1
|
Chen H, Zuo H, Huang J, Liu J, Jiang L, Jiang C, Zhang S, Hu Q, Lai H, Yin B, Yang G, Mai G, Li B, Chi H. Unravelling infiltrating T-cell heterogeneity in kidney renal clear cell carcinoma: Integrative single-cell and spatial transcriptomic profiling. J Cell Mol Med 2024; 28:e18403. [PMID: 39031800 PMCID: PMC11190954 DOI: 10.1111/jcmm.18403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 07/15/2024] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) pathogenesis intricately involves immune system dynamics, particularly the role of T cells within the tumour microenvironment. Through a multifaceted approach encompassing single-cell RNA sequencing, spatial transcriptome analysis and bulk transcriptome profiling, we systematically explored the contribution of infiltrating T cells to KIRC heterogeneity. Employing high-density weighted gene co-expression network analysis (hdWGCNA), module scoring and machine learning, we identified a distinct signature of infiltrating T cell-associated genes (ITSGs). Spatial transcriptomic data were analysed using robust cell type decomposition (RCTD) to uncover spatial interactions. Further analyses included enrichment assessments, immune infiltration evaluations and drug susceptibility predictions. Experimental validation involved PCR experiments, CCK-8 assays, plate cloning assays, wound-healing assays and Transwell assays. Six subpopulations of infiltrating and proliferating T cells were identified in KIRC, with notable dynamics observed in mid- to late-stage disease progression. Spatial analysis revealed significant correlations between T cells and epithelial cells across varying distances within the tumour microenvironment. The ITSG-based prognostic model demonstrated robust predictive capabilities, implicating these genes in immune modulation and metabolic pathways and offering prognostic insights into drug sensitivity for 12 KIRC treatment agents. Experimental validation underscored the functional relevance of PPIB in KIRC cell proliferation, invasion and migration. Our study comprehensively characterizes infiltrating T-cell heterogeneity in KIRC using single-cell RNA sequencing and spatial transcriptome data. The stable prognostic model based on ITSGs unveils infiltrating T cells' prognostic potential, shedding light on the immune microenvironment and offering avenues for personalized treatment and immunotherapy.
Collapse
Affiliation(s)
- Haiqing Chen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Haoyuan Zuo
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- Department of General Surgery (Hepatopancreatobiliary Surgery)Deyang People's HospitalDeyangChina
| | - Jinbang Huang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Jie Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- Department of General SurgeryDazhou Central HospitalDazhouChina
| | - Lai Jiang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Chenglu Jiang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Shengke Zhang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Qingwen Hu
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Haotian Lai
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Bangchao Yin
- Department of PathologySixth People's Hospital of YibinYibinChina
| | - Guanhu Yang
- Department of Specialty MedicineOhio UniversityAthensOhioUSA
| | - Gang Mai
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- Department of General Surgery (Hepatopancreatobiliary Surgery)Deyang People's HospitalDeyangChina
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Hao Chi
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| |
Collapse
|
2
|
Lee CK, Lee JB, Park SJ, Che J, Kwon WS, Kim HS, Jung M, Lee S, Park SR, Koo DH, Lee HW, Bae WK, Jeung HC, Hwang IG, Kim H, Nam CM, Chung HC, Rha SY. Second-line chemoimmunotherapy with nivolumab and paclitaxel in immune-related biomarker-enriched advanced gastric cancer: a multicenter phase Ib/II study. Gastric Cancer 2024; 27:118-130. [PMID: 37906316 DOI: 10.1007/s10120-023-01435-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/29/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND We conducted a trial to evaluate the efficacy and safety of nivolumab and paclitaxel as second-line therapy for immune-related biomarker-enriched advanced gastric cancer (AGC). METHODS This open-label, single-arm, phase Ib/II study was a part of multi-institutional, biomarker-integrated umbrella study conducted in Korea. In phase Ib, patients received nivolumab (3 mg/kg) on Days 1 and 15 and paclitaxel (dose level 1, 70 mg/m2 or dose level 2, 80 mg/m2) on Days 1, 8, 15 every four weeks. In phase II, patients with Epstein-Barr virus-related, deficient mismatch repair or programmed cell death-ligand-1-positive AGC were enrolled. The primary endpoints were recommended phase II dose (RP2D, phase Ib) and progression-free survival (PFS, phase II). Secondary endpoints included objective response rate (ORR), overall survival (OS), safety, and exploratory biomarker analysis. RESULTS Dose level 2 was selected as RP2D. In phase II, 48 patients were enrolled. The median PFS and OS were 3.9 and 11.2 months, respectively. The ORR was 23.3%, and the median response duration was 16.7 months. Grade 3 or higher treatment-related adverse events, mainly neutropenia, occurred in 20 patients (41.7%). Targeted sequencing revealed that patients with RTK/RAS pathway alterations or the HLA-A02 supertype had better survival. Patients with elevated baseline interleukin-1 receptor antagonist levels had worse survival. CONCLUSIONS Although the study did not meet its primary end point, nivolumab and paclitaxel for AGC demonstrated a durable response with manageable toxicity profiles. Genomic analysis or plasma cytokine analysis may provide information for the selection of patients who would benefit more from immunotherapy combined with chemotherapy.
Collapse
Affiliation(s)
- Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
| | - Se Jung Park
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Jingmin Che
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Woo Sun Kwon
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyo Song Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Seulkee Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sook Ryun Park
- Division of Oncology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dong-Hoe Koo
- Division of Hematology/Oncology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyun Woo Lee
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, South Korea
| | - Woo Kyun Bae
- Division of Hematology and Oncology, Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Jeonnam, South Korea
| | - Hei-Cheul Jeung
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - In Gyu Hwang
- Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Chung Mo Nam
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Sun Young Rha
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, South Korea.
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
3
|
Ge Q, Li J, Yang F, Tian X, Zhang M, Hao Z, Liang C, Meng J. Molecular classifications of prostate cancer: basis for individualized risk stratification and precision therapy. Ann Med 2023; 55:2279235. [PMID: 37939258 PMCID: PMC10653710 DOI: 10.1080/07853890.2023.2279235] [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: 05/11/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
Tumour classifications play a pivotal role in prostate cancer (PCa) management. It can predict the clinical outcomes of PCa as early as the disease is diagnosed and then guide therapeutic schemes, such as active monitoring, standalone surgical intervention, or surgery supplemented with postoperative adjunctive therapy, thereby circumventing disease exacerbation and excessive treatment. Classifications based on clinicopathological features, such as prostate cancer-specific antigen, Gleason score, and TNM stage, are still the main risk stratification strategies and have played an essential role in standardized clinical decision-making. However, mounting evidence indicates that clinicopathological parameters in isolation fail to adequately capture the heterogeneity exhibited among distinct PCa patients, such as those sharing identical Gleason scores yet experiencing divergent prognoses. As a remedy, molecular classifications have been introduced. Currently, molecular studies have revealed the characteristic genomic alterations, epigenetic modulations, and tumour microenvironment associated with different types of PCa, which provide a chance for urologists to refine the PCa classification. In this context, numerous invaluable molecular classifications have been devised, employing disparate statistical methodologies and algorithmic approaches, encompassing self-organizing map clustering, unsupervised cluster analysis, and multifarious algorithms. Interestingly, the classifier PAM50 was used in a phase-2 multicentre open-label trial, NRG-GU-006, for further validation, which hints at the promise of molecular classification for clinical use. Consequently, this review examines the extant molecular classifications, delineates the prevailing panorama of clinically pertinent molecular signatures, and delves into eight emblematic molecular classifications, dissecting their methodological underpinnings and clinical utility.
