1
|
Elewa YHA, Abd Elwakil MM, Harashima H, Mohamed SKA, Zahran MH. Microenvironmental Changes in Mediastinal Fat-associated Lymphoid Clusters and Lungs in Early and Late Stages of Metastatic Lung Cancer Induction. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:1228-1243. [PMID: 37749682 DOI: 10.1093/micmic/ozad044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 09/27/2023]
Abstract
The prognosis of metastatic lung melanoma (MLM) has been reported to be poor. An increasing number of studies have reported the function of several immune cells in cancer regression. Although the function of mediastinal fat-associated lymphoid clusters (MFALCs) in the progression of inflammatory lung lesions has been previously reported, the association between MLM progression and MFALCs development has remained unexplored. Herein, we compared the microenvironmental changes in the lungs and MFALCs among phosphate-buffered saline (PBS) and cancer groups at early (1 week) and late (2 weeks) stages following the intravenous injection of B16-F10 melanoma cells into C57BL/6 mice. Except for lung CD4+ helper T-cells and Iba1+ macrophage populations of early stage, we observed a significant increase in the proliferating and immune cell (CD20+ B-lymphocytes, CD3+ T-lymphocytes, CD8+ cytotoxic T-cells, CD16+ natural killer (NK) cells populations, area of high endothelial venules, and lung lymphatic vessels in cancer groups at both the stages as compared with the PBS groups. Furthermore, a significant positive correlation was observed between immune cell populations in MFALCs and the lungs (B- and T-lymphocytes, and NK cells in both stages). Collectively, our findings suggest a promising cancer therapeutic strategy via targeting immune cells in MFALCs.
Collapse
Affiliation(s)
- Yaser Hosny Ali Elewa
- Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mahmoud M Abd Elwakil
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Sherif Kh A Mohamed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mahmoud Hosny Zahran
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
2
|
Evangelista L, Bianchi A, Annovazzi A, Sciuto R, Di Traglia S, Bauckneht M, Lanfranchi F, Morbelli S, Nappi AG, Ferrari C, Rubini G, Panareo S, Urso L, Bartolomei M, D’Arienzo D, Valente T, Rossetti V, Caroli P, Matteucci F, Aricò D, Bombaci M, Caponnetto D, Bertagna F, Albano D, Dondi F, Gusella S, Spimpolo A, Carriere C, Balma M, Buschiazzo A, Gallicchio R, Storto G, Ruffini L, Cervati V, Ledda RE, Cervino AR, Cuppari L, Burei M, Trifirò G, Brugola E, Zanini CA, Alessi A, Fuoco V, Seregni E, Deandreis D, Liberini V, Moreci AM, Ialuna S, Pulizzi S, De Rimini ML. ITA-IMMUNO-PET: The Role of [18F]FDG PET/CT for Assessing Response to Immunotherapy in Patients with Some Solid Tumors. Cancers (Basel) 2023; 15:cancers15030878. [PMID: 36765835 PMCID: PMC9913289 DOI: 10.3390/cancers15030878] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
AIM To examine the role of [18F]FDG PET/CT for assessing response to immunotherapy in patients with some solid tumors. METHODS Data recorded in a multicenter (n = 17), retrospective database between March and November 2021 were analyzed. The sample included patients with a confirmed diagnosis of a solid tumor who underwent serial [18F]FDG PET/CT (before and after one or more cycles of immunotherapy), who were >18 years of age, and had a follow-up of at least 12 months after their first PET/CT scan. Patients enrolled in clinical trials or without a confirmed diagnosis of cancer were excluded. The authors classified cases as having a complete or partial metabolic response to immunotherapy, or stable or progressive metabolic disease, based on a visual and semiquantitative analysis according to the EORTC criteria. Clinical response to immunotherapy was assessed at much the same time points as the serial PET scans, and both the obtained responses were compared. RESULTS The study concerned 311 patients (median age: 67; range: 31-89 years) in all. The most common neoplasm was lung cancer (56.9%), followed by malignant melanoma (32.5%). Nivolumab was administered in 46.3%, and pembrolizumab in 40.5% of patients. Baseline PET and a first PET scan performed at a median 3 months after starting immunotherapy were available for all 311 patients, while subsequent PET scans were obtained after a median 6, 12, 16, and 21 months for 199 (64%), 102 (33%), 46 (15%), and 23 (7%) patients, respectively. Clinical response to therapy was recorded at around the same time points after starting immunotherapy for 252 (81%), 173 (56%), 85 (27%), 40 (13%), and 22 (7%) patients, respectively. After a median 18 (1-137) months, 113 (36.3%) patients had died. On Kaplan-Meier analysis, metabolic responders on the first two serial PET scans showed a better prognosis than non-responders, while clinical response became prognostically informative from the second assessment after starting immunotherapy onwards. CONCLUSIONS [18F]FDG PET/CT could have a role in the assessment of response to immunotherapy in patients with some solid tumors. It can provide prognostic information and thus contribute to a patient's appropriate treatment. Prospective randomized controlled trials are mandatory.
Collapse
Affiliation(s)
- Laura Evangelista
- Nuclear Medicine Unit, Department of Medicine DIMED, University of Padua, 35129 Padua, Italy
- Correspondence:
| | - Andrea Bianchi
- Nuclear Medicine Unit, ASO S.Croce e Carle Cuneo, 12100 Cuneo, Italy
| | - Alessio Annovazzi
- Nuclear Medicine Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Rosa Sciuto
- Nuclear Medicine Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Silvia Di Traglia
- Nuclear Medicine Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Matteo Bauckneht
- Department of Health Sciences (DISSAL), University of Genova, 16126 Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Francesco Lanfranchi
- Department of Health Sciences (DISSAL), University of Genova, 16126 Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Silvia Morbelli
- Department of Health Sciences (DISSAL), University of Genova, 16126 Genova, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Anna Giulia Nappi
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Cristina Ferrari
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Giuseppe Rubini
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Stefano Panareo
- Nuclear Medicine Unit, Azienda Ospedaliero Universitaria di Modena, 41124 Modena, Italy
| | - Luca Urso
- Nuclear Medicine Unit, University of Ferrara, 44121 Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, University of Ferrara, 44121 Ferrara, Italy
| | - Davide D’Arienzo
- Nuclear Medicine Unit, Dept Servizi Sanitari, AORN Ospedali dei Colli, 80131 Naples, Italy
| | - Tullio Valente
- Radiology Department, AORN Ospedali dei Colli, 80131 Naples, Italy
| | - Virginia Rossetti
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST), 47014 Meldola, Italy
| | - Paola Caroli
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST), 47014 Meldola, Italy
| | - Federica Matteucci
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST), 47014 Meldola, Italy
| | - Demetrio Aricò
- Nuclear Medicine Unit, Humanitas Istituto Clinico Catanese, 95045 Misterbianco, Italy
| | - Michelangelo Bombaci
- Nuclear Medicine Unit, Humanitas Istituto Clinico Catanese, 95045 Misterbianco, Italy
| | - Domenica Caponnetto
- Nuclear Medicine Unit, Humanitas Istituto Clinico Catanese, 95045 Misterbianco, Italy
| | | | - Domenico Albano
- Nuclear Medicine Unit, University of Brescia, 25123 Brescia, Italy
| | - Francesco Dondi
- Nuclear Medicine Unit, University of Brescia, 25123 Brescia, Italy
| | - Sara Gusella
- Nuclear Medicine Department, Central Hospital Bolzano (SABES-ASDAA), 39100 Bolzano-Bozen, Italy
| | - Alessandro Spimpolo
- Nuclear Medicine Department, Central Hospital Bolzano (SABES-ASDAA), 39100 Bolzano-Bozen, Italy
| | - Cinzia Carriere
- Dermatology Department, Central Hospital Bolzano (SABES-ASDAA), 39100 Bolzano-Bozen, Italy
| | - Michele Balma
- Nuclear Medicine Unit, ASO S.Croce e Carle Cuneo, 12100 Cuneo, Italy
| | - Ambra Buschiazzo
- Nuclear Medicine Unit, ASO S.Croce e Carle Cuneo, 12100 Cuneo, Italy
| | - Rosj Gallicchio
- Nuclear Medicine Unit, IRCCS CROB Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Giovanni Storto
- Nuclear Medicine Unit, IRCCS CROB Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Livia Ruffini
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, 43126 Parma, Italy
| | - Veronica Cervati
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, 43126 Parma, Italy
