 expression in non-small cell lung cancer revealed by comparative study of matched biopsies and surgical resection samples. Lung Cancer 2023; 181:107230. [PMID: 37150140 DOI: 10.1016/j.lungcan.2023.107230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/09/2023]
Abstract
INTRODUCTION Both MET expression and the PD-L1 tumor proportion score (TPS) are companion diagnostics for treatment of advanced non-small cell lung carcinoma (aNSCLC) patients. We evaluated the rate of correlation between MET expression and the PD-L1 TPS in matched biopsies and surgically resected specimens from NSCLC patients. PATIENTS AND METHODS This retrospective analysis assessed the prevalence and correlation between MET expression (SP44 clone) and the PD-L1 TPS (22C3 clone) by immunohistochemistry together with molecular alterations determined by targeted next-generation sequencing in matched lung biopsy and surgically lung resected specimens from 70 patients with NSCLC. RESULTS The study found a significant correlation between the MET H-score in surgical samples and matched biopsies (P-value < 0.0001), as well as between the PD-L1 TPS in paired biopsies and surgical samples (P-value < 0.0001). However, there was no significant correlation between the MET H-score or expression subgroups and the PD-L1 TPS in both types of paired samples (P-value = 0.47, and P-value = 0.90). The MET H-score was significantly higher in adenocarcinoma compared to squamous cell carcinoma (P-value < 0.0001). A mutational analysis showed that the MET H-score was significantly higher in NSCLC cases with targetable molecular alterations (P-value = 0.0095), while no significant correlation was found for the PD-L1 TPS. CONCLUSIONS Our study found no significant correlation between PD-L1 and MET expression in samples from NSCLC patients, highlighting the importance of personalized treatment strategies based on individual expression profiles. These findings provide valuable insight into the development of effective immunotherapy and targeted therapy for NSCLC patients.
Collapse
Affiliation(s)
- Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Samantha Goffinet
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Jonathan Benzaquen
- FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France; Department of Pulmonary Medicine and Thoracic Oncology, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jacques Boutros
- FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Department of Pulmonary Medicine and Thoracic Oncology, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France
| | - Katia Zahaf
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Salomé Lalvée
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Olivier Bordone
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Virginie Tanga
- Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France
| | - Abel Gómez-Caro
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Charlotte Cohen
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Jean-Philippe Berthet
- Department of Thoracic Surgery, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Charles-Hugo Marquette
- FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France; Department of Pulmonary Medicine and Thoracic Oncology, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Hospital-integrated Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France; FHU OncoAge, Hôpital Pasteur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France; Team 4, Inserm U1081, CNRS UMR 7413, Institute for Research on Cancer and Aging, Nice, France.
| |
Collapse
|
21
|
PD-1/PD-L1 inhibitors in treatment-naïve, advanced non-small cell lung cancer patients with < 1% PD-L1 expression: a meta-analysis of randomized controlled trials. J Cancer Res Clin Oncol 2022; 149:2179-2189. [PMID: 35984491 DOI: 10.1007/s00432-022-04286-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/13/2022] [Indexed: 10/15/2022]
Abstract
BACKGROUND PD-1/PD-L1 inhibitors prolong survival in treatment-naïve, locally advanced, and metastatic non-small cell lung cancer (NSCLC) with positive PD-L1 expression (> 1%/ > 50%). Recent evidence has suggested that tumors with < 1% PD-L1 expression may also be predictive of PD-1/PD-L1 inhibiting agents. METHODS Systematic review and meta-analysis were conducted of randomized controlled trials (RCTs) evaluating PD-1/PD-L1 inhibitors that have assessed tumors with < 1% PD-L1 expression (negative PD-L1 expression). PD-1/PD-L1 inhibitors-chemotherapy combinations (PC) were compared with histology-selected chemotherapy with respect to overall survival (OS) and progression-free survival (PFS). RESULTS Twelve RCTs comprising 5410 participants (PD-1/PD-L1 inhibitors-chemotherapy: 3051; chemotherapy: 2359) met the inclusion criteria. Tumors with PD-L1 expression < 1% were evident in 38.9% of the pooled study population. A significant OS [hazard ratio (HR) 0.71 95% confidence interval (CI) 0.63-0.80, p < 0.00001] and PFS [HR 0.65 95% CI 0.58-0.72, p < 0.00001] benefit of PC was evident in tumors with negative PD-L1 expression. PD-1/PD-L1 inhibitors-chemotherapy combinations were more likely to achieve an objective response than chemotherapy [odds ratio, 1.86; 95% CI, 1.46-2.38, p < 0.00001]. Histologic subtypes and diagnostic assays did not modify the OS and PFS treatment effects for PC compared to chemotherapy. CONCLUSION Tumors harboring < 1% PD-L1 expression are likely to benefit from PD-1/PD-L1 inhibitor-chemotherapy regimens in advanced NSCLC.
Collapse
|
22
|
Liu B, Yan S, Li S, Zhang Q, Yang M, Yang L, Ma J, Li X. Correlation Study of PD-L1, CD4, CD8, and PD-1 in Primary Diffuse Large B-cell Lymphoma of the Central Nervous System. Pathol Res Pract 2022; 239:154008. [DOI: 10.1016/j.prp.2022.154008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/21/2022] [Accepted: 06/29/2022] [Indexed: 11/27/2022]
|
23
|
Rizzo A, Ricci AD. Predictors of response for hepatocellular carcinoma immunotherapy: is there anything on the horizon? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022. [DOI: 10.1080/23808993.2022.2075724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello,” I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Angela Dalia Ricci
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
24
|
Li Y, Wu L, Liu Y, Ma S, Huang B, Feng X, Wang H. A novel multifunctional anti-PD-L1-CD16a-IL15 induces potent cancer cell killing in PD-L1-positive tumour cells. Transl Oncol 2022; 21:101424. [PMID: 35477065 PMCID: PMC9136603 DOI: 10.1016/j.tranon.2022.101424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022] Open
Abstract
Anti-PD-L1 single-domain antibodies were identified from hPD-L1-immunized camels. Three novel multifunctional antibodies, anti-PD-L1-CD16a, anti-PD-L1-IL15, and anti-PD-L1-CD16a-IL15, target PD-L1-positive cancer cells. Anti-PD-L1-IL15 and anti-PD-L1-CD16a-IL15, but not anti-PD-L1-CD16a, stimulate immune cell proliferation in vitro. The anti-PD-L1 antibodies can bind PD-L1-positive cells. Anti-PD-L1-CD16a-IL15 has the strongest antitumour activity, both in vitro and in vivo.
Cancer is the most acute disease and the leading cause of patient death worldwide. Both chemotherapy and molecular-based therapies play an important role in curing cancer. However, the median and overall survival of patients is poor. To date, immune therapies have changed the treatment methods for cancer patients. Programmed death ligand 1 (PD-L1, also known as B-H1, CD274) is a well-studied tumor antigen. PD-L1 is overexpressed in colon cancer, lung cancer, and so on and plays a vital role in cancer development. In this study, anti-PD-L1 single-domain antibodies were identified from recombinant human PD-L1 (rhPD-L1)-immunized llamas. Then, we generated a novel multifunctional anti-PD-L1-CD16a-IL15 antibody targeting PD-L1-positive tumor cells. Anti-PD-L1-CD16a-IL15 was constructed by linking the Interleukin-2 (IL-2) signal peptide, anti-PD-L1 single domain antibody (anti-PD-L1-VHH) and anti-cluster of differentiation 16a single domain antibody (anti-CD16a-VHH), and Interleukin-15/Interleukin-15 receptor alpha (IL15/IL-15Rα). This anti-PD-L1-CD16a-IL15 fusion protein can be expressed and purified from HEK-293F cells. In vitro, our data showed that the anti-PD-L1-CD16a-IL15 fusion protein can recruit T cells and drive natural killer cells (NK) with specific killing of PD-L1-overexpressing tumor cells. Furthermore, in the xenograft model, the anti-PD-L1-CD16a-IL15 fusion protein inhibited tumor growth with human peripheral blood mononuclear cells (PBMCs). These data suggested that the anti-PD-L1-CD16a-IL15 fusion protein has a latent function in antitumour activity, with better guidance for future cancer immunotherapy.