Collapse
Affiliation(s)
- Qintao Ge
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, P.R. China
| | - Jiawei Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, P.R. China
| | - Feixiang Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, P.R. China
| | | | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, P.R. China
| | - Zongyao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, P.R. China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, P.R. China
| | - Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, P.R. China
| |
Collapse
|
4
|
O'Meara CH, Jafri Z, Khachigian LM. Immune Checkpoint Inhibitors, Small-Molecule Immunotherapies and the Emerging Role of Neutrophil Extracellular Traps in Therapeutic Strategies for Head and Neck Cancer. Int J Mol Sci 2023; 24:11695. [PMID: 37511453 PMCID: PMC10380483 DOI: 10.3390/ijms241411695] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has revolutionized the treatment of many cancer types, including head and neck cancers (HNC). When checkpoint and partner proteins bind, these send an "off" signal to T cells, which prevents the immune system from destroying tumor cells. However, in HNC, and indeed many other cancers, more people do not respond and/or suffer from toxic effects than those who do respond. Hence, newer, more effective approaches are needed. The challenge to durable therapy lies in a deeper understanding of the complex interactions between immune cells, tumor cells and the tumor microenvironment. This will help develop therapies that promote lasting tumorlysis by overcoming T-cell exhaustion. Here we explore the strengths and limitations of current ICI therapy in head and neck squamous cell carcinoma (HNSCC). We also review emerging small-molecule immunotherapies and the growing promise of neutrophil extracellular traps in controlling tumor progression and metastasis.
Collapse
Affiliation(s)
- Connor H O'Meara
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, Randwick, NSW 2031, Australia
| | - Zuhayr Jafri
- Vascular Biology and Translational Research, School of Biomedical Sciences, UNSW Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research, School of Biomedical Sciences, UNSW Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
5
|
Hattinger CM, Salaroglio IC, Fantoni L, Godel M, Casotti C, Kopecka J, Scotlandi K, Ibrahim T, Riganti C, Serra M. Strategies to Overcome Resistance to Immune-Based Therapies in Osteosarcoma. Int J Mol Sci 2023; 24:ijms24010799. [PMID: 36614241 PMCID: PMC9821333 DOI: 10.3390/ijms24010799] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/14/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Improving the prognosis and cure rate of HGOSs (high-grade osteosarcomas) is an absolute need. Immune-based treatment approaches have been increasingly taken into consideration, in particular for metastatic, relapsed and refractory HGOS patients, to ameliorate the clinical results currently achieved. This review is intended to give an overview on the immunotherapeutic treatments targeting, counteracting or exploiting the different immune cell compartments that are present in the HGOS tumor microenvironment. The principle at the basis of these strategies and the possible mechanisms that HGOS cells may use to escape these treatments are presented and discussed. Finally, a list of the currently ongoing immune-based trials in HGOS is provided, together with the results that have been obtained in recently completed clinical studies. The different strategies that are presently under investigation, which are generally aimed at abrogating the immune evasion of HGOS cells, will hopefully help to indicate new treatment protocols, leading to an improvement in the prognosis of patients with this tumor.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | | | - Leonardo Fantoni
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Martina Godel
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Chiara Casotti
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
- Correspondence: (C.R.); (M.S.)
| | - Massimo Serra
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Correspondence: (C.R.); (M.S.)
| |
Collapse
|
6
|
Safaeifard F, Goliaei B, Aref AR, Foroughmand-Araabi MH, Goliaei S, Lorch J, Jenkins RW, Barbie DA, Shariatpanahi SP, Rüegg C. Distinct Dynamics of Migratory Response to PD-1 and CTLA-4 Blockade Reveals New Mechanistic Insights for Potential T-Cell Reinvigoration following Immune Checkpoint Blockade. Cells 2022; 11:3534. [PMID: 36428963 PMCID: PMC9688893 DOI: 10.3390/cells11223534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/22/2022] [Accepted: 10/28/2022] [Indexed: 11/10/2022] Open
Abstract
Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1), two clinically relevant targets for the immunotherapy of cancer, are negative regulators of T-cell activation and migration. Optimizing the therapeutic response to CTLA-4 and PD-1 blockade calls for a more comprehensive insight into the coordinated function of these immune regulators. Mathematical modeling can be used to elucidate nonlinear tumor-immune interactions and highlight the underlying mechanisms to tackle the problem. Here, we investigated and statistically characterized the dynamics of T-cell migration as a measure of the functional response to these pathways. We used a previously developed three-dimensional organotypic culture of patient-derived tumor spheroids treated with anti-CTLA-4 and anti-PD-1 antibodies for this purpose. Experiment-based dynamical modeling revealed the delayed kinetics of PD-1 activation, which originates from the distinct characteristics of PD-1 and CTLA-4 regulation, and followed through with the modification of their contributions to immune modulation. The simulation results show good agreement with the tumor cell reduction and active immune cell count in each experiment. Our findings demonstrate that while PD-1 activation provokes a more exhaustive intracellular cascade within a mature tumor environment, the time-delayed kinetics of PD-1 activation outweighs its preeminence at the individual cell level and consequently confers a functional dominance to the CTLA-4 checkpoint. The proposed model explains the distinct immunostimulatory pattern of PD-1 and CTLA-4 blockade based on mechanisms involved in the regulation of their expression and may be useful for planning effective treatment schemes targeting PD-1 and CTLA-4 functions.
Collapse
Affiliation(s)
- Fateme Safaeifard
- Laboratory of Biophysics and Molecular Biology, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614411, Iran
| | - Bahram Goliaei
- Laboratory of Biophysics and Molecular Biology, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614411, Iran
| | - Amir R. Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Xsphera Biosciences Inc., Boston, MA 02210, USA
| | | | - Sama Goliaei
- Faculty of New Sciences & Technologies, University of Tehran, Tehran 1439957131, Iran
| | - Jochen Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Russell W. Jenkins
- MassGeneral Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David A. Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Seyed Peyman Shariatpanahi
- Laboratory of Biophysics and Molecular Biology, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614411, Iran
| | - Curzio Rüegg
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology, and Immunology, Faculty of Sciences and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
7
|
Chen S, Ma S, Wang H, Shao X, Ding B, Guo Z, Chen X, Wang Y. Unraveling the mechanism of alkaloids from Sophora alopecuroides Linn combined with immune checkpoint blockade in the treatment of non-small cell lung cancer based on systems pharmacology. Bioorg Med Chem 2022; 64:116724. [PMID: 35468537 DOI: 10.1016/j.bmc.2022.116724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/07/2022] [Accepted: 03/22/2022] [Indexed: 11/26/2022]
Abstract
Quinolizidine alkaloids, as essential active ingredients extracted from Sophora alopecuroides Linn (SAL), have been proven to be pharmacologically active in a variety of cancers including non-small cell lung cancer (NSCLC). However, whether these alkaloids have substantial benefits in combination with immune checkpoint blockade (ICB) for the treatment of NSCLC is unknown. Here, we explore the potential of these alkaloids in combination with ICB therapy based on a systems pharmacology and bioinformatics approach. We found that 37 alkaloids in SAL have highly similar characteristics in the molecular skeleton, pharmacological properties, and targets. The expression of targets of these alkaloids are significantly correlated with the infiltration level of tumor infiltrating lymphocytes and the expression levels of multiple immune checkpoints in NSCLC. They share similar molecular mechanisms in antitumor immunity. Sophocarpine (Sop) is one of the most representative constituents of these alkaloids. We demonstrated that the Sop promotes PD-L1 expression to improve the effects of PD-L1 blockade treatment via the ADORA1-ATF3 axis. In conclusion, our study identified these alkaloids as promising candidates for the treatment of NSCLC, either alone or in combination with ICB, with potential value for drug development and may provide a promising strategy for improving the survival of NSCLC patients.