| | - Roberta Eufrasia Ledda
- Department of Medicine and Surgery, Unit of Radiological Sciences, University of Parma, 43126 Parma, Italy
| | - Anna Rita Cervino
- Nuclear Medicine Unit, Veneto Institute Of Oncology IOV—IRCSS, 35128 Padua, Italy
| | - Lea Cuppari
- Nuclear Medicine Unit, Veneto Institute Of Oncology IOV—IRCSS, 35128 Padua, Italy
| | - Marta Burei
- Nuclear Medicine Unit, Veneto Institute Of Oncology IOV—IRCSS, 35128 Padua, Italy
| | - Giuseppe Trifirò
- Nuclear Medicine Unit, ICS MAUGERI SPA SB—IRCCS, 35128 Padua, Italy
| | | | | | - Alessandra Alessi
- Nuclear Medicine Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Valentina Fuoco
- Nuclear Medicine Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Ettore Seregni
- Nuclear Medicine Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Désirée Deandreis
- Nuclear Medicine Division, Department of Medical Sciences, University of Turin, 10124 Turin, Italy
| | - Virginia Liberini
- Nuclear Medicine Unit, ASO S.Croce e Carle Cuneo, 12100 Cuneo, Italy
- Nuclear Medicine Division, Department of Medical Sciences, University of Turin, 10124 Turin, Italy
| | - Antonino Maria Moreci
- Nuclear Medicine Unit, Az. Ospedaliera Ospedali Riuniti Villa Sofia-Cervello di Palermo, 90100 Palermo, Italy
| | - Salvatore Ialuna
- Nuclear Medicine Unit, Az. Ospedaliera Ospedali Riuniti Villa Sofia-Cervello di Palermo, 90100 Palermo, Italy
| | - Sabina Pulizzi
- Nuclear Medicine Unit, Az. Ospedaliera Ospedali Riuniti Villa Sofia-Cervello di Palermo, 90100 Palermo, Italy
| | - Maria Luisa De Rimini
- Nuclear Medicine Unit, Dept Servizi Sanitari, AORN Ospedali dei Colli, 80131 Naples, Italy
| |
Collapse
|
3
|
Khadela A, Chavda VP, Postwala H, Ephraim R, Apostolopoulos V, Shah Y. Configuring Therapeutic Aspects of Immune Checkpoints in Lung Cancer. Cancers (Basel) 2023; 15:543. [PMID: 36672492 PMCID: PMC9856297 DOI: 10.3390/cancers15020543] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/25/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Immune checkpoints are unique components of the body's defense mechanism that safeguard the body from immune responses that are potent enough to harm healthy body cells. When proteins present on the surface of T cells recognize and bind to the proteins present on other tumor cells, immune checkpoints are triggered. These proteins are called immunological checkpoints. The T cells receive an on/off signal when the checkpoints interact with companion proteins. This might avert the host's immune system from eliminating cancer cells. The standard care plan for the treatment of non-small cell lung cancer (NSCLC) has been revolutionized with the use of drugs targeting immune checkpoints, in particular programmed cell death protein 1. These drugs are now extended for their potential to manage SCLC. However, it is acknowledged that these drugs have specific immune related adverse effects. Herein, we discuss the use of immune checkpoint inhibitors in patients with NSCLC and SCLC, their outcomes, and future perspectives.
Collapse
Affiliation(s)
- Avinash Khadela
- Department of Pharmacology, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Humzah Postwala
- Pharm. D Section, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| | - Ramya Ephraim
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
- Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Yesha Shah
- Pharm. D Section, L. M. College of Pharmacy, Navrangpura, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
4
|
Altaf R, Jadoon SS, Muhammad SA, Ilyas U, Duan Y. Recent advances in immune checkpoint inhibitors for non-small lung cancer treatment. Front Oncol 2022; 12:1014156. [PMID: 36237320 PMCID: PMC9552217 DOI: 10.3389/fonc.2022.1014156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Lung cancer is one of the deadliest types of cancer responsible for thousands of cancer-related deaths. Its treatment has remained a challenge for researchers, but an increase in the knowledge of molecular pathways and biology of lung cancer has dramatically changed its management in recent decades. Immunotherapies and immunomodulation of lung cancer have previously failed for a long time but thanks to continuous research work and enthusiasm, now, this field is emerging as a novel effective therapy. Now, it is hope with potential benefits and promising results in the treatment of lung cancer. This review article focuses on immune checkpoints inhibitors: CTLA-4 inhibitors (ipilimumab and tremelimumab) and PDL-1 inhibitors (durvalumab and atezolizumab) that can be blocked to treat lung carcinoma. It is also focused on critically analyzing different studies and clinical trials to determine the potential benefits, risks, and adverse events associated with immunotherapeutic treatment.
Collapse
Affiliation(s)
- Reem Altaf
- Henan Provincial Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Department of Pharmacy, Iqra University Islamabad Campus, Islamabad, Pakistan
| | - Sarmad Sheraz Jadoon
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Syed Aun Muhammad
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
- *Correspondence: Syed Aun Muhammad, ; Umair Ilyas, ; Yongtao Duan,
| | - Umair Ilyas
- Department of Pharmaceutics, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
- *Correspondence: Syed Aun Muhammad, ; Umair Ilyas, ; Yongtao Duan,
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
- *Correspondence: Syed Aun Muhammad, ; Umair Ilyas, ; Yongtao Duan,
| |
Collapse
|
5
|
Cao L, Cao Z, Liu H, Liang N, Bing Z, Tian C, Li S. Detection of Potential Mutated Genes Associated with Common Immunotherapy Biomarkers in Non-Small-Cell Lung Cancer Patients. Curr Oncol 2022; 29:5715-5730. [PMID: 36005189 PMCID: PMC9406727 DOI: 10.3390/curroncol29080451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Microsatellite instability (MSI), high tumor mutation burden (TMB-H) and programmed cell death 1 ligand 1 (PD-L1) expression are hot biomarkers related to the improvement of immunotherapy response. Two cohorts of non-small-cell lung cancer (NSCLC) were collected and sequenced via targeted next-generation sequencing. Drug analysis was then performed on the shared genes using three different databases: Drugbank, DEPO and DRUGSURV. A total of 27 common genes were mutated in at least two groups of TMB-H-, MSI- and PD-L1-positive groups. AKT1, SMAD4, SCRIB and AXIN2 were severally involved in PI3K-activated, transforming growth factor beta (TGF-β)-activated, Hippo-repressed and Wnt-repressed pathways. This study provides an understanding of the mutated genes related to the immunotherapy biomarkers of NSCLC.
Collapse
Affiliation(s)
- Lei Cao
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Zhili Cao
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Hongsheng Liu
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Naixin Liang
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Caijuan Tian
- Tianjin Marvel Medical Laboratory, Tianjin Marvelbio Technology Co., Ltd., Tianjin 300381, China
| | - Shanqing Li
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
- Correspondence: ; Tel./Fax: +86-010-6915-2630
| |
Collapse
|
6
|
Kinoshita T, Terai H, Yaguchi T. Clinical Efficacy and Future Prospects of Immunotherapy in Lung Cancer. Life (Basel) 2021; 11:life11101029. [PMID: 34685400 PMCID: PMC8540292 DOI: 10.3390/life11101029] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/20/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022] Open
Abstract
The three major conventional treatments: surgery, chemotherapy, and radiation therapy, have been commonly performed for lung cancer. However, lung cancer is still the leading cause of cancer-related mortality. Immunotherapy has recently emerged as a very effective new treatment modality, and there is now growing enthusiasm for cancer immunotherapy worldwide. However, the results of clinical studies using immunotherapy are not always favorable. Understanding the steps involved in the recognition and eradication of cancer cells by the immune system seems essential to understanding why past immunotherapies have failed and how current therapies can be optimally utilized. In addition, the combination of immunotherapies, such as cancer vaccines and immune checkpoint inhibitors, as well as the combination of these therapies with three conventional therapies, may pave the way for personalized immunotherapy. In this review, we summarize the results of immunotherapies used in phase III clinical trials, including immune checkpoint inhibitors, and discuss the future prospects of immunotherapies in lung cancer treatment.