Collapse
Affiliation(s)
- Yumei Li
- School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Lingjun Wu
- School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Yueying Liu
- Department of Hypertension, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Siwen Ma
- School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Biyi Huang
- School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Xianjing Feng
- School of Pharmacy, Guangxi Medical University, Nanning, China.
| | - Hui Wang
- School of Pharmacy, Guangxi Medical University, Nanning, China.
| |
Collapse
|
25
|
Sunpaweravong P, Thongwatchara P, Chotipanvithayakul R, Sangkhathat S, Thongsuksai P. Expression and Prognostic Significance of c-Myc, ALK, ROS1, BRAF, and PD-L1 Among Patients With Non-Small Cell Lung Cancer. Clin Med Insights Oncol 2022; 16:11795549221092747. [PMID: 35479767 PMCID: PMC9036383 DOI: 10.1177/11795549221092747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/13/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) remains a leading cause of cancer death worldwide, for which better knowledge in molecular prognostic factors is needed to improve clinical outcome. This study aimed to investigate the clinical significance of c-Myc, ALK, ROS1, BRAF, and PD-L1 in NSCLC patients. METHODS Formalin-fixed paraffin-embedded tissue specimens were obtained from 124 NSCLC patients. Of these, 66 matched specimens of normal respiratory epithelial and tumor tissue from patients with stages I-III, who underwent surgical resection, and 58 NSCLC specimens from stage IV patients were recruited into this analysis. Immunohistochemistry staining along with semiquantitative criteria were used to evaluate the expression of the interested proteins. RESULTS Of the 66 patients with stages I-III, positive expression of c-Myc was detected in 12 specimens (18.2%) of NSCLC tissue, whereas none of the normal respiratory epithelial tissue was found to have c-Myc expression (P < .001). Of the 66 NSCLC patients, 28 (43.8%) had PD-L1-positive staining on 1%-49% tumor cells and 7 (10.9%) patients expressed PD-L1 in ⩾50% tumor cells. One (2.3%) adenocarcinoma patient was found to have ROS1 rearrangement. Patients with no expression of c-Myc and PD-L1 (co-negative expression) tended to have a better prognosis than other subgroups. CONCLUSIONS NSCLC tissue significantly expressed more c-Myc and PD-L1, compared with the matched normal respiratory epithelium, emphasizing the important role of these key drivers in tumorigenesis. Therapeutic approach to precisely inhibit the targetable molecular pathways should be considered on an individual patient basis to improve survival outcome.
Collapse
Affiliation(s)
- Patrapim Sunpaweravong
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
- Department of Biomedical Science, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Patcharaporn Thongwatchara
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | | | - Surasak Sangkhathat
- Department of Biomedical Science, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Paramee Thongsuksai
- Department of Biomedical Science, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
26
|
Rizzo A, Cusmai A, Gadaleta-Caldarola G, Palmiotti G. Which role for predictors of response to immune checkpoint inhibitors in hepatocellular carcinoma? Expert Rev Gastroenterol Hepatol 2022; 16:333-339. [PMID: 35403533 DOI: 10.1080/17474124.2022.2064273] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) remains a frequently diagnosed malignancy worldwide, still representing an important cause of cancer-related death. Recent years have seen the emergence of novel systemic treatments for HCC patients, including immune checkpoint inhibitors (ICIs). Nonetheless, several questions regarding HCC immunotherapy remain unanswered, especially in terms of biochemical predictors of response. AREAS COVERED In the current paper, we will discuss available evidence regarding predictive biomarkers of response to HCC immunotherapy. A literature search was conducted in January 2022 of Pubmed/Medline, Cochrane library, and Scopus databases. EXPERT OPINION The identification of predictive biomarkers represents an unmet need in HCC patients receiving ICIs. The HCC medical community is called to further efforts aimed to elucidate the effective role of PD-L1 expression, TMB, MSI, gut microbiota, and other emerging biomarkers.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico "Don Tonino Bello," I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Antonio Cusmai
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico "Don Tonino Bello," I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Gennaro Gadaleta-Caldarola
- Medical Oncology Unit, 'Mons. R. Dimiccoli' Hospital, Barletta (BT), Azienda Sanitaria Locale Barletta, 76121, Italy
| | - Gennaro Palmiotti
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico "Don Tonino Bello," I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124 Bari, Italy
| |
Collapse
|
27
|
Chen Q, Hu J, Hu X, Koh K, Chen H. Current methods and emerging approaches for detection of programmed death ligand 1. Biosens Bioelectron 2022; 208:114179. [PMID: 35364526 DOI: 10.1016/j.bios.2022.114179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 02/08/2023]
Abstract
Various tumor cells overexpress programmed death ligand 1 (PD-L1), a main immune checkpoint protein (ICP) embedded in the tumor cells membrane, to evade immune recognition through the interaction between PD-L1 and its receptor programmed death 1 (PD-1) which is from T-cells for maintaining immune tolerance. So inhibitors targeting the PD-1 or PD-L1 can block the PD-1/PD-L1 signaling pathway to restore the recognition activity of the immune system to tumor cells, which also have been utilized as a novel approach to improve the clinical therapeutic effect for cancer patients. Since not all cancer patients can respond to these inhibitors effectively, previous diagnosis of PD-L1 is significant to target the right treatments for cancer patients. This review pays attention to the PD-L1 detection and recent progress in the measurement of PD-L1 concentration, including various detection methods based on optical sensors as well as electrochemical assays. Apart from above those, we also focus on the prospects of PD-L1 detection in precision medicine.
Collapse
Affiliation(s)
- Qiang Chen
- School of Life Sciences, Shanghai University, Shanghai, 200444, PR China; School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Junjie Hu
- School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Xiaojun Hu
- School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Kwangnak Koh
- Institute of General Education, Pusan National University, Busan, 609-735, Republic of Korea
| | - Hongxia Chen
- School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
28
|
Dyugay IA, Lukyanov DK, Turchaninova MA, Serebrovskaya EO, Bryushkova EA, Zaretsky AR, Khalmurzaev O, Matveev VB, Shugay M, Shelyakin PV, Chudakov DM. Accounting for B-cell Behavior and Sampling Bias Predicts Anti-PD-L1 Response in Bladder Cancer. Cancer Immunol Res 2022; 10:343-353. [PMID: 35013004 PMCID: PMC9381118 DOI: 10.1158/2326-6066.cir-21-0489] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/18/2021] [Accepted: 01/06/2022] [Indexed: 01/07/2023]
Abstract
Cancer immunotherapy is predominantly based on T cell-centric approaches. At the same time, the adaptive immune response in the tumor environment also includes clonally produced immunoglobulins and clonal effector/memory B cells that participate in antigen-specific decisions through their interactions with T cells. Here, we investigated the role of infiltrating B cells in bladder cancer via patient dataset analysis of intratumoral immunoglobulin repertoires. We showed that the IgG1/IgA ratio is a prognostic indicator for several subtypes of bladder cancer and for the whole IMVigor210 anti-PD-L1 immunotherapy study cohort. A high IgG1/IgA ratio associated with the prominence of a cytotoxic gene signature, T-cell receptor signaling, and IL21-mediated signaling. Immunoglobulin repertoire analysis indicated that effector B-cell function, rather than clonally produced antibodies, was involved in antitumor responses. From the T-cell side, we normalized a cytotoxic signature against the extent of immune cell infiltration to neutralize the artificial sampling-based variability in immune gene expression. Resulting metrics reflected proportion of cytotoxic cells among tumor-infiltrating immune cells and improved prediction of anti-PD-L1 responses. At the same time, the IgG1/IgA ratio remained an independent prognostic factor. Integration of the B-cell, natural killer cell, and T-cell signatures allowed for the most accurate prediction of anti-PD-L1 therapy responses. On the basis of these findings, we developed a predictor called PRedIctive MolecUlar Signature (PRIMUS), which outperformed PD-L1 expression scores and known gene signatures. Overall, PRIMUS allows for reliable identification of responders among patients with muscle-invasive urothelial carcinoma, including the subcohort with the low-infiltrated "desert" tumor phenotype.