Collapse
Affiliation(s)
- Sen Chen
- Center of Bioinformatics, College of Life Science, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Shuangxin Ma
- Center of Bioinformatics, College of Life Science, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Haiqing Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Xuexue Shao
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi, Xinjiang 832002, China
| | - Bojiao Ding
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Zihu Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Xuetong Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| | - Yonghua Wang
- Center of Bioinformatics, College of Life Science, Northwest A & F University, Yangling, Shaanxi 712100, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| |
Collapse
|
8
|
Zhang Z, Bu L, Luo J, Guo J. Targeting protein kinases benefits cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188738. [PMID: 35660645 DOI: 10.1016/j.bbcan.2022.188738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/16/2022] [Accepted: 05/28/2022] [Indexed: 02/07/2023]
Abstract
Small-molecule kinase inhibitors have been well established and successfully developed in the last decades for cancer target therapies. However, intrinsic or acquired drug resistance is becoming the major barrier for their clinical application. With the development of immunotherapies, in particular the discovery of immune checkpoint inhibitors (ICIs), the combination of ICIs with other therapies have recently been extensively explored, among which combination of ICIs with kinase inhibitors achieves promising clinical outcome in a plethora of cancer types. Here we comprehensively summarize the potent roles of protein kinases in modulating immune checkpoints both in tumor and immune cells, and reshaping tumor immune microenvironments by evoking innate immune response and neoantigen generation or presentation. Moreover, the clinical trial and approval of combined administration of kinase inhibitors with ICIs are collected, highlighting the precise strategies to benefit cancer immune therapies.
Collapse
Affiliation(s)
- Zhengkun Zhang
- Department of Urology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Lang Bu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Junhang Luo
- Department of Urology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Jianping Guo
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
9
|
CSNK2 in cancer: pathophysiology and translational applications. Br J Cancer 2022; 126:994-1003. [PMID: 34773100 PMCID: PMC8980014 DOI: 10.1038/s41416-021-01616-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/29/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022] Open
Abstract
Protein kinase CSNK2 (CK2) is a pleiotropic serine/threonine kinase frequently dysregulated in solid and hematologic malignancies. To consolidate a wide range of biological and clinically oriented data from this unique kinase in cancer, this systematic review summarises existing knowledge from in vitro, in vivo and pre-clinical studies on CSNK2 across 24 different human cancer types. CSNK2 mRNA transcripts, protein levels and activity were found to be routinely upregulated in cancer, and commonly identified phosphotargets included AKT, STAT3, RELA, PTEN and TP53. Phenotypically, it frequently influenced evasion of apoptosis, enhancement of proliferation, cell invasion/metastasis and cell cycle control. Clinically, it held prognostic significance across 14 different cancers, and its inhibition in xenograft experiments resulted in a positive treatment response in 12. In conjunction with commentary on preliminary studies of CSNK2 inhibitors in humans, this review harmonises an extensive body of CSNK2 data in cancer and reinforces its emergence as an attractive target for cancer therapy. Continuing to investigate CSNK2 will be crucial to advancing our understanding of CSNK2 biology, and offers the promise of important new discoveries scientifically and clinically.
Collapse
|
10
|
Bruns IB, Beltman JB. Quantifying the contribution of transcription factor activity, mutations and microRNAs to CD274 expression in cancer patients. Sci Rep 2022; 12:4374. [PMID: 35289334 PMCID: PMC8921511 DOI: 10.1038/s41598-022-08356-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/03/2022] [Indexed: 12/15/2022] Open
Abstract
Immune checkpoint inhibitors targeting the programmed cell death protein 1 (PD-1)/programmed cell death protein ligand 1 (PD-L1) axis have been remarkably successful in inducing tumor remissions in several human cancers, yet a substantial number of patients do not respond to treatment. Because this may be partially due to the mechanisms giving rise to high PD-L1 expression within a patient, it is highly relevant to fully understand these mechanisms. In this study, we conduct a bioinformatic analysis to quantify the relative importance of transcription factor (TF) activity, microRNAs (miRNAs) and mutations in determining PD-L1 (CD274) expression at mRNA level based on data from the Cancer Genome Atlas. To predict individual CD274 levels based on TF activity, we developed multiple linear regression models by taking the expression of target genes of the TFs known to directly target PD-L1 as independent variables. This analysis showed that IRF1, STAT1, NFKB and BRD4 are the most important regulators of CD274 expression, explaining its mRNA levels in 90–98% of the patients. Because the remaining patients had high CD274 levels independent of these TFs, we next investigated whether mutations associated with increased CD274 mRNA levels, and low levels of miRNAs associated with negative regulation of CD274 expression could cause high CD274 levels in these patients. We found that mutations or miRNAs offered an explanation for high CD274 levels in 81–100% of the underpredicted patients. Thus, CD274 expression is largely explained by TF activity, and the remaining unexplained cases can largely be explained by mutations or low miRNA abundance.
Collapse
Affiliation(s)
- Imke B Bruns
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Joost B Beltman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
11
|
Akbari H, Taghizadeh-Hesary F, Bahadori M. Mitochondria determine response to anti-programmed cell death protein-1 (anti-PD-1) immunotherapy: An evidence-based hypothesis. Mitochondrion 2021; 62:151-158. [PMID: 34890822 DOI: 10.1016/j.mito.2021.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 11/21/2021] [Accepted: 12/01/2021] [Indexed: 02/06/2023]
Abstract
Immunotherapy based on programmed cell death protein-1 (PD-1) is a promising approach in oncology. However, a significant fraction of patients remain unresponsive. Therefore, it is imperative to clarify the relevant predictive factors. A decrease in cellular adenosine triphosphate (c-ATP) level can predispose to cellular dysfunction. ATP is a prerequisite for proper T cell migration and activation. Therefore, a decrease in the c-ATP level impairs T cell function and promotes cancer progression. This article gives an overview of the potential predictive factors of PD-1 blockade. Besides, it highlights the pivotal role of mitochondria in response to anti-PD-1 therapies.
Collapse
Affiliation(s)
- Hassan Akbari
- Department of Pathology, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Traditional Medicine School, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Moslem Bahadori
- Professor Emeritus, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Thangam R, Patel KD, Kang H, Paulmurugan R. Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy-Current Status and Future Perspectives. Vaccines (Basel) 2021; 9:vaccines9080935. [PMID: 34452059 PMCID: PMC8402739 DOI: 10.3390/vaccines9080935] [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: 06/14/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/17/2022] Open
Abstract
Engineering polymeric nanoparticles for their shape, size, surface chemistry, and functionalization using various targeting molecules has shown improved biomedical applications for nanoparticles. Polymeric nanoparticles have created tremendous therapeutic platforms, particularly applications related to chemo- and immunotherapies in cancer. Recently advancements in immunotherapies have broadened this field in immunology and biomedical engineering, where "immunoengineering" creates solutions to target translational science. In this regard, the nanoengineering field has offered the various techniques necessary to manufacture and assemble multifunctional polymeric nanomaterial systems. These include nanoparticles functionalized using antibodies, small molecule ligands, targeted peptides, proteins, and other novel agents that trigger and encourage biological systems to accept the engineered materials as immune enhancers or as vaccines to elevate therapeutic functions. Strategies to engineer polymeric nanoparticles with therapeutic and targeting molecules can provide solutions for developing immune vaccines via maintaining the receptor storage in T- and B cells. Furthermore, cancer immunotherapy using polymeric nanomaterials can serve as a gold standard approach for treating primary and metastasized tumors. The current status of the limited availability of immuno-therapeutic drugs highlights the importance of polymeric nanomaterial platforms to improve the outcomes via delivering anticancer agents at localized sites, thereby enhancing the host immune response in cancer therapy. This review mainly focuses on the potential scientific enhancements and recent developments in cancer immunotherapies by explicitly discussing the role of polymeric nanocarriers as nano-vaccines. We also briefly discuss the role of multifunctional nanomaterials for their therapeutic impacts on translational clinical applications.