Collapse
Affiliation(s)
- Tomonari Kinoshita
- Division of General Thoracic Surgery, Department of Surgery, School of Medicine, Keio University, Tokyo 160-8582, Japan
- Correspondence: ; Tel.: +81-3-5363-3806
| | - Hideki Terai
- Division of Pulmonary Medicine, Department of Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan;
| | - Tomonori Yaguchi
- Center for Cancer Immunotherapy and Immunobiology, Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan;
| |
Collapse
|
7
|
Li WP, Yen CJ, Wu BS, Wong TW. Recent Advances in Photodynamic Therapy for Deep-Seated Tumors with the Aid of Nanomedicine. Biomedicines 2021; 9:69. [PMID: 33445690 PMCID: PMC7828119 DOI: 10.3390/biomedicines9010069] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Photodynamic therapy (PDT) works through photoactivation of a specific photosensitizer (PS) in a tumor in the presence of oxygen. PDT is widely applied in oncology to treat various cancers as it has a minimally invasive procedure and high selectivity, does not interfere with other treatments, and can be repeated as needed. A large amount of reactive oxygen species (ROS) and singlet oxygen is generated in a cancer cell during PDT, which destroys the tumor effectively. However, the efficacy of PDT in treating a deep-seated tumor is limited due to three main reasons: Limited light penetration depth, low oxygen concentration in the hypoxic core, and poor PS accumulation inside a tumor. Thus, PDT treatments are only approved for superficial and thin tumors. With the advancement of nanotechnology, PDT to treat deep-seated or thick tumors is becoming a reachable goal. In this review, we provide an update on the strategies for improving PDT with nanomedicine using different sophisticated-design nanoparticles, including two-photon excitation, X-ray activation, targeting tumor cells with surface modification, alteration of tumor cell metabolism pathways, release of therapeutic gases, improvement of tumor hypoxia, and stimulation of host immunity. We focus on the difficult-to-treat pancreatic cancer as a model to demonstrate the influence of advanced nanomedicine in PDT. A bright future of PDT application in the treatment of deep-seated tumors is expected.
Collapse
Affiliation(s)
- Wei-Peng Li
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Jui Yen
- Division of Hematology and Oncology, Department of Internal Medicine, Graduate Institute of Clinical Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan;
| | - Bo-Sheng Wu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Tak-Wah Wong
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
8
|
Wu Z, Man S, Sun R, Li Z, Wu Y, Zuo D. Recent advances and challenges of immune checkpoint inhibitors in immunotherapy of non-small cell lung cancer. Int Immunopharmacol 2020; 85:106613. [DOI: 10.1016/j.intimp.2020.106613] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
|
9
|
Liang H, Xu Y, Chen M, Zhong W, Wang M, Zhao J. Patterns of response in metastatic NSCLC during PD-1 or PD-L1 inhibitor therapy: Comparison of the RECIST 1.1 and iRECIST criteria. Thorac Cancer 2020; 11:1068-1075. [PMID: 32129934 PMCID: PMC7113040 DOI: 10.1111/1759-7714.13367] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/26/2022] Open
Abstract
Background Immunotherapy plays an important role in advanced non‐small cell lung cancer (NSCLC). However, radiological evaluation is challenging due to the potential inflammatory effects of immunotherapy, which can lead to atypical response patterns. Identifying these atypical responses is critical to making treatment decisions and prognostication. Methods We performed a retrospective analysis of consecutive advanced NSCLC patients treated with immunotherapy (alone or in combination). We collected patients' clinical and pathological data, analyzed the proportion of patients who continued immunotherapy beyond progressive disease (PD) per RECIST 1.1, and compared the differences in response patterns between the RECIST 1.1 and iRECIST criteria. Results A total of 43 patients treated at the Peking Union Medical College, China from January 2018 to April 2019 were included. Continued immunotherapy beyond PD per RECIST 1.1 was observed in 10 (33.3%, 10/30) patients, of which there were discordant assessments (30%, 3/10) between the RECIST 1.1 and iRECIST, which were evaluated as PD by RECIST 1.1 and immune unconfirmed PD by iRECIST. Among seven patients with immune confirmed PD, one (1/30, 3.3%) had pseudoprogression. Patients who continued immunotherapy beyond PD (n = 10) experienced significantly prolonged overall survival (not reached vs. 8.1 months: hazard ratio, 2.8; 95% confidence interval: 2.7–13.6, P = 0.03) compared with patients who did not continue immunotherapy beyond PD (n = 20). Conclusions RECIST 1.1 evaluation underestimated the benefit of immunotherapy. Further research is required to optimize iRECIST and establish some criteria for selecting patients who will benefit from continued immunotherapy beyond PD per RECIST 1.1.
Collapse
Affiliation(s)
- Hongge Liang
- Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yan Xu
- Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Minjiang Chen
- Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Zhong
- Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mengzhao Wang
- Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jing Zhao
- Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
10
|
Yang T, Shi X, Kang Y, Zhu M, Fan M, Zhang D, Zhang Y. Novel compounds TAD-1822-7-F2 and F5 inhibited HeLa cells growth through the JAK/Stat signaling pathway. Biomed Pharmacother 2018; 103:118-126. [PMID: 29649626 DOI: 10.1016/j.biopha.2018.03.174] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 10/17/2022] Open
Abstract
Cervical carcinoma remains the second most common malignancy with a high mortality rate among women worldwide. TAD-1822-7-F2 (F2) and TAD-1822-7-F5 (F5) are novel compounds synthesized on the chemical structure of taspine derivatives, and show an effective suppression for HeLa cells. Our study aims to confirm the potential targets of F2 and F5, and investigate the underlying mechanism of the inhibitory effect on HeLa cells. In this study, Real Time Cell Analysis and crystal violet staining assay were conducted to investigate the effect of F2 and F5 on HeLa cells proliferation. And the analytical methods of surface plasmon resonance and quartz crystal microbalance were established and employed to study the interaction between F2 and F5 and potential target protein JAK2, suggesting that both compounds have strong interaction with the JAK2 protein. Western blot analysis, immunofluorescence staining study and PCR was conducted to investigate the molecules of JAK/Stat signaling pathway. Interestingly, F2 and F5 showed diverse regulation for signaling molecules because of their different chemical structure. F2 increased the expression of JAK2 and downregulated the level of P-JAK1 and P-JAK2, and decreased P-Stat3 (Ser727). While F5 could increase the expression of JAK2 and naturally decrease the phosphorylation of JAK1 and Tyk2, and decreased the expression of P-Stat6. Moreover, F2 and F5 showed the same downregulation on the P-Stat3 (Tyr705). Therefore, F2 and F5 could target the JAK2 protein and prevent the phosphorylation of JAKs to suppress the phosphorylation of the downstream effector Stats, which suggested that F2 and F5 have great potential to be the inhibitors of the JAK/Stat signaling pathway.
Collapse
Affiliation(s)
- Tianfeng Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, PR China
| | - Xianpeng Shi
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, PR China
| | - Yuan Kang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, PR China
| | - Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, PR China
| | - Mengying Fan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, PR China
| | - Dongdong Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, PR China.
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, PR China.
| |
Collapse
|
11
|
Tackling Cancer Resistance by Immunotherapy: Updated Clinical Impact and Safety of PD-1/PD-L1 Inhibitors. Cancers (Basel) 2018; 10:cancers10020032. [PMID: 29370105 PMCID: PMC5836064 DOI: 10.3390/cancers10020032] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/21/2018] [Accepted: 01/23/2018] [Indexed: 02/07/2023] Open
Abstract
Cancer therapy has been constantly evolving with the hope of finding the most effective agents with the least toxic effects to eradicate tumors. Cancer immunotherapy is currently among the most promising options, fulfilling this hope in a wide range of tumors. Immunotherapy aims to activate immunity to fight cancer in a very specific and targeted manner; however, some abnormal immune reactions known as immune-related adverse events (IRAEs) might occur. Therefore, many researchers are aiming to define the most proper protocols for managing these complications without interfering with the anticancer effect. One of these targeted approaches is the inhibition of the interaction between the checkpoint protein, programmed death-receptor 1 (PD-1), and its ligand, programmed death-ligand 1 (PD-L1), via a class of antibodies known as PD-1/PD-L1 inhibitors. These antibodies achieved prodigious success in a wide range of malignancies, including those where optimal treatment is not yet fully identified. In this review, we have critically explored and discussed the outcome of the latest PD-1 and PD-L1 inhibitor studies in different malignancies compared to standard chemotherapeutic alternatives with a special focus on the clinical efficacy and safety. The approval of the clinical applications of nivolumab, pembrolizumab, atezolizumab, avelumab, and durvalumab in the last few years clearly highlights the hopeful future of PD-1/PD-L1 inhibitors for cancer patients. These promising results of PD-1/PD-L1 inhibitors have encouraged many ongoing preclinical and clinical trials to explore the extent of antitumor activity, clinical efficacy and safety as well as to extend their applications.