Collapse
Affiliation(s)
- Ilya A. Dyugay
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.,Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Daniil K. Lukyanov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.,Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Maria A. Turchaninova
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Ekaterina O. Serebrovskaya
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Ekaterina A. Bryushkova
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Molecular Biology Department, Lomonosov Moscow State University, Moscow, Russia
| | - Andrew R. Zaretsky
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Oybek Khalmurzaev
- Department of Urology, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vsevolod B. Matveev
- Department of Urology, Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Mikhail Shugay
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Pavel V. Shelyakin
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Dmitriy M. Chudakov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.,Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Corresponding Author: Dmitriy M. Chudakov, Genomics of Adaptive Immunity, IBCH RAS, Miklukho-Maklaya, 16/10, Moscow 117997, Russia. Phone: 7 (495) 335-01-00; E-mail:
| |
Collapse
|
29
|
Artificial intelligence-assisted system for precision diagnosis of PD-L1 expression in non-small cell lung cancer. Mod Pathol 2022; 35:403-411. [PMID: 34518630 DOI: 10.1038/s41379-021-00904-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
Standardized programmed death-ligand 1 (PD-L1) assessment in non-small cell lung cancer (NSCLC) is challenging, owing to inter-observer variability among pathologists and the use of different antibodies. There is a strong demand for the development of an artificial intelligence (AI) system to obtain high-precision scores of PD-L1 expression in clinical diagnostic scenarios. We developed an AI system using whole slide images (WSIs) of the 22c3 assay to automatically assess the tumor proportion score (TPS) of PD-L1 expression based on a deep learning (DL) model of tumor detection. Tests were performed to show the diagnostic ability of the AI system in the 22c3 assay to assist pathologists and the reliability of the application in the SP263 assay. A robust high-performance DL model for automated tumor detection was devised with an accuracy and specificity of 0.9326 and 0.9641, respectively, and a concrete TPS value was obtained after tumor cell segmentation. The TPS comparison test in the 22c3 assay showed strong consistency between the TPS calculated with the AI system and trained pathologists (R = 0.9429-0.9458). AI-assisted diagnosis test confirmed that the repeatability and efficiency of untrained pathologists could be improved using the AI system. The Ventana PD-L1 (SP263) assay showed high consistency in TPS calculations between the AI system and pathologists (R = 0.9787). In conclusion, a high-precision AI system is proposed for the automated TPS assessment of PD-L1 expression in the 22c3 and SP263 assays in NSCLC. Our study also indicates the benefits of using an AI-assisted system to improve diagnostic repeatability and efficiency for pathologists.
Collapse
|
30
|
Saleh RR, Scott JL, Meti N, Perlon D, Fazelzad R, Ocana A, Amir E. Prognostic Value of Programmed Death Ligand-1 Expression in Solid Tumors Irrespective of Immunotherapy Exposure: A Systematic Review and Meta-Analysis. Mol Diagn Ther 2022; 26:153-168. [PMID: 35106739 DOI: 10.1007/s40291-022-00576-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND The programmed cell death-1/programmed cell death ligand-1 (PD-L1) pathway, which plays a crucial role in cancer immune surveillance, is the target of several approved immunotherapeutic agents and is used as a predictive biomarker in some solid tumors. However, its use as a prognostic marker (i.e., regardless of therapy used) is not established clearly with available data demonstrating inconsistent prognostic impact of PD-L1 expression in solid tumors. METHODS We conducted a systematic literature search of electronic databases and identified publications exploring the effect of PD-L1 expression on overall survival and/or disease-free survival. Hazard ratios were pooled in a meta-analysis using generic inverse-variance and random-effects modeling. We used the Deeks method to explore subgroup differences based on disease site, stage of disease, and method of PD-L1 quantification. RESULTS One hundred and eighty-six studies met the inclusion criteria. Programmed cell death ligand-1 expression was associated with worse overall survival (hazard ratio 1.33, 95% confidence interval 1.26-1.39; p < 0.001). There was significant heterogeneity between disease sites (subgroup p = 0.002) with pancreatic, hepatocellular, and genitourinary cancers associated with the highest magnitude of adverse outcomes. Programmed cell death ligand-1 was also associated with worse overall disease-free survival (hazard ratio 1.19, 95% confidence interval 1.09-1.30; p < 0.001). Stage of disease did not significantly affect the results (subgroup p = 0.52), nor did the method of quantification via immunohistochemistry or messenger RNA (subgroup p = 0.70). CONCLUSIONS High expression of PD-L1 is associated with worse survival in solid tumors albeit with significant heterogeneity among tumor types. The effect is consistent in early-stage and metastatic disease and is not sensitive to method of PD-L1 quantification. These data can provide additional information for the counseling of patients with cancer about prognosis.
Collapse
Affiliation(s)
- Ramy R Saleh
- Department of Medical Oncology, McGill University, Montreal, QC, Canada
| | - Jordan L Scott
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada
| | - Nicholas Meti
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada
| | - Danielle Perlon
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada
| | - Rouhi Fazelzad
- Information Specialist, Library and Information Services, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Alberto Ocana
- Hospital Clinico San Carlos and Instituto de Investigación Sanitaria San Carlos (IdISSC), and Centro Regional de Investigaciones Biomedicas (CRIB), Centro de Investigación Biomédica en Red Cáncerci (CIBERONC), Universidad Castilla La Mancha (UCLM), Madrid, Spain
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
31
|
Campelos S, Polónia A, Caramelo A, Curado M, Eloy C. p40/PD-L1 and TTF1/PD-L1 Immunohistochemical Double Staining Contributes to Intraindividual Variability Associated With PD-L1 Staining Interpretation in Lung Cancer Samples. Appl Immunohistochem Mol Morphol 2022; 30:165-170. [PMID: 35262521 DOI: 10.1097/pai.0000000000000999] [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: 12/30/2020] [Accepted: 11/08/2021] [Indexed: 12/24/2022]
Abstract
The important developments achieved in recent years with a consequent paradigm shift in the treatment of non-small cell lung cancer (NSCLC), including the latest immune checkpoint inhibitors, have led to an increasing need to optimize the scarce material usually available in the diagnosis of these tumors. In this sense, this study intends to evaluate the performance of double immunohistochemistry (IHC) in comparison to simple IHC for programmed death-ligand 1 (PD-L1) evaluation with 22C3 clone for selection to therapy with pembrolizumab. For that, 38 histologic samples of NSCLC small biopsies sent to our laboratory were selected. Double IHC were performed with the doublets TTF1/PD-L1 and p40/PD-L1, after all the usual diagnostic routine and molecular study was performed. The slides were interpreted by 2 independent pathologists and the results obtained were compared with each other and with the results obtained at diagnosis. A perfect agreement was observed when comparing the immunoexpression of TTF1 and p40 in double IHC in relation to single IHC. Although the agreement was substantial in the analysis of the positive/negative PD-L1 IHC (81.6% to 92.1%; κ=0.610 to 0.829) and in the analysis of the 50% cut-off (86.8% to 89.5%; κ=0.704 to 0.759), it fell short of the expected and desirable agreement for a biomarker such as PD-L1, since this result will have a major role in the institution of a treatment. In conclusion, this small series does not allow us to recommend this methodology for the evaluation of the PD-L1 biomarker in double staining IHC with the 22C3 clone for therapy selection.