Collapse
Affiliation(s)
- Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Correspondence: (R.T.); (R.P.)
| | - Kapil D. Patel
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Department of Biomicrosystem Technology, Korea University, Seoul 02841, Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Correspondence: (R.T.); (R.P.)
| |
Collapse
|
13
|
Abstract
Breast cancer, as a heterogeneous disease, includes a wide range of pathological and clinical behaviors. Current treatment protocols, including radiotherapy, chemotherapy, and hormone replacement therapy, are mainly associated with poor response and high rate of recurrence. Therefore, more efforts are needed to develop alternative therapies for this type of cancer. Immunotherapy, as a novel strategy in cancer treatment, has a potential in treating breast cancer patients. Although breast cancer has long been considered problematic to treat with immunotherapy, as it is immunologically "cold," numerous newer preclinical and clinical reports now recommend that immunotherapy has the capability to treat breast cancer patients. In this review, we highlight the different immunotherapy strategies in breast cancer treatment.
Collapse
|
14
|
Zhang T, Zheng S, Liu Y, Li X, Wu J, Sun Y, Liu G. DNA damage response and PD-1/PD-L1 pathway in ovarian cancer. DNA Repair (Amst) 2021; 102:103112. [PMID: 33838550 DOI: 10.1016/j.dnarep.2021.103112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 03/17/2021] [Accepted: 03/27/2021] [Indexed: 12/15/2022]
Abstract
Ovarian cancer has a poor prognosis due to drug resistance, relapse and metastasis. In recent years, immunotherapy has been applied in numerous cancers clinically. However, the effect of immunotherapy monotherapy in ovarian cancer is limited. DNA damage response (DDR) is an essential factor affecting the efficacy of tumor immunotherapy. Defective DNA repair may lead to carcinogenesis and tumor genomic instability, but on the other hand, it may also portend particular vulnerability of tumors and can be used as biomarkers for immunotherapy patient selection. Programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1) pathway mediates tumor immune escape, which may be a promising target for immunotherapy. Therefore, further understanding of the mechanism of PD-L1 expression after DDR may help guide the development of immunotherapy in ovarian cancer. In this review, we present the DNA damage repair pathway and summarize how DNA damage repair affects the PD-1/PD-L1 pathway in cancer cells. And then we look for biomarkers that affect efficacy or prognosis. Finally, we review the progress of PD-1/PD-L1-based immunotherapy in combination with other therapies that may affect the DDR pathway in ovarian cancer.
Collapse
Affiliation(s)
- Tianyu Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China.
| | - Shuangshuang Zheng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China.
| | - Yang Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China.
| | - Xiao Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China.
| | - Jing Wu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China.
| | - Yue Sun
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China.
| | - Guoyan Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, 300052, China.
| |
Collapse
|
15
|
Simonian M, Haji Ghaffari M, Negahdari B. Immunotherapy for Breast Cancer Treatment. IRANIAN BIOMEDICAL JOURNAL 2021; 25:140-56. [PMID: 33724757 PMCID: PMC8183391 DOI: 10.29252/ibj.25.3.140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/12/2020] [Indexed: 06/12/2023]
Abstract
Breast cancer, as a heterogeneous disease, includes a wide range of pathological and clinical behaviors. Current treatment protocols, including radiotherapy, chemotherapy, and hormone replacement therapy, are mainly associated with poor response and high rate of recurrence. Therefore, more efforts are needed to develop alternative therapies for this type of cancer. Immunotherapy, as a novel strategy in cancer treatment, has a potential in treating breast cancer patients. Although breast cancer has long been considered problematic to treat with immunotherapy, as it is immunologically "cold," numerous newer preclinical and clinical reports now recommend that immunotherapy has the capability to treat breast cancer patients. In this review, we highlight the different immunotherapy strategies in breast cancer treatment.
Collapse
Affiliation(s)
| | | | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Mao C, Zeng X, Zhang C, Yang Y, Xiao X, Luan S, Zhang Y, Yuan Y. Mechanisms of Pharmaceutical Therapy and Drug Resistance in Esophageal Cancer. Front Cell Dev Biol 2021; 9:612451. [PMID: 33644048 PMCID: PMC7905099 DOI: 10.3389/fcell.2021.612451] [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: 09/30/2020] [Accepted: 01/04/2021] [Indexed: 02/05/2023] Open
Abstract
Pharmaceutical therapies are essential for esophageal cancer (EC). For the advanced EC, the neoadjuvant therapy regimen, including chemotherapy plus radiotherapy and/or immunotherapy, is effective to achieve clinical benefit, even pathological complete response. For the unresectable, recurrent, and metastatic EC, the pharmaceutical therapy is the limited effective regimen to alleviate the disease and prolong the progression-free survival and overall survival. In this review, we focus on the pharmaceutical applications in EC treatment including cytotoxic agents, molecular targeted antibodies, and immune checkpoint inhibitors (ICIs). The chemotherapy regimen is based on cytotoxic agents such as platinum-based complexes, fluorinated pyrimidines and taxenes. Although the cytotoxic agents have been developed in past decades, the standard chemotherapy regimen is still the cisplatin and 5-FU or paclitaxel because the derived drugs have no significant advantages of overcoming the shortcomings of side effects and drug resistance. The targeted molecular therapy is an essential supplement for chemotherapy; however, there are only a few targeted therapies available in clinical practice. Trastuzumab and ramucirumab are the only two molecular therapy drugs which are approved by the US Food and Drug Administration to treat advanced and/or metastatic EC. Although the targeted therapy usually achieves effective benefits in the early stage therapy of EC, the patients will always develop drug resistance during treatment. ICIs have had a significant impact on routine clinical practice in cancer treatment. The anti-programmed cell death-1 monoclonal antibodies pembrolizumab and nivolumab, as the ICIs, are recommended for advanced EC by several clinical trials. However, the significant issues of pharmaceutical treatment are still the dose-limiting side effects and primary or secondary drug resistance. These defects of pharmaceutical therapy restrain the clinical application and diminish the effectiveness of treatment.
Collapse
Affiliation(s)
- Chengyi Mao
- Department of Thoracic Surgery West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yushang Yang
- Department of Thoracic Surgery West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiao
- Department of Thoracic Surgery West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Luan
- Department of Thoracic Surgery West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Nursing Key Laboratory of Sichuan Province, Chengdu, China
- Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Yuan
- Department of Thoracic Surgery West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Yu CC, Wortman JC, He TF, Solomon S, Zhang RZ, Rosario A, Wang R, Tu TY, Schmolze D, Yuan Y, Yost SE, Li X, Levine H, Atwal G, Lee PP. Physics approaches to the spatial distribution of immune cells in tumors. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2021; 84:022601. [PMID: 33232952 DOI: 10.1088/1361-6633/abcd7b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The goal of immunotherapy is to mobilize the immune system to kill cancer cells. Immunotherapy is more effective and, in general, the prognosis is better, when more immune cells infiltrate the tumor. We explore the question of whether the spatial distribution rather than just the density of immune cells in the tumor is important in forecasting whether cancer recurs. After reviewing previous work on this issue, we introduce a novel application of maximum entropy to quantify the spatial distribution of discrete point-like objects. We apply our approach to B and T cells in images of tumor tissue taken from triple negative breast cancer patients. We find that the immune cells are more spatially dispersed in good clinical outcome (no recurrence of cancer within at least 5 years of diagnosis) compared to poor clinical outcome (recurrence within 3 years of diagnosis). Our results highlight the importance of spatial distribution of immune cells within tumors with regard to clinical outcome, and raise new questions on their role in cancer recurrence.