Collapse
|
12
|
Zhang X, Zhang Y, Xu J, Wang H, Zheng X, Lou Y, Han B. Antigen presentation of the Oct4 and Sox2 peptides by CD154-activated B lymphocytes enhances the killing effect of cytotoxic T lymphocytes on tumor stem-like cells derived from cisplatin-resistant lung cancer cells. J Cancer 2018; 9:367-374. [PMID: 29344283 PMCID: PMC5771344 DOI: 10.7150/jca.20821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 11/21/2017] [Indexed: 12/11/2022] Open
Abstract
The present study investigated whether antigen presentation of the Oct4 and Sox2 peptides by CD154-activated B lymphocytes can enhance the killing effect of CD8+ cytotoxic T lymphocytes (CTLs) on lung stem-like cancer cells (SLCCs). The CTLs were generated using an accelerated co-cultured dendritic cells (DC) (acDC) assay by incubating human peripheral blood mononuclear cells (PBMCs) from non-small-cell lung cancer patients with antigen peptides of Oct4 and Sox2 in the presence of several DC-activating agents. CD154+ NIH3T3 cells prepared by CD154 lentiviral transfection were used as feeder layer to activate primary B cells (CD19+) obtained from PBMCs. Activated B cells were co-cultured with CTLs to present antigen peptides of Oct4 and Sox2. CTLs co-cultured with activated B cells were evaluated for the levels of secreted inflammatory cytokines using ELISA. In addition, the killing effect of the CTLs on SLCCs derived from cisplatin-resistant strain of human lung cancer cell line PC9 was evaluated by flow cytometry using CFSE labeling of the target cells. After the acDC assay, the PBMCs exhibited a significant (p<0.01) increase in the population of CD8+/CD3+ cells, indicating successful preparation of CTLs. The primary B cells cultured on the CD154+ NIH3T3 feeder layer resulted in significant (p<0.01) increase in the proportions of population expressing CD80, CD86, or HLA-A, indicating successful activation of the B cells. The co-culture of CTLs with CD154-activated B cells presenting the Oct4 and Sox2 peptides caused significant increase in the levels of secretory inflammatory cytokines and exhibited enhanced killing of the SLCCs derived from cisplatin-resistant PC9 cells. Antigen presentation of the Oct4 and Sox2 peptides by CD154-activated B cells can enhance the killing effect of CTLs towards lung SLCCs.
Collapse
Affiliation(s)
- Xueyan Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Yanwei Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Jianlin Xu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Huimin Wang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Xiaoxuan Zheng
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Yuqing Lou
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Baohui Han
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| |
Collapse
|
13
|
Tazdait M, Mezquita L, Lahmar J, Ferrara R, Bidault F, Ammari S, Balleyguier C, Planchard D, Gazzah A, Soria JC, Marabelle A, Besse B, Caramella C. Patterns of responses in metastatic NSCLC during PD-1 or PDL-1 inhibitor therapy: Comparison of RECIST 1.1, irRECIST and iRECIST criteria. Eur J Cancer 2017; 88:38-47. [PMID: 29182990 DOI: 10.1016/j.ejca.2017.10.017] [Citation(s) in RCA: 249] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 10/22/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors are an important tool in the therapeutic strategy against metastatic non-small cell lung cancer (NSCLC); however, radiological evaluation is challenging due to the emergence of atypical patterns of responses. Several evaluation criteria have been proposed, such as the Response Evaluation Criteria in Solid Tumours (RECIST), version 1.1, immune -related RECIST (irRECIST) and iRECIST, but have not been systematically compared in a homogeneous population. PATIENTS AND METHODS We conducted a monocentric retrospective analysis of consecutive advanced NSCLC patients treated with an anti-programmed cell death-1 or anti-program death-ligand 1. Response patterns and the discordance between RECIST 1.1, irRECIST and iRECIST guidelines were described, and associations of response patterns and clinical outcome were explored. RESULTS Overall, 160 patients treated between February 2013 and October 2016 were included. Atypical responses were observed in 20 patients (13%), including eight pseudoprogressions (PsPDs) (5%) and 12 dissociated responses (8%). Thirteen of the 20 patients demonstrated clinical benefit. Per the RECIST 1.1, 37 patients (23%) showed an objective response or stable disease, and 123 patients (77%) exhibited progression. Eighty progressive patients were assessable for irRECIST and iRECIST: 15 patients were assessed differently; however, only three (3.8%) mismatches with a theoretical impact on the therapeutic decision were identified. Patients with PsPD or dissociated response had higher overall survival than patients with true progression. CONCLUSION Atypical responses (PsPD/dissociated response) occurred in 13% of NSCLC patients under immune checkpoint inhibitors. Based on survival analyses, the RECIST 1.1 evaluation underestimated the benefit of immune checkpoint inhibitors in 11% of the progressive patients. Immune-related RECIST and iRECIST identified these unconventional responses, with a 3.8% discrepancy rate.
Collapse
Affiliation(s)
- M Tazdait
- Department of Radiology, Gustave Roussy Cancer Campus, Villejuif, France
| | - L Mezquita
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - J Lahmar
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - R Ferrara
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - F Bidault
- Department of Radiology, Gustave Roussy Cancer Campus, Villejuif, France
| | - S Ammari
- Department of Radiology, Gustave Roussy Cancer Campus, Villejuif, France
| | - C Balleyguier
- Department of Radiology, Gustave Roussy Cancer Campus, Villejuif, France
| | - D Planchard
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - A Gazzah
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - J C Soria
- Department of Drug Development (DITEP), Gustave Roussy Institute, Villejuif, France
| | - A Marabelle
- Department of Drug Development (DITEP), Gustave Roussy Institute, Villejuif, France
| | - B Besse
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - C Caramella
- Department of Radiology, Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
14
|
Song H, Liu S, Zhao Z, Sun W, Wei X, Ma X, Zhao P, Gao D. Increased cycles of DC/CIK immunotherapy decreases frequency of Tregs in patients with resected NSCLC. Int Immunopharmacol 2017; 52:197-202. [PMID: 28941416 DOI: 10.1016/j.intimp.2017.09.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 01/26/2023]
Abstract
Regulatory T cells (Tregs) suppress antitumor immune responses. Cycles of Dendritic cells (DC) vaccination combined with cytokine-induced killer (CIK) cells (DC/CIK) treatment were significantly related with good prognosis. Therefore, we investigated whether increased cycles of immunotherapy could decrease frequency of Tregs in patients with resected non-small cell lung cancer (NSCLC). Previous study from our laboratory has determined that the optimal cutoff point of the cycle count was 3cycles. We examined the levels of Tregs and the related cytokines by flow cytometric and cytokine analysis in these patients after more than (≥) 3cycles or less than (<) 3cycles of DC/CIK cell treatment. Significant reduction of Tregs frequency, Treg-generated cytokines level and recurrence rate were presented in patients received with ≥3cycles of DC/CIK cell treatment compared with patients with <3cycles of treatment. Interestingly, Tregs frequency and the related cytokines level were similar between patients suffered tumor recurrence and patients without recurrence in both groups. Together, our findings reveal that increased cycle count of DC/CIK cell immunotherapy contribute to decline of Tregs frequency and cancer recurrence rate in patients with resected NSCLC.