Collapse
Affiliation(s)
- Sofia Campelos
- Laboratory of Pathology, Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto
- Institute for Research and Innovation in Health, University of Porto
| | - António Polónia
- Laboratory of Pathology, Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto
- Institute for Research and Innovation in Health, University of Porto
| | - Ana Caramelo
- Laboratory of Pathology, Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto
- Institute for Research and Innovation in Health, University of Porto
| | - Mónica Curado
- Laboratory of Pathology, Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto
- Institute for Research and Innovation in Health, University of Porto
| | - Catarina Eloy
- Laboratory of Pathology, Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto
- Institute for Research and Innovation in Health, University of Porto
- FMUP-Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
32
|
Xu Q, Cha Q, Qin H, Liu B, Wu X, Shi J. Identification of Master Regulators Driving Disease Progression, Relapse, and Drug Resistance in Lung Adenocarcinoma. FRONTIERS IN BIOINFORMATICS 2022; 2:813960. [PMID: 36304306 PMCID: PMC9580914 DOI: 10.3389/fbinf.2022.813960] [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: 11/15/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Backgrounds: Lung cancer is the leading cause of cancer related death worldwide. Current treatment strategies primarily involve surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy, determined by TNM stages, histologic types, and genetic profiles. Plenty of studies have been trying to identify robust prognostic gene expression signatures. Even for high performance signatures, they usually have few shared genes. This is not totally unexpected, since a prognostic signature is associated with patient survival and may contain no upstream regulators. Identification of master regulators driving disease progression is a vital step to understand underlying molecular mechanisms and develop new treatments. Methods: In this study, we have utilized a robust workflow to identify potential master regulators that drive poor prognosis in patients with lung adenocarcinoma. This workflow takes gene expression signatures that are associated with poor survival of early-stage lung adenocarcinoma, EGFR-TKI resistance, and responses to immune checkpoint inhibitors, respectively, and identifies recurrent master regulators from seven public gene expression datasets by a regulatory network-based approach. Results: We have found that majority of the master regulators driving poor prognosis in early stage LUAD are cell-cycle related according to Gene Ontology annotation. However, they were demonstrated experimentally to promote a spectrum of processes such as tumor cell proliferation, invasion, metastasis, and drug resistance. Master regulators predicted from EGFR-TKI resistance signature and the EMT pathway signature are largely shared, which suggests that EMT pathway functions as a hub and interact with other pathways such as hypoxia, angiogenesis, TNF-α signaling, inflammation, TNF-β signaling, Wnt, and Notch signaling pathways. Master regulators that repress immunotherapy are enriched with MYC targets, E2F targets, oxidative phosphorylation, and mTOR signaling. Conclusion: Our study uncovered possible mechanisms underlying recurrence, resistance to targeted therapy, and immunotherapy. The predicted master regulators may serve as potential therapeutic targets in patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Qiong Xu
- Department of Respiratory Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiongfang Cha
- Department of Respiratory Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Qin
- Department of Respiratory Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Liu
- Department of Respiratory Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueling Wu
- Department of Respiratory Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xueling Wu, ; Jiantao Shi,
| | - Jiantao Shi
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Xueling Wu, ; Jiantao Shi,
| |
Collapse
|
33
|
Rizzo A, Ricci AD, Di Federico A, Frega G, Palloni A, Tavolari S, Brandi G. Predictive Biomarkers for Checkpoint Inhibitor-Based Immunotherapy in Hepatocellular Carcinoma: Where Do We Stand? Front Oncol 2022; 11:803133. [PMID: 34976841 PMCID: PMC8718608 DOI: 10.3389/fonc.2021.803133] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) remains the sixth most commonly diagnosed malignancy worldwide, still representing an important cause of cancer-related death. Over the next few years, novel systemic treatment options have emerged. Among these, immune checkpoint inhibitors (ICIs) have been widely evaluated and are under assessment, as monotherapy or in combination with other anticancer agents in treatment-naïve and previously treated patients. In particular, the approval of the PD-L1 inhibitor atezolizumab plus the antiangiogenic agent bevacizumab as front-line treatment for advanced HCC has led to the adoption of this combination in this setting, and the IMbrave 150 phase III trial has established a novel standard of care. However, several questions remain unanswered, including the identification of reliable predictors of response to ICIs in HCC patients. In the current paper, we will provide an updated overview of potentially useful predictive biomarkers of response to immunotherapy in advanced HCC. A literature search was conducted in September 2021 of Pubmed/Medline, Cochrane library and Scopus databases.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Angela Dalia Ricci
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Giorgio Frega
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Palloni
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Simona Tavolari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
34
|
Liu D, Gao S, Zhai Y, Yang X, Zhai G. Research progress of tumor targeted drug delivery based on PD-1/PD-L1. Int J Pharm 2022; 616:121527. [DOI: 10.1016/j.ijpharm.2022.121527] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 12/16/2022]
|
35
|
Kumar S, Pandey M, Mir IA, Mukhopadhyay A, Sharawat SK, Jain D, Saikia J, Malik PS, Kumar S, Mohan A. Evaluation of the programmed death-ligand 1 mRNA expression and immunopositivity and their correlation with survival outcomes in Indian lung cancer patients. Hum Cell 2021; 35:286-298. [PMID: 34786661 DOI: 10.1007/s13577-021-00647-4] [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: 09/23/2021] [Accepted: 11/11/2021] [Indexed: 11/26/2022]
Abstract
The presence of membranous immunopositivity of programmed death-ligand 1 (PD-L1) in tumors serves as a key determinant of response to immune checkpoint inhibitors. However, there are very limited studies on the evaluation of the PD-L1 mRNA expression and immunopositivity and their correlation with therapeutic response and survival outcomes, especially in Indian lung cancer patients. In this prospective study, conducted between 2017 and 2020, we collected biopsies and surgically resected tumors from 173 lung cancer patients. PD-L1 immunopositivity and mRNA expression were determined by immunohistochemistry using SP263 assay and qRT-PCR, respectively. PD-L1 expression was correlated with various clinicopathological variables, response to therapy, and survival outcomes using appropriate statistical methods. The median age was 60 years (range 33-81 years) with the majority of patients being male (86.5%) and smokers (83%). Histologically, the majority of patients were non-small cell lung cancer (89.4%) and of squamous cell carcinoma histology (64.3%). PD-L1 immunopositivity in tumor cells (tumor proportion score (TPS) ≥ 1%) was detected in 37.6%, while high immunopositivity (TPS ≥ 50%) was detected in 16.8% of lung cancer patients. Almost 76% of lung cancer patients with PD-L1 TPS ≥ 50% belonged to PD-L1 mRNA high-expression group. PD-L1 mRNA expression and immunopositivity did not correlate with response to therapy and survival outcomes. We conclude that PD-L1 immunopositivity and mRNA expression do not seem to serve as a prognostic biomarker for lung cancer patients treated with chemotherapy. More prospective studies should be planned to evaluate the predictive and prognostic relevance of PD-L1 expression in Indian lung cancer patients being treated with immune checkpoint inhibitors.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/mortality
- Female
- Gene Expression
- Humans
- Immune Checkpoint Inhibitors/therapeutic use
- Immunohistochemistry
- India/epidemiology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/mortality
- Male
- Middle Aged
- Prospective Studies
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Survival Rate
- Time Factors
Collapse
Affiliation(s)
- Sachin Kumar
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| | - Monu Pandey
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Ishfaq A Mir
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Abhirup Mukhopadhyay
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Surender K Sharawat
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Jyoutishman Saikia
- Department of Surgical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Prabhat S Malik
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sunil Kumar
- Department of Surgical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Anant Mohan
- Department of Pulmonary Critical Care and Sleep Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| |
Collapse
|
36
|
Umar MI, Hassan W, Murtaza G, Buabeid M, Arafa E, Irfan HM, Asmawi MZ, Huang X. The Adipokine Component in the Molecular Regulation of Cancer Cell Survival, Proliferation and Metastasis. Pathol Oncol Res 2021; 27:1609828. [PMID: 34588926 PMCID: PMC8473628 DOI: 10.3389/pore.2021.1609828] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 12/22/2022]
Abstract
A hormonal imbalance may disrupt the rigorously monitored cellular microenvironment by hampering the natural homeostatic mechanisms. The most common example of such hormonal glitch could be seen in obesity where the uprise in adipokine levels is in virtue of the expanding bulk of adipose tissue. Such aberrant endocrine signaling disrupts the regulation of cellular fate, rendering the cells to live in a tumor supportive microenvironment. Previously, it was believed that the adipokines support cancer proliferation and metastasis with no direct involvement in neoplastic transformations and tumorigenesis. However, the recent studies have reported discrete mechanisms that establish the direct involvement of adipokine signaling in tumorigenesis. Moreover, the individual adipokine profile of the patients has never been considered in the prognosis and staging of the disease. Hence, the present manuscript has focused on the reported extensive mechanisms that culminate the basis of poor prognosis and diminished survival rate in obese cancer patients.