Collapse
Affiliation(s)
- Clare C Yu
- Department of Physics and Astronomy, University of California, Irvine, Irvine, CA 92697, United States of America
- Department of Immuno-Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Juliana C Wortman
- Department of Physics and Astronomy, University of California, Irvine, Irvine, CA 92697, United States of America
| | - Ting-Fang He
- Department of Immuno-Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Shawn Solomon
- Department of Immuno-Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Robert Z Zhang
- Department of Immuno-Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Anthony Rosario
- Department of Immuno-Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Roger Wang
- Department of Immuno-Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Travis Y Tu
- Department of Immuno-Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Daniel Schmolze
- Department of Pathology, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Yuan Yuan
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Susan E Yost
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| | - Xuefei Li
- Department of Bioengineering and the Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, United States of America
| | - Herbert Levine
- Department of Bioengineering and the Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, United States of America
- Department of Bioengineering and Department of Physics, Northeastern University, Boston, MA 02115, United States of America
| | - Gurinder Atwal
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, United States of America
| | - Peter P Lee
- Department of Immuno-Oncology, City of Hope Comprehensive Cancer Center and Beckman Research Institute, 1500 East Duarte Road, Duarte, CA 91010, United States of America
| |
Collapse
|
18
|
Arora M, Kumari S, Singh J, Chopra A, Chauhan SS. Expression pattern, regulation, and clinical significance of TOX in breast cancer. Cancer Immunol Immunother 2021; 70:349-363. [PMID: 32757053 PMCID: PMC10992774 DOI: 10.1007/s00262-020-02689-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022]
Abstract
Thymocyte selection-associated high mobility group box protein (TOX) is a transcription factor implicated in the regulation of T cell exhaustion during chronic infection and cancer. While TOX is being targeted for cancer immunotherapy, limited information is available about its significance in breast cancer and other solid tumors. We performed a comprehensive analysis of TOX gene expression, its epigenetic regulation, protein localization, relation to tumor infiltrating immune cell composition, and prognostic significance in breast cancer using publicly available datasets. Our results suggest an inverse correlation between TOX expression and DNA methylation in tumor cells. However, its expression is elevated in tumor infiltrating immune cells (TIICs), which may compensates for the total TOX levels in the tumor as a whole. Furthermore, higher TOX levels in tumors are associated with T cell exhaustion signatures along with presence of active inflammatory response, including elevated levels of T cell effector cytokines. Survival analysis also confirmed that higher expression of TOX is associated with better prognosis in breast cancer. Therefore, expression of TOX may serve as a novel prognostic marker for this malignancy.
Collapse
Affiliation(s)
- Mohit Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sarita Kumari
- Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Jay Singh
- Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
19
|
Msaouel P, Genovese G, Gao J, Sen S, Tannir NM. TAM kinase inhibition and immune checkpoint blockade- a winning combination in cancer treatment? Expert Opin Ther Targets 2021; 25:141-151. [PMID: 33356674 DOI: 10.1080/14728222.2021.1869212] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Immune checkpoint inhibitors (ICI) have shown great promise in a wide spectrum of malignancies. However, responses are not always durable, and this mode of treatment is only effective in a subset of patients. As such, there exists an unmet need for novel approaches to bolster ICI efficacy.Areas covered: We review the role of the Tyro3, Axl, and Mer (TAM) receptor tyrosine kinases in promoting tumor-induced immune suppression and discuss the benefits that may be derived from combining ICI with TAM kinase-targeted tyrosine kinase inhibitors. We searched the MEDLINE Public Library of Medicine (PubMed) and EMBASE databases and referred to ClinicalTrials.gov for relevant ongoing studies.Expert opinion: Targeting of TAM kinases may improve the efficacy of immune checkpoint blockade. However, it remains to be determined whether this effect will be better achieved by the selective targeting of each TAM receptor, depending on the context, or by multi-receptor TAM inhibitors. Triple inhibition of all TAM receptors is more likely to be associated with an increased risk for adverse events. Clinical trial designs should use high-resolution clinical endpoints and proper control arms to determine the synergistic effects of combining TAM inhibition with immune checkpoint blockade.
Collapse
Affiliation(s)
- Pavlos Msaouel
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
20
|
Li L, Wei JR, Dong J, Lin QG, Tang H, Jia YX, Tan W, Chen QY, Zeng TT, Xing S, Qin YR, Zhu YH, Li Y, Guan XY. Laminin γ2-mediating T cell exclusion attenuates response to anti-PD-1 therapy. SCIENCE ADVANCES 2021; 7:7/6/eabc8346. [PMID: 33536206 PMCID: PMC7857690 DOI: 10.1126/sciadv.abc8346] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 12/17/2020] [Indexed: 05/10/2023]
Abstract
PD-1/PD-L1 blockade therapies provide notable clinical benefits for patients with advanced cancers, but the factors influencing the effectiveness of the treatment remain incompletely cataloged. Here, the up-regulation of laminin γ2 (Ln-γ2) predicted the attenuated efficacy of anti-PD-1 drugs and was associated with unfavorable outcomes in patients with lung cancer or esophageal cancer. Furthermore, Ln-γ2 was transcriptionally activated by transforming growth factor-β1 (TGF-β1) secreted from cancer-associated fibroblasts via JNK/AP1 signaling, which blocked T cell infiltration into the tumor nests by altering the expression of T cell receptors. Coadministration of the TGF-β receptor inhibitor galunisertib and chemotherapy drugs provoked vigorous antitumor activity of anti-PD-1 therapy in mouse tumor models. Therefore, Ln-γ2 may represent a useful biomarker to optimize clinical decisions and predict the response of cancer patients to treatment with anti-PD-1 drugs.
Collapse
Affiliation(s)
- Lei Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong 00852, China
- Department of Clinical Oncology Center, The University of Hongkong-Shenzhen Hospital, Shenzhen 518058, China
| | - Jia-Ru Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Jun Dong
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Qing-Guang Lin
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hong Tang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China
| | - Yong-Xu Jia
- Department of Clinical Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Wanlin Tan
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Qing-Yun Chen
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ting-Ting Zeng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shan Xing
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yan-Ru Qin
- Department of Clinical Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Ying-Hui Zhu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yan Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong 00852, China
- Department of Clinical Oncology Center, The University of Hongkong-Shenzhen Hospital, Shenzhen 518058, China
- Department of Clinical Oncology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
21
|
Bansal D, Reimers MA, Knoche EM, Pachynski RK. Immunotherapy and Immunotherapy Combinations in Metastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2021; 13:cancers13020334. [PMID: 33477569 PMCID: PMC7831137 DOI: 10.3390/cancers13020334] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/01/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
Although most prostate cancers are localized, and the majority are curable, recurrences occur in approximately 35% of men. Among patients with prostate-specific antigen (PSA) recurrence and PSA doubling time (PSADT) less than 15 months after radical prostatectomy, prostate cancer accounted for approximately 90% of the deaths by 15 years after recurrence. An immunosuppressive tumor microenvironment (TME) and impaired cellular immunity are likely largely responsible for the limited utility of checkpoint inhibitors (CPIs) in advanced prostate cancer compared with other tumor types. Thus, for immunologically "cold" malignancies such as prostate cancer, clinical trial development has pivoted towards novel approaches to enhance immune responses. Numerous clinical trials are currently evaluating combination immunomodulatory strategies incorporating vaccine-based therapies, checkpoint inhibitors, and chimeric antigen receptor (CAR) T cells. Other trials evaluate the efficacy and safety of these immunomodulatory agents' combinations with standard approaches such as androgen deprivation therapy (ADT), taxane-based chemotherapy, radiotherapy, and targeted therapies such as tyrosine kinase inhibitors (TKI) and poly ADP ribose polymerase (PARP) inhibitors. Here, we will review promising immunotherapies in development and ongoing trials for metastatic castration-resistant prostate cancer (mCRPC). These novel trials will build on past experiences and promise to usher a new era to treat patients with mCRPC.