Collapse
Affiliation(s)
- Haiping Song
- Department of Oncology, The Affiliated Central Hospital of Qingdao University, 127 Siliu South Road, Qingdao 266042, China
| | - Shujuan Liu
- Department of Oncology, The Affiliated Central Hospital of Qingdao University, 127 Siliu South Road, Qingdao 266042, China
| | - Ziyun Zhao
- Department of Clinical Laboratory, The Affiliated Central Hospital of Qingdao University, 127 Siliu South Road, Qingdao 266042, China
| | - Weihong Sun
- Biotherapy Center, The Affiliated Central Hospital of Qingdao University, 127 Siliu South Road, Qingdao 266042, China.
| | - Xiaofang Wei
- Biotherapy Center, The Affiliated Central Hospital of Qingdao University, 127 Siliu South Road, Qingdao 266042, China
| | - Xuezhen Ma
- Department of Oncology, The Affiliated Central Hospital of Qingdao University, 127 Siliu South Road, Qingdao 266042, China.
| | - Peng Zhao
- Biotherapy Center, The Affiliated Central Hospital of Qingdao University, 127 Siliu South Road, Qingdao 266042, China
| | - Daiqing Gao
- Biotherapy Center, The Affiliated Central Hospital of Qingdao University, 127 Siliu South Road, Qingdao 266042, China
| |
Collapse
|
15
|
Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, Lin NU, Litière S, Dancey J, Chen A, Hodi FS, Therasse P, Hoekstra OS, Shankar LK, Wolchok JD, Ballinger M, Caramella C, de Vries EGE. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 2017; 18:e143-e152. [PMID: 28271869 PMCID: PMC5648544 DOI: 10.1016/s1470-2045(17)30074-8] [Citation(s) in RCA: 1666] [Impact Index Per Article: 208.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/25/2016] [Accepted: 11/30/2016] [Indexed: 12/11/2022]
Abstract
Tumours respond differently to immunotherapies compared with chemotherapeutic drugs, raising questions about the assessment of changes in tumour burden-a mainstay of evaluation of cancer therapeutics that provides key information about objective response and disease progression. A consensus guideline-iRECIST-was developed by the RECIST working group for the use of modified Response Evaluation Criteria in Solid Tumours (RECIST version 1.1) in cancer immunotherapy trials, to ensure consistent design and data collection, facilitate the ongoing collection of trial data, and ultimate validation of the guideline. This guideline describes a standard approach to solid tumour measurements and definitions for objective change in tumour size for use in trials in which an immunotherapy is used. Additionally, it defines the minimum datapoints required from future trials and those currently in development to facilitate the compilation of a data warehouse to use to later validate iRECIST. An unprecedented number of trials have been done, initiated, or are planned to test new immune modulators for cancer therapy using a variety of modified response criteria. This guideline will allow consistent conduct, interpretation, and analysis of trials of immunotherapies.
Collapse
Affiliation(s)
- Lesley Seymour
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada.
| | | | | | - Robert Ford
- Clinical Trials Imaging Consulting, LLC, Belle Mead, NJ, USA
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA
| | - Sumithra Mandrekar
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Janet Dancey
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Alice Chen
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
| | - Lalitha K Shankar
- Diagnostic Imaging Branch, National Cancer Institute, Bethesda, MD, USA
| | - Jedd D Wolchok
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcus Ballinger
- Weill Cornell Medical and Graduate Colleges, New York, NY, USA; Ludwig Institute for Cancer Research, New York, NY, USA; Genentech Inc, San Francisco, CA, USA
| | | | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
16
|
Hamilton G, Rath B. Avelumab: combining immune checkpoint inhibition and antibody-dependent cytotoxicity. Expert Opin Biol Ther 2017; 17:515-523. [PMID: 28274143 DOI: 10.1080/14712598.2017.1294156] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Immune checkpoint inhibition holds great promise for selected tumors. The human monoclonal antibody (mAB) avelumab is directed to programmed death ligand-1 (PD-L1) and is supposed to inhibit the immunosuppressive PD-L1/PD-1 interaction and, furthermore, effect antibody-dependent cytotoxicity (ADCC) lysis of tumor cells. Areas covered: This article presents an overview of the current means to activate the antitumor immune defense by targeting PD-1 or PD-L1 with mABs and their possible role in ADCC-mediated tumor cell elimination. Expert opinion: Avelumab contains a Fc region which can bind cognate receptors on immune effector cells and induce ADCC-mediated tumor cell lysis, in contrast to other mABs directed to PD-1/PD-L1 which lack the ability to trigger ADCC due to belonging to the IgG4 subclass or possessing a mutated Fc region. Preclinical and clinical data indicate that avelumab can be safely administered to cancer patients with a toxicity profile comparable to other mABs and without lysis of PD-L1-positive activated immune cells. This antibody yielded durable responses in a phase II trial in advanced Merkel cell carcinoma patients. Tumor cell lysis by avelumab prevents cells from resorting to alternative checkpoints as shown by targeting PD-1 and the upregulation of TIM-3.
Collapse
Affiliation(s)
- Gerhard Hamilton
- a Department of Surgery , Medical University of Vienna , Vienna , Austria
| | - Barbara Rath
- a Department of Surgery , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
17
|
Ren J, Gwin WR, Zhou X, Wang X, Huang H, Jiang N, Zhou L, Agarwal P, Hobeika A, Crosby E, Hartman ZC, Morse MA, H Eng K, Lyerly HK. Adaptive T cell responses induced by oncolytic Herpes Simplex Virus-granulocyte macrophage-colony-stimulating factor therapy expanded by dendritic cell and cytokine-induced killer cell adoptive therapy. Oncoimmunology 2016; 6:e1264563. [PMID: 28507788 DOI: 10.1080/2162402x.2016.1264563] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/07/2016] [Accepted: 11/18/2016] [Indexed: 02/08/2023] Open
Abstract
Purpose: Although local oncolytic viral therapy (OVT) may enhance tumor lysis, antigen release, and adaptive immune responses, systemic antitumor responses post-therapy are limited. Adoptive immunotherapy with autologous dendritic cells (DC) and cytokine-induced killer cells (DC-CIK) synergizes with systemic therapies. We hypothesized that OVT with Herpes Simplex Virus-granulocyte macrophage-colony-stimulating factor (HSV-GM-CSF) would induce adaptive T cell responses that could be expanded systemically with sequential DC-CIK therapy. Patients and Methods: We performed a pilot study of intratumoral HSV-GM-CSF OVT followed by autologous DC-CIK cell therapy. In addition to safety and clinical endpoints, we monitored adaptive T cell responses by quantifying T cell receptor (TCR) populations in pre-oncolytic therapy, post-oncolytic therapy, and after DC-CIK therapy. Results: Nine patients with advanced malignancy were treated with OVT (OrienX010), of whom seven experienced stable disease (SD). Five of the OVT treated patients underwent leukapheresis, generation, and delivery of DC-CIKs, and two had SD, whereas three progressed. T cell receptor sequencing of TCR β sequences one month after OVT therapy demonstrates a dynamic TCR repertoire in response to OVT therapy in the majority of patients with the systematic expansion of multiple T cell clone populations following DC-CIK therapy. This treatment was well tolerated and long-term event free and overall survival was observed in six of the nine patients. Conclusions: Strategies inducing the local activation of tumor-specific immune responses can be combined with adoptive cellular therapies to expand the adaptive T cell responses systemically and further studies are warranted.
Collapse
Affiliation(s)
- Jun Ren
- Beijing Key Lab for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - William R Gwin
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Xinna Zhou
- Beijing Key Lab for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Wang
- Beijing Key Lab for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hongyan Huang
- Beijing Key Lab for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ni Jiang
- Beijing Key Lab for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lei Zhou
- Beijing Key Lab for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Pankaj Agarwal
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Erika Crosby
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Zachary C Hartman
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Michael A Morse
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Kevin H Eng
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - H Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
18
|
Jeremic B, Cihoric N, Dubinsky P, Filipovic N. Adjuvant immunotherapy in resected early non-small cell lung cancer-battle lost, hopefully not the war! J Thorac Dis 2016; 8:1886-90. [PMID: 27618979 DOI: 10.21037/jtd.2016.07.11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Branislav Jeremic
- Institute of Lung Diseases, Sremska Kamenica, Kragujevac, Serbia;; BioIRC Centre for Biomedical Research, Kragujevac, Serbia
| | - Nikola Cihoric
- Department of Radiation Oncology, Inselspital, Bern, Switzerland
| | - Pavol Dubinsky
- University Hospital to East Slovakia Institute of Oncology, Kosice, Slovakia
| | | |
Collapse
|
19
|
Implications of MDSCs-targeting in lung cancer chemo-immunotherapeutics. Pharmacol Res 2016; 110:25-34. [DOI: 10.1016/j.phrs.2016.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/23/2016] [Accepted: 05/04/2016] [Indexed: 12/23/2022]
|
20
|
Conway EM, Pikor LA, Kung SHY, Hamilton MJ, Lam S, Lam WL, Bennewith KL. Macrophages, Inflammation, and Lung Cancer. Am J Respir Crit Care Med 2016; 193:116-30. [PMID: 26583808 DOI: 10.1164/rccm.201508-1545ci] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lung cancer is the leading cause of cancer mortality worldwide, and at only 18%, it has one of the lowest 5-year survival rates of all malignancies. With its highly complex mutational landscape, treatment strategies against lung cancer have proved largely ineffective. However with the recent success of immunotherapy trials in lung cancer, there is renewed enthusiasm in targeting the immune component of tumors. Macrophages make up the majority of the immune infiltrate in tumors and are a key cell type linking inflammation and cancer. Although the mechanisms through which inflammation promotes cancer are not fully understood, two connected hypotheses have emerged: an intrinsic pathway, driven by genetic alterations that lead to neoplasia and inflammation, and an extrinsic pathway, driven by inflammatory conditions that increase cancer risk. Here, we discuss the contribution of macrophages to these pathways and subsequently their roles in established tumors. We highlight studies investigating the association of macrophages with lung cancer prognosis and discuss emerging therapeutic strategies for targeting macrophages in the tumor microenvironment.