Collapse
Affiliation(s)
| | - Waseem Hassan
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Elshaimaa Arafa
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | | | - Mohd Zaini Asmawi
- School of Pharmaceutical Sciences, University of Science Malaysia, Pulau Pinang, Malaysia
| | - Xianju Huang
- College of Pharmacy, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
37
|
Gao H, Wu Y, Shi J, Zhang X, Liu T, Hu B, Jia B, Wan Y, Liu Z, Wang F. Nuclear imaging-guided PD-L1 blockade therapy increases effectiveness of cancer immunotherapy. J Immunother Cancer 2021; 8:jitc-2020-001156. [PMID: 33203663 PMCID: PMC7674096 DOI: 10.1136/jitc-2020-001156] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 02/06/2023] Open
Abstract
Objectives Strategies to improve the responsiveness of programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) checkpoint blockade therapy remain an essential topic in cancer immunotherapy. In this study, we developed a new radiolabeled nanobody-based imaging probe 99mTc-MY1523 targeting PD-L1 for the enhanced therapeutic efficacy of PD-L1 blockade immunotherapy by the guidance of 99mTc-MY1523 SPECT/CT imaging. Methods The binding affinity and specificity of nanobody MY1523 were measured in vitro. MY1523 was radiolabeled with 99mTc by a site-specific transpeptidation of Sortase-A, and the biodistribution and single photon emission CT (SPECT)/CT were performed in mice bearing different tumors. We used interferon-γ (IFN-γ) as an intervention means to establish animal models with different levels of PD-L1 expression, then investigated the ability of 99mTc-MY1523 SPECT/CT for the in vivo non-invasive measurement of PD-L1 expression in tumors. Finally, the PD-L1 blockade immunotherapies guided by 99mTc-MY1523 SPECT/CT were carried out in MC-38, A20, and 4T1 tumor-bearing mouse models, followed by the testing of tumor infiltration T cells. Results MY1523 exhibited a high binding affinity and specificity to PD-L1 and had no competitive binding with the therapeutic antibody. 99mTc-MY1523 was prepared with high specific activity and radiochemical purity. It was found that tumor PD-L1 expression was dynamically upregulated by IFN-γ intervention in MC-38, A20, and 4T1 tumor-bearing mouse models, as indicated by 99mTc-MY1523 SPECT/CT. The PD-L1 blockade therapy initiated during the therapeutic time window determined by 99mTc-MY1523 SPECT/CT imaging significantly enhanced the therapeutic efficacy in all animal models, while the tumor growth was effectively suppressed, and the survival time of mice was evidently prolonged. A correlation between dynamically upregulated PD-L1 expression and improved PD-L1 blockade therapy effectiveness was revealed, and the markedly increased infiltration of effector T cells into tumors was verified after the imaging-guided therapy. Conclusion Our results demonstrated that 99mTc-MY1523 SPECT/CT allowed a real-time, quantitative and dynamic mapping of PD-L1 expression in vivo, and the imaging-guided PD-L1 blockade immunotherapy significantly enhanced the therapeutic efficacy. This strategy merits translation into clinical practice for the better management of combination therapies with radiotherapy or chemotherapy.
Collapse
Affiliation(s)
- Hannan Gao
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yue Wu
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiyun Shi
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xin Zhang
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Tianyu Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Biao Hu
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yakun Wan
- Shanghai Novamab Biopharmaceuticals Co., Ltd, Shanghai, China
| | - Zhaofei Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Fan Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China .,Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
38
|
Rizzo A, Ricci AD. PD-L1, TMB, and other potential predictors of response to immunotherapy for hepatocellular carcinoma: how can they assist drug clinical trials? Expert Opin Investig Drugs 2021; 31:415-423. [PMID: 34429006 DOI: 10.1080/13543784.2021.1972969] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) represents the sixth most commonly diagnosed malignancy worldwide, accounting for millions of deaths annually. Despite immune checkpoint inhibitors (ICIs) reported important results, only a minority of HCC patients benefit from these treatments, and the identification of predictive biomarkers of response still remains a highly unmet need. AREAS COVERED Herein, we provide a timely overview of available evidence on biochemical predictors of response to immunotherapy in advanced HCC patients; we speculate on how PD-L1, TMB, and other emerging biomarkers could assist drug clinical trials in the near future. A literature search was conducted in June 2021 using Pubmed/Medline, Cochrane library, and Scopus databases. EXPERT OPINION Reliable predictors of response to ICIs are of pivotal importance to allow a proper stratification and selection of HCC patients that could derive more benefit from immunotherapy. Well-designed, multicenter clinical trials specifically focused on predictive biomarkers are warranted in this setting, where most of evidence currently derives from retrospective, single-center studies with small sample size.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Medical Oncology, Irccs Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italia
| | - Angela Dalia Ricci
- Medical Oncology, Irccs Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italia
| |
Collapse
|
39
|
Choi Y, Kwon O, Choi CM, Jeong MK. A Pilot Study of Whether the Cold-Heat Syndrome Type is Associated with Treatment Response and Immune Status in Patients with Non-Small Cell Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:9920469. [PMID: 34239594 PMCID: PMC8241512 DOI: 10.1155/2021/9920469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/04/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022]
Abstract
The cold-heat syndrome type (ZHENG) is one of the essential elements of syndrome differentiation in East Asian Medicine. This pilot study aimed to explore the characteristics of non-small cell lung cancer (NSCLC) patients treated with immune checkpoint inhibitors (ICIs) based on the cold-heat syndrome type. Twenty NSCLC patients treated with ICI monotherapy were included in the study and completed the cold-heat syndrome differentiation questionnaire. Demographic and clinical characteristics of the included patients were obtained through electronic medical records. Additionally, blood samples of 10 patients were analyzed with cytokine level and immune profiling. Patients were divided into two groups of cold type (n = 9) and non-cold type (n = 11), according to the cold symptoms questionnaire's cutoff point. No significant difference between the two groups was observed in clinical response to ICIs (p=0.668). Progression-free survival (PFS) seemed to be longer in patients with non-cold type than cold type (p=0.332). In patients with adenocarcinoma, the non-cold type showed longer PFS than the cold type (p=0.036). Also, there were more patients with PD-L1 negative in the cold type compared to the non-cold type (p=0.050). In immune profiling, the proportion of effector memory CD8 T-cells was higher in patients with cold type than with non-cold type (p=0.015), and the proportion of terminal effector CD8 T-cells was lower in patients with cold type than with non-cold type (p=0.005). This pilot study has shown the potential for differences in prognosis and immune status between patients with cold and non-cold types. Hopefully, it provides essential information and insight into NSCLC patients' characteristics from the perspective of syndrome differentiation. Further large-scale observational studies and intervention studies are required.
Collapse
Affiliation(s)
- Yujin Choi
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Ojin Kwon
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Chang-Min Choi
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Mi-Kyung Jeong
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| |
Collapse
|
40
|
Kern R, Panis C. CTLA-4 Expression and Its Clinical Significance in Breast Cancer. Arch Immunol Ther Exp (Warsz) 2021; 69:16. [PMID: 34148159 DOI: 10.1007/s00005-021-00618-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022]
Abstract
Breast cancer is the leading cause of women's death among all cancers. The main reason associated with this is the development of metastasis and therapy-resistant breast carcinoma (BC), which pose the main challenge of oncology nowadays. Evidence suggest that these tumors seem to have inhibitory mechanisms that may favor their progression and surveillance. Cancer cells can evade antitumor T cell responses by expressing some immune inhibitory molecules such as the cytotoxic T-lymphocyte antigen-4 (CTLA-4), whose clinical meaning has emerged in the last few years and is poorly understood in the BC context. This systematic literature review aims at identifying studies on CTLA-4 expression in BC, and address what is known about its clinical meaning. A literature search was performed in PubMed and LILACS databases, using the MESH terms "breast cancer"; "CTLA-4 Antigen/antagonists and inhibitors"; and "Lymphocytes, Tumor-Infiltrating/immunology", published in the last 10 years. In total, 12 studies were included in this review. Systematic review used the Preferred Reporting Items for Systematic Reviews and Meta-Analyses. Despite the small number of eligible studies, the literature reports some associations between CTLA-4 expression in the tumor microenvironment and worse BC outcomes, regardless of its molecular subtype. CTLA-4 expression in BC is a putative marker of clinical significance and a rationale therapeutic target in the emerging field of immunotherapy.