Collapse
|
22
|
Lambrou GI, Adamaki M, Hatziagapiou K, Vlahopoulos S. Gene Expression and Resistance to Glucocorticoid-Induced Apoptosis in Acute Lymphoblastic Leukemia: A Brief Review and Update. Curr Drug Res Rev 2021; 12:131-149. [PMID: 32077838 DOI: 10.2174/2589977512666200220122650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/29/2019] [Accepted: 01/23/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Resistance to glucocorticoid (GC)-induced apoptosis in Acute Lymphoblastic Leukemia (ALL), is considered one of the major prognostic factors for the disease. Prednisolone is a corticosteroid and one of the most important agents in the treatment of acute lymphoblastic leukemia. The mechanics of GC resistance are largely unknown and intense ongoing research focuses on this topic. AIM The aim of the present study is to review some aspects of GC resistance in ALL, and in particular of Prednisolone, with emphasis on previous and present knowledge on gene expression and signaling pathways playing a role in the phenomenon. METHODS An electronic literature search was conducted by the authors from 1994 to June 2019. Original articles and systematic reviews selected, and the titles and abstracts of papers screened to determine whether they met the eligibility criteria, and full texts of the selected articles were retrieved. RESULTS Identification of gene targets responsible for glucocorticoid resistance may allow discovery of drugs, which in combination with glucocorticoids may increase the effectiveness of anti-leukemia therapies. The inherent plasticity of clinically evolving cancer justifies approaches to characterize and prevent undesirable activation of early oncogenic pathways. CONCLUSION Study of the pattern of intracellular signal pathway activation by anticancer drugs can lead to development of efficient treatment strategies by reducing detrimental secondary effects.
Collapse
Affiliation(s)
- George I Lambrou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Athens, Greece
| | - Maria Adamaki
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Athens, Greece
| | - Kyriaki Hatziagapiou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Athens, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Athens, Greece
| |
Collapse
|
23
|
Id1 and PD-1 Combined Blockade Impairs Tumor Growth and Survival of KRAS-mutant Lung Cancer by Stimulating PD-L1 Expression and Tumor Infiltrating CD8 + T Cells. Cancers (Basel) 2020; 12:cancers12113169. [PMID: 33126649 PMCID: PMC7693788 DOI: 10.3390/cancers12113169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/19/2020] [Accepted: 10/25/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Lung adenocarcinoma is the most frequent lung cancer subtype. Many of those adenocarcinomas of the lung are driven by the KRAS gene. Although immunotherapy has significantly improved the clinical outcomes of patients with lung adenocarcinomas, many patients do not benefit from that therapeutic strategy. Id1 is a protein involved in immunosuppression. Here we aimed to test whether a combined blockade of Id1 and PD-1 is able to improve outcomes of mice models with KRAS-driven lung adenocarcinoma. Abstract The use of PD-1/PD-L1 checkpoint inhibitors in advanced NSCLC is associated with longer survival. However, many patients do not benefit from PD-1/PD-L1 blockade, largely because of immunosuppression. New immunotherapy-based combinations are under investigation in an attempt to improve outcomes. Id1 (inhibitor of differentiation 1) is involved in immunosuppression. In this study, we explored the potential synergistic effect of the combination of Id1 inhibition and pharmacological PD-L1 blockade in three different syngeneic murine KRAS-mutant lung adenocarcinoma models. TCGA analysis demonstrated a negative and statistically significant correlation between PD-L1 and Id1 expression levels. This observation was confirmed in vitro in human and murine KRAS-driven lung cancer cell lines. In vivo experiments in KRAS-mutant syngeneic and metastatic murine lung adenocarcinoma models showed that the combined blockade targeting Id1 and PD-1 was more effective than each treatment alone in terms of tumor growth impairment and overall survival improvement. Mechanistically, multiplex quantification of CD3+/CD4+/CD8+ T cells and flow cytometry analysis showed that combined therapy favors tumor infiltration by CD8+ T cells, whilst in vivo CD8+ T cell depletion led to tumor growth restoration. Co-culture assays using CD8+ cells and tumor cells showed that T cells present a higher antitumor effect when tumor cells lack Id1 expression. These findings highlight that Id1 blockade may contribute to a significant immune enhancement of antitumor efficacy of PD-1 inhibitors by increasing PD-L1 expression and harnessing tumor infiltration of CD8+ T lymphocytes.
Collapse
|
24
|
Camorani S, Passariello M, Agnello L, Esposito S, Collina F, Cantile M, Di Bonito M, Ulasov IV, Fedele M, Zannetti A, De Lorenzo C, Cerchia L. Aptamer targeted therapy potentiates immune checkpoint blockade in triple-negative breast cancer. J Exp Clin Cancer Res 2020; 39:180. [PMID: 32892748 PMCID: PMC7487859 DOI: 10.1186/s13046-020-01694-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a uniquely aggressive cancer with high rates of relapse due to resistance to chemotherapy. TNBC expresses higher levels of programmed cell death-ligand 1 (PD-L1) compared to other breast cancers, providing the rationale for the recently approved immunotherapy with anti-PD-L1 monoclonal antibodies (mAbs). A huge effort is dedicated to identify actionable biomarkers allowing for combination therapies with immune-checkpoint blockade. Platelet-derived growth factor receptor β (PDGFRβ) is highly expressed in invasive TNBC, both on tumor cells and tumor microenvironment. We recently proved that tumor growth and lung metastases are impaired in mouse models of human TNBC by a high efficacious PDGFRβ aptamer. Hence, we aimed at investigating the effectiveness of a novel combination treatment with the PDGFRβ aptamer and anti-PD-L1 mAbs in TNBC. METHODS The targeting ability of the anti-human PDGFRβ aptamer toward the murine receptor was verified by streptavidin-biotin assays and confocal microscopy, and its inhibitory function by transwell migration assays. The anti-proliferative effects of the PDGFRβ aptamer/anti-PD-L1 mAbs combination was assessed in human MDA-MB-231 and murine 4 T1 TNBC cells, both grown as monolayer or co-cultured with lymphocytes. Tumor cell lysis and cytokines secretion by lymphocytes were analyzed by LDH quantification and ELISA, respectively. Orthotopic 4 T1 xenografts in syngeneic mice were used for dissecting the effect of aptamer/mAb combination on tumor growth, metastasis and lymphocytes infiltration. Ex vivo analyses through immunohistochemistry, RT-qPCR and immunoblotting were performed. RESULTS We show that the PDGFRβ aptamer potentiates the anti-proliferative activity of anti-PD-L1 mAbs on both human and murine TNBC cells, according to its human/mouse cross-reactivity. Further, by binding to activated human and mouse lymphocytes, the aptamer enhances the anti-PD-L1 mAb-induced cytotoxicity of lymphocytes against tumor cells. Importantly, the aptamer heightens the antibody efficacy in inhibiting tumor growth and lung metastases in mice. It acts on both tumor cells, inhibiting Akt and ERK1/2 signaling pathways, and immune populations, increasing intratumoral CD8 + T cells and reducing FOXP3 + Treg cells. CONCLUSION Co-treatment of PDGFRβ aptamer with anti-PD-L1 mAbs is a viable strategy, thus providing for the first time an evidence of the efficacy of PDGFRβ/PD-L1 co-targeting combination therapy in TNBC.
Collapse
Affiliation(s)
- Simona Camorani
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", CNR, Via S. Pansini 5, 80131, Naples, Italy
| | - Margherita Passariello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
- Ceinge-Biotecnologie Avanzate s.c.a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Lisa Agnello
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", CNR, Via S. Pansini 5, 80131, Naples, Italy
| | - Silvia Esposito
- Ceinge-Biotecnologie Avanzate s.c.a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Francesca Collina
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Monica Cantile
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Maurizio Di Bonito
- Pathology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Ilya V Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", CNR, Via S. Pansini 5, 80131, Naples, Italy
| | - Antonella Zannetti
- Institute of Biostructure and Bioimaging, CNR, Via T. De Amicis 95, 80145, Naples, Italy
| | - Claudia De Lorenzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy.