Collapse
Affiliation(s)
- Emma M Conway
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Larissa A Pikor
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Sonia H Y Kung
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Melisa J Hamilton
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Stephen Lam
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Wan L Lam
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Kevin L Bennewith
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| |
Collapse
|
21
|
Steven A, Fisher SA, Robinson BW. Immunotherapy for lung cancer. Respirology 2016; 21:821-33. [PMID: 27101251 DOI: 10.1111/resp.12789] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 01/22/2016] [Accepted: 02/09/2016] [Indexed: 12/13/2022]
Abstract
Treatment of lung cancer remains a challenge, and lung cancer is still the leading cause of cancer-related mortality. Immunotherapy has previously failed in lung cancer but has recently emerged as a very effective new therapy, and there is now growing worldwide enthusiasm in cancer immunotherapy. We summarize why immune checkpoint blockade therapies have generated efficacious and durable responses in clinical trials and why this has reignited interest in this field. Cancer vaccines have also been explored in the past with marginal success. Identification of optimal candidate neoantigens may improve cancer vaccine efficacy and may pave the way to personalized immunotherapy, alone or in combination with other immunotherapy such as immune checkpoint blockade. Understanding the steps in immune recognition and eradication of cancer cells is vital to understanding why previous immunotherapies failed and how current therapies can be used optimally. We hold an optimistic view for the future prospect in lung cancer immunotherapy.
Collapse
Affiliation(s)
- Antonius Steven
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia.,National Centre for Asbestos Related Diseases (NCARD), Perth, Western Australia, Australia
| | - Scott A Fisher
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia.,National Centre for Asbestos Related Diseases (NCARD), Perth, Western Australia, Australia
| | - Bruce W Robinson
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia.,National Centre for Asbestos Related Diseases (NCARD), Perth, Western Australia, Australia
| |
Collapse
|
22
|
Zhao L, Wang WJ, Zhang JN, Zhang XY. 5-Fluorouracil and interleukin-2 immunochemotherapy enhances immunogenicity of non-small cell lung cancer A549 cells through upregulation of NKG2D ligands. Asian Pac J Cancer Prev 2016; 15:4039-44. [PMID: 24935593 DOI: 10.7314/apjcp.2014.15.9.4039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to investigate the anti-cancer effects and mechanisms of immunochemotherapy of 5-fluorouracil (5-FU) and interleukin-2 (IL-2) on non-small cell lung cancer (NSCLC) A549 cells. MATERIALS AND METHODS In order to detect whether 5-FU+IL-2 could effectively inhibit tumor growth in vivo, we established an A549-bearing nude mouse model. The cytotoxicity of natural killer (NK) cells was evaluated using a standard chromium release assay. To evaluate the relevance of NK cells in 5-FU+IL-2- mediated tumor inhibitory effects, we depleted NK cells in A549-bearing mice by injecting anti-asialo-GM-1 antibodies. Effects of 5-FU+IL-2 on the expression and promoter activity of NKG2D ligands (MICA/MICB) in A549 cells in vitro were also assessed. RESULTS In A549-bearing nude mice, combination therapy significantly inhibited tumor growth in comparison with monotherapy with 5-FU or IL-2 and enhanced the recognition and lysis of tumor cells by NK cells. Further study of mechanisms showed that NK cells played a vital role in the anticancer immune response of 5-FU+IL-2 immunochemotherapy. In addition, the combination therapy synergistically stimulated the expression and promoter activity of MICA/MICB. CONCLUSIONS 5-FU and IL-2 immunochemotherapy significantly inhibited tumor growth and activated NK cytotoxicity in vivo, and these effects were partly impaired after depleting NK cells in tumor-bearing mice. Combination treatment of 5-FU and IL-2 upregulated the expression and the promoter activity of MICA/MICB in A549 cells, which enhanced the recognition of A549 cells by NK cells. All of the data indicated that immunochemotherapy of 5-FU and IL-2 may provide a new treatment option for patients with lung cancer.
Collapse
Affiliation(s)
- Lei Zhao
- Institute of Frontier Medical Science, Jilin University, Changchun, Jilin, China E-mail :
| | | | | | | |
Collapse
|
23
|
Zhang J, Liu J, Chen H, Wu W, Li X, Wu Y, Wang Z, Zhang K, Li Y, Weng Y, Liao H, Gu L. Specific immunotherapy generates CD8(+) CD196(+) T cells to suppress lung cancer growth in mice. Immunol Res 2016; 64:1033-40. [PMID: 26910585 DOI: 10.1007/s12026-016-8793-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
That specific immunotherapy can inhibit cancer growth has been recognized; its efficiency is to be improved. This study aimed to inhibit lung cancer (LC) growth in a mouse model by using an LC-specific vaccination. In this study, a LC mouse model was created by adoptive transplantation with LC cells. The tumor-bearing mice were vaccinated with LC cell extracts plus adjuvant TNBS or adoptive transplantation with specific CD8(+) CD196(+) T cells. The results showed that the vaccination with LC extracts (LCE)/TNBS markedly inhibited the LC growth and induced CD8(+) CD196(+) T cells in LC tissue and the spleen. These CD8(+) CD196(+) T cells proliferated and produce high levels of perforin upon exposure to LCE and specifically induced LC cell apoptosis. Exposure to TNBS induced RAW264.7 cells to produce macrophage inflammatory protein-3α; the latter activated signal transducer and activator of transcription 3 and further induced perforin expression in the CD8(+) CD196(+) T cells. Adoptive transfer with specific CD8(+) CD196(+) T cells suppressed LC growth in mice. In conclusion, immunization with LC extracts and TNBS can induce LC-specific CD8(+) CD196(+) T cells in LC-bearing mice and inhibit LC growth.
Collapse
Affiliation(s)
- Jian Zhang
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Jing Liu
- Infectious Disease Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Huiguo Chen
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Weibin Wu
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Xiaojun Li
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Yonghui Wu
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Zhigang Wang
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Kai Zhang
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Yun Li
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Yimin Weng
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Hongying Liao
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Lijia Gu
- Thoracic Surgery Department, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou, 510630, China.
| |
Collapse
|
24
|
Zhang Q, Wu YL. Safety and efficacy of targeted agents monotherapy in advanced NSCLC. Expert Rev Clin Pharmacol 2015; 9:143-55. [PMID: 26558941 DOI: 10.1586/17512433.2016.1101340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The emergence of targetted therapy has revolutionised the treatment of advanced NSCLC. Increasing numbers of driver genes and related targetted agents have supplied more powerful weapons for conquering NSCLC. Depending on whether there are clear targets and therapeutic effects, we can now rank targetted agents into three categories: agents with explicit targets and pure effects, agents with theoretical targets but with no effective biomarkers, agents with vague targets and lower effects. The latest clinical data on the safety and efficacy of monotherapy with three kinds of agents will be reviewed respectively in this article.