Collapse
Affiliation(s)
- Rodrigo Kern
- Laboratory of Tumor Biology, State University of West Paraná, UNIOESTE, Francisco Beltrão, Brazil
| | - Carolina Panis
- Laboratory of Tumor Biology, State University of West Paraná, UNIOESTE, Francisco Beltrão, Brazil.
- State University of Western Paraná, Health Sciences Center, Vitório Traiano Highway, Km 2, Francisco Beltrão, PR, Brazil.
| |
Collapse
|
41
|
Challenges and Opportunities in the Statistical Analysis of Multiplex Immunofluorescence Data. Cancers (Basel) 2021; 13:cancers13123031. [PMID: 34204319 PMCID: PMC8233801 DOI: 10.3390/cancers13123031] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Immune modulation is considered a hallmark of cancer initiation and progression, and has offered promising opportunities for therapeutic manipulation. Multiplex immunofluorescence (mIF) technology has enabled the tumor immune microenvironment (TIME) to be studied at an increased scale, in terms of both the number of markers and the number of samples. Another benefit of mIF technology is the ability to measure not only the abundance but also the spatial location of multiple cells types within a tissue sample simultaneously, allowing for assessment of the co-localization of different types of immune markers. Thus, the use of mIF technologies have enable researchers to characterize patient, clinical, and tumor characteristics in the hope of identifying patients whom might benefit from immunotherapy treatments. In this review we outline some of the challenges and opportunities in the statistical analyses of mIF data to study the TIME. Abstract Immune modulation is considered a hallmark of cancer initiation and progression. The recent development of immunotherapies has ushered in a new era of cancer treatment. These therapeutics have led to revolutionary breakthroughs; however, the efficacy of immunotherapy has been modest and is often restricted to a subset of patients. Hence, identification of which cancer patients will benefit from immunotherapy is essential. Multiplex immunofluorescence (mIF) microscopy allows for the assessment and visualization of the tumor immune microenvironment (TIME). The data output following image and machine learning analyses for cell segmenting and phenotyping consists of the following information for each tumor sample: the number of positive cells for each marker and phenotype(s) of interest, number of total cells, percent of positive cells for each marker, and spatial locations for all measured cells. There are many challenges in the analysis of mIF data, including many tissue samples with zero positive cells or “zero-inflated” data, repeated measurements from multiple TMA cores or tissue slides per subject, and spatial analyses to determine the level of clustering and co-localization between the cell types in the TIME. In this review paper, we will discuss the challenges in the statistical analysis of mIF data and opportunities for further research.
Collapse
|
42
|
Jain E, Sharma S, Aggarwal A, Bhardwaj N, Dewan A, Kumar A, Jain D, Bhattacharya M, Saurav GK, Kini L, Mohanty SK. PD-L1 expression and its clinicopathologic and genomic correlation in the non-small cell lung carcinoma patients: An Indian perspective. Pathol Res Pract 2021; 228:153497. [PMID: 34053784 DOI: 10.1016/j.prp.2021.153497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Immunotherapy with checkpoint inhibitor programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) antibodies targeting the cellular immune checkpoints is the present area of interest showing promising results in patients with advanced non-small cell lung cancer (NSCLC). As there is paucity of PD-L1 expression data from the Indian perspective, we studied the correlation of clinicopathologic profile and oncogenic driver mutations in these patients. MATERIALS AND METHODS Samples from 252 advanced NSCLCs patients were studied for PD-L1 expression through immunohistochemistry using rabbit anti-human PD-L1 monoclonal antibody (clone SP263) on Ventana BenchMark ULTRA autostainer. Simultaneously, genetic mutations were studied by next generation sequencing (for EGFR, ALK, ROS, MET, and BRAF). PD-L1 expression was analyzed for association with clinicopathologic features and various mutations. RESULTS PD-L1 positivity was seen in 134 patients (53.2 %). It was twice more prevalent in males than females. No significant correlation was observed between PD-L1 expression with age, gender, site of testing (primary vs. metastatic tumors), smoking status, tumor laterality, stage, or histologic type; however, there was significant difference among solid and acinar types of adenocarcinoma combined together vs. other adenocarcinoma subtypes (p = 0.013), and well and moderately differentiated vs. poorly differentiated tumors (p = 0.022). When types/extent of PD-L1 positivity (≥25 %) were compared with demographics, clinical, and pathologic variables, significant differences were observed across the tumor grades (high-grade vs. low-grade) (p = 0.009) and stages (p = 0.039). The PD-L1 expression failed to demonstrate any statistical significance with oncogenic drivers. High PD-L1 expression (TPS ≥ 50) was observed in 27.6 % patients, and it was more prevalent in female patients (32.4 %), aged ≥60 years (33.8 %), smokers (27.3 %), poorly differentiated (36.8 %) and stage IV tumors (28.2 %). Exon 19 deletion was more prevalent in PD-L1 negative tumors whereas exon 21 substitution (L858R) was seen more in PD-L1 positive tumors. CONCLUSIONS This is the largest Indian study demonstrating PD-L1 expression in NSCLC patients comparing with clinicopathologic and genomic parameters. PD-L1 expression was significantly associated with high-grade, solid, and acinar types of adenocarcinoma and advanced tumors. High PD-L1 expression was more prevalent in female patients, aged ≥60 years, smokers, and poorly differentiated and stage IV tumors (28.2 %). Exon 19 deletion was more in PD-L1 negative tumors whereas exon 21 substitution (L858R) was more in PD-L1 positive tumors. PD-L1 is a potential predictive marker stratifying patients who benefit from PD-1 pathway-targeted therapy.
Collapse
Affiliation(s)
- Ekta Jain
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Shivani Sharma
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Aditi Aggarwal
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Nitin Bhardwaj
- Indian Council of Medical Research and National Institute of Malaria Research, New Delhi, 110029, India.
| | - Aditi Dewan
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Abhishek Kumar
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Deepika Jain
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Munmun Bhattacharya
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Gauraw Kumar Saurav
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Lata Kini
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| | - Sambit Kumar Mohanty
- Department of Pathology, CORE Diagnostics, 406, Udyog Vihar III, Gurgaon, Haryana 122001, India.
| |
Collapse
|
43
|
Tejerina E, Garca Tobar L, Echeveste JI, de Andrea CE, Vigliar E, Lozano MD. PD-L1 in Cytological Samples: A Review and a Practical Approach. Front Med (Lausanne) 2021; 8:668612. [PMID: 34026795 PMCID: PMC8139418 DOI: 10.3389/fmed.2021.668612] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/09/2021] [Indexed: 12/25/2022] Open
Abstract
With a growing number of predictive biomarkers needed to manage patients with non-small cell lung cancer (NSCLC), there has been a paradigm shift in care and handling of diagnostic samples. Among the various testing methods, immunohistochemistry (IHC) is the most cost- effective and widely available. Furthermore, over the past decade immunotherapy has emerged as one of the most promising cancer treatments. In this scenario IHC is the most used testing method available for PDL-1/PD1 immunotherapy. Several monoclonal antibodies targeting programmed death 1 (PD-1)/programmed death ligand-1 (PD-L1) pathways have been integrated into standard-of-care treatments of a wide range of cancer types, once provided evidence of PD-L1 expression in tumor cells by immunohistochemistry (IHC). Since currently available PD-L1 assays have been developed on formalin-fixed paraffin embedded (FFPE) histological specimens, a growing body of research is being dedicated to confirm the feasibility of applying PDL-1 assays also to cytological samples. Albeit promising results have been reported, several important issues still need to be addressed. Among these are the type of cytological samples, pre-analytical issues, cyto-histological correlation, and inter-observer agreement. This review briefly summarizes the knowledge of the role of cytopathology in the analysis of PD-L1 by immunocytochemistry (ICC) and future directions of cytopathology in the immunotherapy setting.