- Ceinge-Biotecnologie Avanzate s.c.a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", CNR, Via S. Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
25
|
Haibe Y, El Husseini Z, El Sayed R, Shamseddine A. Resisting Resistance to Immune Checkpoint Therapy: A Systematic Review. Int J Mol Sci 2020; 21:E6176. [PMID: 32867025 PMCID: PMC7504220 DOI: 10.3390/ijms21176176] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/11/2020] [Accepted: 07/12/2020] [Indexed: 12/14/2022] Open
Abstract
The treatment landscape in oncology has witnessed a major revolution with the introduction of checkpoint inhibitors: anti-PD1, anti-PDL1 and anti-CTLA-4. These agents enhance the immune response towards cancer cells instead of targeting the tumor itself, contrary to standard chemotherapy. Although long-lasting durable responses have been observed with immune checkpoints inhibitors, the response rate remains relatively low in many cases. Some patients respond in the beginning but then eventually develop acquired resistance to treatment and progress. Other patients having primary resistance never respond. Multiple studies have been conducted to further elucidate these variations in response in different tumor types and different individuals. This paper provides an overview of the mechanisms of resistance to immune checkpoint inhibitors and highlights the possible therapeutic approaches under investigation aiming to overcome such resistance in order to improve the clinical outcomes of cancer patients.
Collapse
Affiliation(s)
| | | | | | - Ali Shamseddine
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (Z.E.H.); (R.E.S.)
| |
Collapse
|
26
|
Pérez-Ruiz E, Melero I, Kopecka J, Sarmento-Ribeiro AB, García-Aranda M, De Las Rivas J. Cancer immunotherapy resistance based on immune checkpoints inhibitors: Targets, biomarkers, and remedies. Drug Resist Updat 2020; 53:100718. [PMID: 32736034 DOI: 10.1016/j.drup.2020.100718] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/09/2020] [Accepted: 07/13/2020] [Indexed: 01/05/2023]
Abstract
Cancer is one of the main public health problems in the world. Systemic therapies such as chemotherapy and more recently target therapies as well as immunotherapy have improved the prognosis of this large group of complex malignant diseases. However, the frequent emergence of multidrug resistance (MDR) mechanisms is one of the major impediments towards curative treatment of cancer. While several mechanisms of drug chemoresistance are well defined, resistance to immunotherapy is still insufficiently unclear due to the complexity of the immune response and its dependence on the host. Expression and regulation of immune checkpoint molecules (such as PD-1, CD279; PD-L1, CD274; and CTLA-4, CD152) play a key role in the response to immunotherapy. In this regard, immunotherapy based on immune checkpoints inhibitors (ICIs) is a common clinical approach for treatment of patients with poor prognosis when other first-line therapies have failed. Unfortunately, about 70 % of patients are classified as non-responders, or they progress after initial response to these ICIs. Multiple factors can be related to immunotherapy resistance: characteristics of the tumor microenvironment (TME); presence of tumor infiltrating lymphocytes (TILs), such as CD8 + T cells associated with treatment-response; presence of tumor associated macrophages (TAMs); activation of certain regulators (like PIK3γ or PAX4) found present in non-responders; a low percentage of PD-L1 expressing cells; tumor mutational burden (TMB); gain or loss of antigen-presenting molecules; genetic and epigenetic alterations correlated with resistance. This review provides an update on the current state of immunotherapy resistance presenting targets, biomarkers and remedies to overcome such resistance.
Collapse
Affiliation(s)
- Elisabeth Pérez-Ruiz
- Medical Oncology Department, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Costa del Sol, 29603 Marbella, Malaga, Spain.
| | - Ignacio Melero
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra (CUN), Center for Applied Medical Research (CIMA), Universidad de Navarra (UNAV), 31008 Pamplona, Spain.
| | - Joanna Kopecka
- Department of Oncology, Turin School of Medicine, University of Turin, 10126 Turin, Italy.
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology and University Clinic of Hematology and Coimbra Institute for Clinical and Biomedical Research - Group of Environment Genetics and Oncobiology (iCBR/CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), and Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
| | - Marilina García-Aranda
- Research Unit, Hospital Costa del Sol, Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Instituto de Investigación Biomédica de Málaga (IBIMA), 29603 Marbella, Malaga, Spain.
| | - Javier De Las Rivas
- Cancer Research Center (CiC-IBMCC, CSIC/USAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), and Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain.
| |
Collapse
|
27
|
Balibegloo M, Rezaei N. Development and clinical application of bispecific antibody in the treatment of colorectal cancer. Expert Rev Clin Immunol 2020; 16:689-709. [PMID: 32536227 DOI: 10.1080/1744666x.2020.1783249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Treatment of colorectal cancer as one of the most commonly diagnosed and a frequent cause of cancer-related deaths is of great challenges in health-related issues. AREAS COVERED Immunotherapy is the fourth pillar of cancer treatment which provides more novel therapeutic options with expanding investigational potentials. One of the modalities in immunotherapy is the use of bispecific antibodies. Despite demonstrating many promising roles, it still needs more advanced studies to identify the actual pros and cons. In this review, the application of bispecific antibody in the treatment of colorectal cancer has been explained, based on preclinical and clinical studies. The literature search was conducted mainly through PubMed in June and September 2019. EXPERT OPINION Bispecific antibody is in its early stages in colorectal cancer treatment, requiring modern technologies in manufacturing, better biomarkers and more specific target antigens, more studies on individual genetic variations, and conducting later phase clinical trials and systematic reviews to achieve better survival benefits.
Collapse
Affiliation(s)
- Maryam Balibegloo
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN) , Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences , Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN) , Tehran, Iran
| |
Collapse
|
28
|
Affiliation(s)
- Carmine Finelli
- Department of Internal Medicine, Ospedale Cav. R. Apicella - ASL Napoli 3 Sud, Via di Massa, 1, 80040 Pollena (Napoli), Italy
| |
Collapse
|
29
|
17-Aminogeldanamycin Inhibits Constitutive Nuclear Factor-Kappa B (NF-κB) Activity in Patient-Derived Melanoma Cell Lines. Int J Mol Sci 2020; 21:ijms21113749. [PMID: 32466509 PMCID: PMC7312877 DOI: 10.3390/ijms21113749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022] Open
Abstract
Melanoma remains incurable skin cancer, and targeting heat shock protein 90 (HSP90) is a promising therapeutic approach. In this study, we investigate the effect of 17-aminogeldanamycin, a potent HSP90 inhibitor, on nuclear factor-kappa B (NF-κB) activity in BRAFV600E and NRASQ61R patient-derived melanoma cell lines. We performed time-lapse microscopy and flow cytometry to monitor changes in cell confluence and viability. The NF-κB activity was determined by immunodetection of phospho-p65 and assessment of expression of NF-κB-dependent genes by quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, and enzyme-linked immunosorbent assay (ELISA). Constitutive activity of p65/NF-κB was evident in all melanoma cell lines. Differences in its level might be associated with genetic alterations in CHUK, IL1B, MAP3K14, NFKBIE, RIPK1, and TLR4, while differences in transcript levels of NF-κB-inducible genes revealed by PCR array might result from the contribution of other regulatory mechanisms. 17-Aminogeldanamycin markedly diminished the level of phospho-p65, but the total p65 protein level was unaltered, indicating that 17-aminogeldanamycin inhibited activation of p65/NF-κB. This conclusion was supported by significantly reduced expression of selected NF-κB-dependent genes: cyclin D1 (CCND1), C-X-C motif chemokine ligand 8 (CXCL8), and vascular endothelial growth factor (VEGF), as shown at transcript and protein levels, as well as secretion of IL-8 and VEGF. Our study indicates that 17-aminogeldanamycin can be used for efficient inhibition of NF-κB activity and the simultaneous diminution of IL-8 and VEGF levels in the extracellular milieu of melanoma.