Collapse
Affiliation(s)
- Qi Zhang
- a Graduate School , Southern Medical University , Guangzhou , Guangdong , PR China.,b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , Guangdong , PR China
| | - Yi-Long Wu
- b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , Guangdong , PR China
| |
Collapse
|
25
|
Tang C, Wang X, Soh H, Seyedin S, Cortez MA, Krishnan S, Massarelli E, Hong D, Naing A, Diab A, Gomez D, Ye H, Heymach J, Komaki R, Allison JP, Sharma P, Welsh JW. Combining radiation and immunotherapy: a new systemic therapy for solid tumors? Cancer Immunol Res 2015; 2:831-8. [PMID: 25187273 DOI: 10.1158/2326-6066.cir-14-0069] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
With the recent success of checkpoint inhibitors and other immunomodulating agents, there has been renewed interest in the combination of such agents with radiation. The biologic premise behind such a strategy is that the tumor-antigen release achieved by localized radiation will promote specific tumor targeting by the adaptive immune system, which can be augmented further by systemic immune-stimulating agents. In this manner, clinicians hope to induce a phenomenon known as the abscopal effect, whereby localized radiation results in immune-mediated tumor regression in disease sites well outside of the radiation field. Herein, we present a comprehensive overview of the early clinical and preclinical evidence behind this approach.
Collapse
Affiliation(s)
- Chad Tang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaohong Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hendrick Soh
- University of California Irvine School of Medicine, Irvine, California
| | - Steven Seyedin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunil Krishnan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Erminia Massarelli
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Gomez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Huiping Ye
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ristuko Komaki
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Padmanee Sharma
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
26
|
Immature myeloid cells and tolerogenic cytokine profile in lung adenocarcinoma metastatic lymph nodes assessed by endobronchial ultrasound. Tumour Biol 2015; 37:953-61. [DOI: 10.1007/s13277-015-3885-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 08/03/2015] [Indexed: 01/15/2023] Open
|
27
|
Reinmuth N, Heigener DF. Immunotherapy. Lung Cancer 2015. [DOI: 10.1183/2312508x.10010814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
28
|
Wang M, Cao JX, Liu YS, Xu BL, Li D, Zhang XY, Li JL, Liu JL, Wang HB, Wang ZX. Evaluation of tumour vaccine immunotherapy for the treatment of advanced non-small cell lung cancer: a systematic meta-analysis. BMJ Open 2015; 5:e006321. [PMID: 25872936 PMCID: PMC4401843 DOI: 10.1136/bmjopen-2014-006321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Our meta-analysis performed a systematic evaluation on the therapeutic efficacy and safety of tumour vaccines for the treatment of advanced non-small cell lung cancer (NSCLC). DESIGN Systematic review and meta-analysis of randomised controlled trials (RCT). DATA SOURCES PubMed, the Cochrane Center Register of Controlled Trials, Science Direct and EMBASE were searched from January 1980 until January 2015. ELIGIBILITY CRITERIA FOR SELECTING STUDIES RCT were included; the control arm had to receive either placebo or chemotherapy or no treatment. MAIN OUTCOME MEASURES The quality of the data from individual papers was assessed for overall survival (OS), clinical response rate and side effects. RESULTS Overall, 11 RCT of advanced NSCLC with a total of 3986 patients were conducted for meta-analysis. The results showed that the vaccine arm significantly extended primary endpoint median overall survival compared with control group (p<0.00001) (HR 0.760; 95% CI 0.644 to 0.896; p=0.001). Three subgroup patients with tumour vaccine at 1-year, 2-year and 3-year survival rates also gained significant benefits compared with their corresponding control group (p=0.0004, 0.03 and 0.19, respectively). Besides, a significant improvement in median time to progression (TTP), median progression-free survival (PFS) and a trend of improvement in objective response rate were observed after tumour vaccine treatment (p=0.001, 0.005 and 0.05, respectively; median PFS HR 0.842; 95% CI 0.744 to 0.954; p=0.007). A few severe adverse effects occurred in the tumour vaccine group, but fewer side effects were observed in the vaccine group compared with the control group (p<0.00001). CONCLUSIONS Taken together, NSCLC tumour vaccines markedly prolong median OS (p<0.00001), median TTP (p=0.001) and median PFS (p=0.005), improve clinical response rate (p=0.05) and lessen adverse side effects (p<0.00001). Our meta-analysis suggests tumour vaccines improve the efficacy of the treatment, and also provide superiority in treatment of patients with advanced NSCLC among a variety of immunotherapy strategies.
Collapse
Affiliation(s)
- Min Wang
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Jun-Xia Cao
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Yi-Shan Liu
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Bei-Lei Xu
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Duo Li
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Xiao-Yan Zhang
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Jun-Li Li
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Jin-Long Liu
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Hai-Bo Wang
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Zheng-Xu Wang
- Biotherapy Center, General Hospital of Beijing Military Command, Beijing, People's Republic of China
| |
Collapse
|
29
|
Scagliotti GV, Bironzo P, Vansteenkiste JF. Addressing the unmet need in lung cancer: The potential of immuno-oncology. Cancer Treat Rev 2015; 41:465-75. [PMID: 25936526 DOI: 10.1016/j.ctrv.2015.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/19/2015] [Accepted: 04/02/2015] [Indexed: 12/15/2022]
Abstract
Chemotherapy is currently the standard of care for non-oncogene-driven advanced non-small cell lung cancer (NSCLC). Due to improvements in chemotherapeutic choices and supportive care, patients currently typically undergo multiple lines of chemotherapy as their disease progresses. Although treatments have improved over recent years, limited benefits are seen, especially in patients receiving later-line chemotherapy, as response rates can be low, response duration short and survival poor. Furthermore, only a small percentage of patients derive benefit from later-line therapy, with most experiencing deteriorating quality of life and significant toxicities. More recently, molecular targeted therapies have provided improvements in outcomes. However, these treatments only offer a clear benefit in subsets of tumours harbouring the appropriate genomic alteration (mutation, amplification, translocation). Most of the genomic abnormalities susceptible to therapeutic intervention are detected in adenocarcinoma, mainly in never smokers, while alterations in the genome of other histological subtypes are known but specific agents targeting these alterations have yet to be developed. Thus, the therapeutic management of these subtypes represents an ongoing challenge. Recent advances in immunotherapy have highlighted the potential of immuno-oncology based treatments for NSCLC, offering the potential to provide durable responses and outcomes regardless of histology or mutation status. This review discusses the current unmet medical needs in NSCLC, the limits of current first-line and later-line chemotherapy and targeted agents, and the emergence of new therapeutic strategies.
Collapse
Affiliation(s)
- G V Scagliotti
- University of Torino, Department of Oncology, Orbassano, Torino, Italy.
| | - P Bironzo
- University of Torino, Department of Oncology, Orbassano, Torino, Italy
| | - J F Vansteenkiste
- Respiratory Oncology Unit and Trial Unit, Department of Pulmonology, University Hospitals KU Leuven, Leuven, Belgium
| |
Collapse
|
30
|
Villaruz LC, Kalyan A, Zarour H, Socinski MA. Immunotherapy in lung cancer. Transl Lung Cancer Res 2015; 3:2-14. [PMID: 25806276 DOI: 10.3978/j.issn.2218-6751.2013.10.13] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 10/17/2013] [Indexed: 12/16/2022]
Abstract
Immunotherapy has emerged in recent years as a promising therapeutic approach in lung cancer. Two approaches are of particular interest: immune checkpoint inhibition, which aims to counteract the physiologic mechanisms of immune tolerance co-opted by some tumors, and vaccine therapy, which enables enhanced exposure to tumor antigen. Immune checkpoint therapies include the monoclonal antibody blockade of the cytotoxic T-lymphocyte antigen-4 (CTLA-4) with ipilimumab, as well as antibody blockade of the programmed cell death-1 (PD-1) receptor and the PD-1 ligand. These immune checkpoint therapies have been evaluated in both non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) with early evidence of activity. Vaccines include antigen specific therapies which induce specific antitumor immunity against relevant tumor-associated antigens. In lung cancer, these include the melanoma-associated antigen-A3 (MAGE-A3), membrane-associated glycoprotein (MUC-1), and the epidermal growth factor receptor (EGFR). Whole tumor vaccines have also been evaluated in lung cancer and influence the patient's immune system to allow recognition of the tumor as foreign creating de novo immunity. This review summarizes the evidence to date for the efficacy and safety of immunotherapies in lung cancer.