Collapse
Affiliation(s)
- Eva Tejerina
- Department of Pathology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Laura Garca Tobar
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain
| | - Jos I Echeveste
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain
| | | | - Elena Vigliar
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Mara D Lozano
- Department of Pathology, Clinica University of Navarra, Pamplona, Spain
| |
Collapse
|
44
|
Hofman P. Next-Generation Sequencing with Liquid Biopsies from Treatment-Naïve Non-Small Cell Lung Carcinoma Patients. Cancers (Basel) 2021; 13:2049. [PMID: 33922637 PMCID: PMC8122958 DOI: 10.3390/cancers13092049] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 12/16/2022] Open
Abstract
Recently, the liquid biopsy (LB), a non-invasive and easy to repeat approach, has started to compete with the tissue biopsy (TB) for detection of targets for administration of therapeutic strategies for patients with advanced stages of lung cancer at tumor progression. A LB at diagnosis of late stage non-small cell lung carcinoma (NSCLC) is also being performed. It may be asked if a LB can be complementary (according to the clinical presentation or systematics) or even an alternative to a TB for treatment-naïve advanced NSCLC patients. Nucleic acid analysis with a TB by next-generation sequencing (NGS) is gradually replacing targeted sequencing methods for assessment of genomic alterations in lung cancer patients with tumor progression, but also at baseline. However, LB is still not often used in daily practice for NGS. This review addresses different aspects relating to the use of LB for NGS at diagnosis in advanced NSCLC, including its advantages and limitations.
Collapse
Affiliation(s)
- Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Université Côte d’Azur, CHU Nice, FHU OncoAge, Pasteur Hospital, 30 avenue de la voie romaine, BP69, CEDEX 01, 06001 Nice, France; ; Tel.: +33-4-92-03-88-55 or +33-4-92-03-87-49; Fax: +33-4-92-88-50
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d’Azur, CHU Nice, FHU OncoAge, 06001 Nice, France
| |
Collapse
|
45
|
Kuksin M, Morel D, Aglave M, Danlos FX, Marabelle A, Zinovyev A, Gautheret D, Verlingue L. Applications of single-cell and bulk RNA sequencing in onco-immunology. Eur J Cancer 2021; 149:193-210. [PMID: 33866228 DOI: 10.1016/j.ejca.2021.03.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 02/08/2023]
Abstract
The rising interest for precise characterization of the tumour immune contexture has recently brought forward the high potential of RNA sequencing (RNA-seq) in identifying molecular mechanisms engaged in the response to immunotherapy. In this review, we provide an overview of the major principles of single-cell and conventional (bulk) RNA-seq applied to onco-immunology. We describe standard preprocessing and statistical analyses of data obtained from such techniques and highlight some computational challenges relative to the sequencing of individual cells. We notably provide examples of gene expression analyses such as differential expression analysis, dimensionality reduction, clustering and enrichment analysis. Additionally, we used public data sets to exemplify how deconvolution algorithms can identify and quantify multiple immune subpopulations from either bulk or single-cell RNA-seq. We give examples of machine and deep learning models used to predict patient outcomes and treatment effect from high-dimensional data. Finally, we balance the strengths and weaknesses of single-cell and bulk RNA-seq regarding their applications in the clinic.
Collapse
Affiliation(s)
- Maria Kuksin
- ENS de Lyon, 15 Parvis René Descartes, 69007, Lyon, France; Département d'Innovations Thérapeutiques et Essais Précoces (DITEP), Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France
| | - Daphné Morel
- Département d'Innovations Thérapeutiques et Essais Précoces (DITEP), Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France; Département de Radiothérapie, Gustave Roussy Cancer Campus, Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France; INSERM UMR1030, Molecular Radiotherapy and Therapeutic Innovations, Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
| | - Marine Aglave
- INSERM US23, CNRS UMS 3655, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France
| | | | - Aurélien Marabelle
- Département d'Innovations Thérapeutiques et Essais Précoces (DITEP), Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France; INSERM U1015, Gustave Roussy, Université Paris Saclay, France
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, F-75005, Paris, France; INSERM, U900, F-75005, Paris, France; MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, F-75006, Paris, France; Laboratory of Advanced Methods for High-dimensional Data Analysis, Lobachevsky University, 603000, Nizhny Novgorod, Russia
| | - Daniel Gautheret
- Institute for Integrative Biology of the Cell, UMR 9198, CEA, CNRS, Université Paris-Saclay, Gif-Sur-Yvette, France; IHU PRISM, Gustave Roussy Cancer Campus, Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France; Université Paris-Saclay, France
| | - Loïc Verlingue
- Département d'Innovations Thérapeutiques et Essais Précoces (DITEP), Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France; INSERM UMR1030, Molecular Radiotherapy and Therapeutic Innovations, Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France; Institut Curie, PSL Research University, F-75005, Paris, France; Université Paris-Saclay, France.
| |
Collapse
|
46
|
Kim H, Hong JY, Lee J, Park SH, Park JO, Park YS, Lim HY, Kang WK, Kim KM, Kim ST. Clinical sequencing to assess tumor mutational burden as a useful biomarker to immunotherapy in various solid tumors. Ther Adv Med Oncol 2021; 13:1758835921992992. [PMID: 33717226 PMCID: PMC7917846 DOI: 10.1177/1758835921992992] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/13/2021] [Indexed: 01/05/2023] Open
Abstract
Background: Immune checkpoint inhibitors (ICIs) have become established as a new
therapeutic paradigm in various solid cancers. Predictive biomarkers to ICIs
have not yet been fully established. Tumor mutational burden (TMB) has been
considered as a useful marker to indicate patients who benefit from
ICIs. Methods: We performed next-generation sequencing, including TMB analysis, as a routine
clinical practice in 501 patients with advanced gastrointestinal (GI),
genitourinary (GU), or rare cancers. The TruSight™ Oncology 500
assay from Illumina was used as a cancer panel. Results: In total, 11.6% (58/501) were identified with tumors with high TMB and
MSI-high status was confirmed in seven out of 501 cases (1.4%). High TMB was
observed in 11.6% of patients with various solid tumors, including: GU
cancers (36.0%, 9/25), colorectal cancer (15.2%, 23/151), biliary tract
cancer (14.6%, 7/48), melanoma (14.3%, 3/21), gastric cancer (11.2%,
13/116), hepatocellular carcinoma (8.3%, 1/12), other GI tract cancers
(4.5%, 1/22), and sarcoma (1.7%, 1/60). The objective response rate (ORR) to
ICIs was 75% (nine out of 12) in solid tumor patients with high TMB and 25%
(30 out of 40) in those with non-high TMB. Patients with high TMB had better
ORR to ICIs than those with non-high TMB (p = 0.004).
Univariate analysis revealed that the status of PD-L1 expression and of TMB
(high versus non-high) had significant association in
response to ICIs. However, in multivariate analysis, the status of TMB (high
versus non-high) was only significantly related to the
response to ICIs (p = 0.036). Conclusion: In the present study, we analyzed the TMB using a cancer panel for various
solid tumor patients in routine clinical practice and also demonstrated the
usefulness of TMB to predict the efficacy for ICIs.
Collapse
Affiliation(s)
- Hana Kim
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Korea
| | - Jung Yong Hong
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Korea
| | - Jeeyun Lee
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Korea
| | - Se Hoon Park
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Korea
| | - Joon Oh Park
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Korea
| | - Young Suk Park
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Korea
| | - Ho Yeong Lim
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Korea
| | - Won Ki Kang
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Korea
| | - Seung Tae Kim
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| |
Collapse
|
47
|
Muñoz-San Martín C, Gamella M, Pedrero M, Montero-Calle A, Pérez-Ginés V, Camps J, Arenas M, Barderas R, Pingarrón JM, Campuzano S. Anticipating metastasis through electrochemical immunosensing of tumor hypoxia biomarkers. Anal Bioanal Chem 2021; 414:399-412. [PMID: 33635388 DOI: 10.1007/s00216-021-03240-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/26/2022]
Abstract
Metastasis is responsible for about 90% of cancer-associated deaths. In the context of solid tumors, the low oxygen concentration in the tumor microenvironment (hypoxia) is one of the key factors contributing to metastasis. Tumor cells adapt to these conditions by overexpressing certain proteins such as programmed death ligand 1 (PD-L1) and hypoxia-inducible factor 1 alpha (HIF-1α). However, the determination of these tumor hypoxia markers that can be used to follow-up tumor progression and improve the efficiency of therapies has been scarcely addressed using electrochemical biosensors. In this work, we report the first electrochemical bioplatform for the determination of PD-L1 as well as the first one allowing its simultaneous determination with HIF-1α. The target proteins were captured and enzymatically labeled on magnetic microbeads and amperometric detection was undertaken on the surface of screen-printed dual carbon electrodes using the hydrogen peroxide/peroxidase/hydroquinone system. Sandwich immunoassays were implemented for both the HIF-1α and PD-L1 sensors and the analytical characteristics were evaluated providing LOD values of 86 and 279 pg mL-1 for the amperometric determination of PD-L1 and HIF-1α standards, respectively. The developed electrochemical immunoplatforms are competitive versus the only electrochemical immunosensor reported for the determination of HIF-1α and the "gold standard" ELISA methodology for the single determination of both proteins in terms of assay time, compatibility with the simultaneous determination of both proteins making their use suitable for untrained users at the point of attention. The dual amperometric immunosensor was applied to the simultaneous determination of HIF-1α and PD-L1 in cancer cell lysates. The analyses lasted only 2 h and just 0.5 μg of the sample was required.