Collapse
|
30
|
Emami N, Pakchin PS, Ferdousi R. Computational predictive approaches for interaction and structure of aptamers. J Theor Biol 2020; 497:110268. [PMID: 32311376 DOI: 10.1016/j.jtbi.2020.110268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
Aptamers are short single-strand sequences that can bind to their specific targets with high affinity and specificity. Usually, aptamers are selected experimentally via systematic evolution of ligands by exponential enrichment (SELEX), an evolutionary process that consists of multiple cycles of selection and amplification. The SELEX process is expensive, time-consuming, and its success rates are relatively low. To overcome these difficulties, in recent years, several computational techniques have been developed in aptamer sciences that bring together different disciplines and branches of technologies. In this paper, a complementary review on computational predictive approaches of the aptamer has been organized. Generally, the computational prediction approaches of aptamer have been proposed to carry out in two main categories: interaction-based prediction and structure-based predictions. Furthermore, the available software packages and toolkits in this scope were reviewed. The aim of describing computational methods and tools in aptamer science is that aptamer scientists might take advantage of these computational techniques to develop more accurate and more sensitive aptamers.
Collapse
Affiliation(s)
- Neda Emami
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Samadi Pakchin
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Ferdousi
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
31
|
Nguyen HD, Liao YC, Ho YS, Chen LC, Chang HW, Cheng TC, Liu D, Lee WR, Shen SC, Wu CH, Tu SH. The α9 Nicotinic Acetylcholine Receptor Mediates Nicotine-Induced PD-L1 Expression and Regulates Melanoma Cell Proliferation and Migration. Cancers (Basel) 2019; 11:E1991. [PMID: 31835799 PMCID: PMC6966517 DOI: 10.3390/cancers11121991] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023] Open
Abstract
Cigarette smoking is associated with an increased risk of melanoma metastasis. Smokers show higher PD-L1 expression and better responses to PD-1/PD-L1 inhibitors than nonsmokers. Here, we investigate whether nicotine, a primary constituent of tobacco, induces PD-L1 expression and promotes melanoma cell proliferation and migration, which is mediated by the α9 nicotinic acetylcholine receptor (α9-nAChR). α9-nAChR overexpression in melanoma using melanoma cell lines, human melanoma tissues, and assessment of publicly available databases. α9-nAChR expression was significantly correlated with PD-L1 expression, clinical stage, lymph node status, and overall survival (OS). Overexpressing or knocking down α9-nAChR in melanoma cells up- or downregulated PD-L1 expression, respectively, and affected melanoma cell proliferation and migration. Nicotine-induced α9-nAChR activity promoted melanoma cell proliferation through stimulation of the α9-nAChR-mediated AKT and ERK signaling pathways. In addition, nicotine-induced α9-nAchR activity promoted melanoma cell migration via activation of epithelial-mesenchymal transition (EMT). Moreover, PD-L1 expression was upregulated in melanoma cells after nicotine treatment via the transcription factor STAT3 binding to the PD-L1 promoter. These results highlight that nicotine-induced α9-nAChR activity promotes melanoma cell proliferation, migration, and PD-L1 upregulation. This study may reveal important insights into the mechanisms underlying nicotine-induced melanoma growth and metastasis through α9-nAChR-mediated carcinogenic signals and PD-L1 expression.
Collapse
Affiliation(s)
- Hai Duong Nguyen
- International Master Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - You-Cheng Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (Y.-C.L.); (W.-R.L.); (S.-C.S.)
| | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan; (Y.-S.H.); (L.-C.C.)
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan
- Department of Medical Laboratory, Taipei Medical University Hospital, Taipei 110, Taiwan;
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
| | - Li-Ching Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan; (Y.-S.H.); (L.-C.C.)
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Hui-Wen Chang
- Department of Medical Laboratory, Taipei Medical University Hospital, Taipei 110, Taiwan;
| | - Tzu-Chun Cheng
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
| | - Donald Liu
- Department of Dermatology, Taipei Medical University Shuang Ho Hospital, New Taipei City 237, Taiwan;
| | - Woan-Ruoh Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (Y.-C.L.); (W.-R.L.); (S.-C.S.)
- Department of Dermatology, Taipei Medical University Shuang Ho Hospital, New Taipei City 237, Taiwan;
- Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Shing-Chuan Shen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (Y.-C.L.); (W.-R.L.); (S.-C.S.)
- Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- International Master/PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 101, Taiwan
| | - Chih-Hsiung Wu
- Department of Surgery, EnChu Kong Hospital, New Taipei City 237, Taiwan;
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Shih-Hsin Tu
- Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
32
|
García-Aranda M, Redondo M. Immunotherapy: A Challenge of Breast Cancer Treatment. Cancers (Basel) 2019; 11:E1822. [PMID: 31756919 PMCID: PMC6966503 DOI: 10.3390/cancers11121822] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/16/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in women and is a leading cause of cancer death in women worldwide. Despite the significant benefit of the use of conventional chemotherapy and monoclonal antibodies in the prognosis of breast cancer patients and although the recent approval of the anti-PD-L1 antibody atezolizumab in combination with chemotherapy has been a milestone for the treatment of patients with metastatic triple-negative breast cancer, immunologic treatment of breast tumors remains a great challenge. In this review, we summarize current breast cancer classification and standard of care, the main obstacles that hinder the success of immunotherapies in breast cancer patients, as well as different approaches that could be useful to enhance the response of breast tumors to immunotherapies.
Collapse
Affiliation(s)
- Marilina García-Aranda
- Research Unit, Hospital Costa del Sol, Autovía A-7, km 187, 29603 Marbella, Spain;
- Research Network in Health Services in Chronic Diseases (Red de Investigación en Servicios de Salud en Enfermedades Crónicas, REDISSEC), Carlos III Health Institute (Instituto de Salud Carlos III). Av. de Monforte de Lemos, 5. 28029 Madrid, Spain
- Malaga Biomedical Research Institute (Instituto de Investigación Biomédica de Málaga, IBIMA), Calle Doctor Miguel Díaz Recio, 28. 29010 Málaga, Spain
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain
| | - Maximino Redondo
- Research Unit, Hospital Costa del Sol, Autovía A-7, km 187, 29603 Marbella, Spain;
- Research Network in Health Services in Chronic Diseases (Red de Investigación en Servicios de Salud en Enfermedades Crónicas, REDISSEC), Carlos III Health Institute (Instituto de Salud Carlos III). Av. de Monforte de Lemos, 5. 28029 Madrid, Spain
- Malaga Biomedical Research Institute (Instituto de Investigación Biomédica de Málaga, IBIMA), Calle Doctor Miguel Díaz Recio, 28. 29010 Málaga, Spain
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain
| |
Collapse
|
33
|
Hattinger CM, Patrizio MP, Magagnoli F, Luppi S, Serra M. An update on emerging drugs in osteosarcoma: towards tailored therapies? Expert Opin Emerg Drugs 2019; 24:153-171. [PMID: 31401903 DOI: 10.1080/14728214.2019.1654455] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: Current treatment of conventional and non-conventional high-grade osteosarcoma (HGOS) is based on the surgical removal of primary tumor and, when possible, of metastases and local reccurrence, together with systemic pre- and post-operative chemotherapy with drugs that have been used since decades. Areas covered: This review is intended to summarize the new agents and therapeutic strategies that are under clinical evaluation in HGOS, with the aim to increase the cure probability of this highly malignant bone tumor, which has not significantly improved during the last 30-40 years. The list of drugs, compounds and treatment modalities presented and discussed here has been generated by considering only those that are included in presently ongoing and recruiting clinical trials, or which have been completed in the last 2 years with reported results, on the basis of the information obtained from different and continuously updated databases. Expert opinion: Despite HGOS is a rare tumor, several clinical trials are presently evaluating different treatment strategies, which may hopefully positively impact on the outcome of patients who experience unfavorable prognosis when treated with conventional therapies.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Maria Pia Patrizio
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Federica Magagnoli
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Silvia Luppi
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Massimo Serra
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| |
Collapse
|