Collapse
Affiliation(s)
- Cristina Muñoz-San Martín
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Maria Gamella
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - María Pedrero
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Ana Montero-Calle
- UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Víctor Pérez-Ginés
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovirai Virgili, 43204, Reus, Spain
| | - Meritxell Arenas
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovirai Virgili, 43204, Reus, Spain
| | - Rodrigo Barderas
- UFIEC, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
| | - José M Pingarrón
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Susana Campuzano
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| |
Collapse
|
48
|
Rizzo A, Brandi G. Biochemical predictors of response to immune checkpoint inhibitors in unresectable hepatocellular carcinoma. Cancer Treat Res Commun 2021; 27:100328. [PMID: 33549983 DOI: 10.1016/j.ctarc.2021.100328] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/28/2020] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) represents the most commonly diagnosed liver cancer worldwide, and the overall survival of patients with unresectable disease is poor. In the last five years, immune checkpoint inhibitors (ICIs) have revolutionized the treatment scenario of several hematological and solid tumors, and these agents have been actively explored in unresectable HCC. Firstly, promising findings of phase I and II clinical studies reporting durable responses and a tolerable safety profile have led to the assessment of ICIs as single agents in phase III clinical studies; however, the latter have provided controversial results, and the activity of ICI monotherapy seems limited to a small subgroup of patients. Conversely, the IMbrave150 trial recently showed that, among patients with previously untreated unresectable HCC, treatment with atezolizumab plus bevacizumab resulted in significantly longer overall survival and progression-free survival compared to sorafenib monotherapy. In addition, the activity of several other ICIs is under investigation, as combination immunotherapy as well as combinations of immunotherapy with antiangiogenic agents. Nonetheless, there are currently no validated predictive biomarkers able to guide treatment choice in this setting, where the identification of specific predictors of response to ICIs represents a major challenge. In this review, we aim to provide a critical overview of recent evidence on biochemical predictors of response to ICIs in patients with unresectable HCC, especially focusing on PD-L1, TMB, MSI, and other emerging biomarkers.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy; Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni, 15 Bologna, Italy.
| | - Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy; Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni, 15 Bologna, Italy
| |
Collapse
|
49
|
Mograbi B, Heeke S, Hofman P. The Importance of STK11/ LKB1 Assessment in Non-Small Cell Lung Carcinomas. Diagnostics (Basel) 2021; 11:196. [PMID: 33572782 PMCID: PMC7912095 DOI: 10.3390/diagnostics11020196] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Despite the recent implementation of immunotherapy as a single treatment or in combination with chemotherapy for first-line treatment of advanced non-small cell lung cancer (NSCLC), many patients do not benefit from this regimen due to primary treatment resistance or toxicity. Consequently, there is an urgent need to develop efficient biomarkers that can select patients who will benefit from immunotherapy thereby providing the appropriate treatment and avoiding toxicity. One of the biomarkers recently described for the stratification of NSCLC patients undergoing immunotherapy are mutations in STK11/LKB1, which are often associated with a lack of response to immunotherapy in some patients. Therefore, the purpose of this review is to describe the different cellular mechanisms associated with STK11/LKB1 mutations, which may explain the lack of response to immunotherapy. Moreover the review addresses the co-occurrence of additional mutations that may influence the response to immunotherapy and the current clinical studies that have further explored STK11/LKB1 as a predictive biomarker. Additionally this work includes the opportunities and limitations to look for the STK11/LKB1 status in the therapeutic strategy for NSCLC patients.
Collapse
Affiliation(s)
- Baharia Mograbi
- Centre Antoine Lacassagne, CNRS, FHU OncoAge, Team 4, INSERM, IRCAN, Université Côte d’Azur, 06000 Nice, France;
| | - Simon Heeke
- Department of Thoracic Head and Neck Medical Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Paul Hofman
- Centre Antoine Lacassagne, CNRS, FHU OncoAge, Team 4, INSERM, IRCAN, Université Côte d’Azur, 06000 Nice, France;
- CHU Nice, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, Pasteur Hospital, Université Côte d’Azur, 06000 Nice, France
- CHU Nice, FHU OncoAge, Hospital-Integrated Biobank BB-0033-00025, Université Côte d’Azur, 06000 Nice, France
| |
Collapse
|
50
|
Guarize J, Rocco EG, de Marinis F, Sedda G, Bertolaccini L, Donghi SM, Casiraghi M, Tonno CD, Barberis M, Spaggiari L. Prospective evaluation of EBUS-TBNA specimens for programmed death-ligand 1 expression in non-small cell lung cancer patients: a pilot study. J Bras Pneumol 2021; 47:e20200584. [PMID: 34259745 PMCID: PMC8332653 DOI: 10.36416/1806-3756/e20200584] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/06/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE EBUS-TBNA cytological sampling is routinely performed for pathological diagnosis, mediastinal staging, and molecular testing in lung cancer patients. EBUS-TBNA samples are not formally accepted for testing programmed death-ligand 1 (PD-L1) expression. The objective of the study was to compare the feasibility, reproducibility, and accuracy of PD-L1 expression assessment in cytological specimens and histological samples. METHODS We prospectively collected histological (transbronchial forceps biopsy) and cytological (EBUS-TBNA) samples from peribronchial neoplastic lesions during an endoscopic procedure at the same target lesion for the pathological diagnosis and molecular assessment of stage IV non-small cell lung cancer (NSCLC). RESULTS Fifteen patients underwent the procedure. Adequate cytological samples (at least 100 neoplastic cells) were obtained in 12 cases (92.3%). Assessment of PD-L1 expression was similar between histological and cytological samples (agreement rate = 92%). Sensitivity and diagnostic accuracy of EBUS-TBNA cytological specimens were 88.9% and 100%, respectively. CONCLUSIONS The evaluation of PD-L1 expression in EBUS-TBNA cytological specimens is feasible and presents good reproducibility when compared with routine histological samples. EBUS-TBNA cytological samples could be used for the assessment of PD-L1 expression in patients with NSCLC as a minimally invasive approach in stage IV NSCLC cancer patients.
Collapse
Affiliation(s)
- Juliana Guarize
- . Pneumologia Interventistica, Divisione di Chirurgia Toracica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
| | - Elena Guerini Rocco
- . Divisione di Anatomia Patologica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
| | - Filippo de Marinis
- . Divisione di Oncologia Toracica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
| | - Giulia Sedda
- . Divisione di Chirurgia Toracica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
| | - Luca Bertolaccini
- . Divisione di Chirurgia Toracica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
| | - Stefano Maria Donghi
- . Pneumologia Interventistica, Divisione di Chirurgia Toracica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
| | - Monica Casiraghi
- . Divisione di Chirurgia Toracica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
| | - Clementina Di Tonno
- . Divisione di Anatomia Patologica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
| | - Massimo Barberis
- . Divisione di Anatomia Patologica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
| | - Lorenzo Spaggiari
- . Divisione di Chirurgia Toracica, Istituto Europeo di Oncologia - IEO - Istituto di Ricovero e Cura a Carattere Scientifico - IRCCS - Milano, Italia
- . Dipartimento di Oncologia ed Emato-Oncologia, Università degli Studi di Milano, Milano, Italia
| |
Collapse
|