Collapse
Affiliation(s)
- Liza C Villaruz
- University of Pittsburgh Cancer Institute, University of Pittsburgh, School of Medicine/Hematology-Oncology, Lung and Thoracic Malignancies Program, Pittsburgh, PA 15232, USA
| | - Aparna Kalyan
- University of Pittsburgh Cancer Institute, University of Pittsburgh, School of Medicine/Hematology-Oncology, Lung and Thoracic Malignancies Program, Pittsburgh, PA 15232, USA
| | - Hassane Zarour
- University of Pittsburgh Cancer Institute, University of Pittsburgh, School of Medicine/Hematology-Oncology, Lung and Thoracic Malignancies Program, Pittsburgh, PA 15232, USA
| | - Mark A Socinski
- University of Pittsburgh Cancer Institute, University of Pittsburgh, School of Medicine/Hematology-Oncology, Lung and Thoracic Malignancies Program, Pittsburgh, PA 15232, USA
| |
Collapse
|
31
|
Swaika A, Hammond WA, Joseph RW. Current state of anti-PD-L1 and anti-PD-1 agents in cancer therapy. Mol Immunol 2015; 67:4-17. [PMID: 25749122 DOI: 10.1016/j.molimm.2015.02.009] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/05/2015] [Accepted: 02/08/2015] [Indexed: 02/07/2023]
Abstract
Immunotherapy for the treatment of cancer is rapidly evolving from therapies that globally and non-specifically simulate the immune system to more targeted activation of individual components of the immune system. The net result of this targeted approach is decreased toxicity and increased efficacy of immunotherapy. More specifically, therapies that inhibit the interaction between programmed death ligand 1 (PD-L1), present on the surface of tumor or antigen-presenting cells, and programmed death 1 (PD-1), present on the surface of activated lymphocytes, are generating much excitement and enthusiasm, even in malignancies that are not traditionally considered to be immunogenic. Herein, we review the current landscape of anti-PD-1 and anti-PD-L1 therapies in the world of oncology. We have performed a comprehensive literature search on the data available through PubMed, Medline, Scopus, the ClinicalTrials.gov registry, and abstracts from major oncology meetings in order to summarize the clinical data of anti-PD-1/PD-L1 therapies.
Collapse
Affiliation(s)
- Abhisek Swaika
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, USA
| | - William A Hammond
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, USA
| | - Richard W Joseph
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, USA.
| |
Collapse
|
32
|
Abstract
Despite the availability of radiotherapy, cytotoxic agents, and targeted agents, a high unmet medical need remains for novel therapies that improve treatment outcomes in patients with lung cancer who are ineligible for surgical resection. Building upon the early promise shown with general immunostimulatory agents, immuno-oncology is at the forefront of research in this field, with several novel agents currently under investigation. In particular, agents targeting immune checkpoints, such as the cytotoxic T-lymphocyte antigen-4 (CTLA-4) receptor and programmed death-1 (PD-1) receptor, have shown in early clinical trials potential for improving tumor responses and survival in patients with non-small cell lung cancer (NSCLC). Here, we examine the rationale for targeting immune checkpoints in lung cancer and review the clinical data from studies with immune checkpoint inhibitors currently in development. The challenges associated with optimizing treatment with these agents in lung cancer also are discussed.
Collapse
Affiliation(s)
- Martin Reck
- Department of Thoracic Oncology, LungenClinic Grosshansdorf, Airway Research Center North (ARCN), Grosshansdorf, Germany.
| | - Luis Paz-Ares
- Department of Medical Oncology, Instituto de Biomedicina de Sevilla - IBIS (Hospital Universitario Virgen del Rocio, Universidad de Sevilla and CSIC), Seville, Spain
| |
Collapse
|
33
|
Han RX, Liu X, Pan P, Jia YJ, Yu JC. Effectiveness and safety of chemotherapy combined with dendritic cells co-cultured with cytokine-induced killer cells in the treatment of advanced non-small-cell lung cancer: a systematic review and meta-analysis. PLoS One 2014; 9:e108958. [PMID: 25268709 PMCID: PMC4182599 DOI: 10.1371/journal.pone.0108958] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 09/06/2014] [Indexed: 12/03/2022] Open
Abstract
Background Lung cancer, particularly non-small-cell lung cancer (NSCLC) is the leading cause of cancer mortality. Chemotherapy combined dendritic cells co-cultured with cytokine-induced killer cells (DC-CIK) immunotherapy has been applied in advanced NSCLC patients' treatment, but couldn't provide consistent beneficial results. Therefore, it is necessary to evaluate the efficiency and safety of combination therapy to promote the application. Methods A literature search for randomized controlled trials of NSCLC was conducted in PubMed database. Before meta-analysis was performed, studies were evaluated heterogeneity. Pooled risk ratios (RRs) were estimated and 95% confidence intervals (CIs) were calculated using a fixed-effect model. Sensitivity analysis was also performed. Results Six eligible trials were enrolled. Efficiency and safety of chemotherapy followed by DC-CIK immunotherapy (experimental group) and chemotherapy alone (control group) were compared. 1-year overall survival (OS) (P = 0.02) and progression free survival (PFS) (P = 0.005) in the experimental group were significantly increased compared with the control. Disease control rate (DCR) (P = 0.006) rose significantly in experimental group. However, no significant differences between the two groups were observed in 2-year OS (P = 0.21), 2-year PFS (P = 0.10), overall response rate (ORR) (P = 0.76) and partial response (PR) (P = 0.22). Temporary fever, anemia, leukopenia and nausea were the four major adverse events (AEs) treated by chemotherapy. The incidence of anemia, leukopenia and nausea in the experimental group was obviously lower than the control group. Temporary fever rate was higher in experimental group than that in the control, but could be alleviated by taking sufficient rest. Conclusions Chemotherapy combined with DC-CIK immunotherapy showed superiority in DCR, 1-year OS and PFS, and no more AEs appeared, however, there was no significant improvement in ORR, PR, 2-year OS and PFS. As a whole, the combination therapy is safer but modest in efficacy for advanced NSCLC patients.
Collapse
Affiliation(s)
- Rui-xian Han
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xu Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pan Pan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ying-jie Jia
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- * E-mail: (JCY); (YJJ)
| | - Jian-chun Yu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- * E-mail: (JCY); (YJJ)
| |
Collapse
|
34
|
Lee JM, Dubinett SM, Sharma S. Immunologic Approaches to Lung Cancer Therapy. Lung Cancer 2014. [DOI: 10.1002/9781118468791.ch29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Massarelli E, Papadimitrakopoulou V, Welsh J, Tang C, Tsao AS. Immunotherapy in lung cancer. Transl Lung Cancer Res 2014; 3:53-63. [PMID: 25806281 PMCID: PMC4367607 DOI: 10.3978/j.issn.2218-6751.2014.01.01] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 01/17/2014] [Indexed: 12/13/2022]
Abstract
Survival rates for metastatic lung cancer including non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) are poor with 5-year survival of less than 5%. The use of molecular targeted therapies has improved median overall survival (OS) in a limited group of NSCLC patients whose tumors harbor specific genetic alterations. However for a large group of NSCLC and SCLC molecular alterations are not available to lead to direct targeted therapies. Recent favorable results of newer trials of therapeutic vaccines and checkpoint inhibitors have proven against the common belief that lung cancer is nonimmunogenic. In particular, the checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and the programmed death-1 (PD-1) pathway have shown durable clinical responses with manageable toxicity. Several phase II and III clinical trials testing the association of different schedule of chemotherapy and immunotherapy or immunotherapy alone are ongoing in lung cancer and important results are expected in the near future. However, more studies are needed to understand the optimal combination of immunotherapeutic agents with chemotherapy and radiation therapy for the treatment of NSCLC and SCLC.
Collapse
|
36
|
Perez CA, Santos ES, Raez LE. Active immunotherapy for non-small-cell lung cancer: moving toward a reality. Expert Rev Anticancer Ther 2014; 11:1599-605. [DOI: 10.1586/era.11.155] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
37
|
Zielinski C, Knapp S, Mascaux C, Hirsch F. Rationale for targeting the immune system through checkpoint molecule blockade in the treatment of non-small-cell lung cancer. Ann Oncol 2013; 24:1170-9. [PMID: 23393121 PMCID: PMC3629900 DOI: 10.1093/annonc/mds647] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/28/2012] [Accepted: 12/03/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Treatments of non-small-cell lung cancer (NSCLC)-particularly of the squamous subtype-are limited. In this article, we describe the immunomodulatory environment in NSCLC and the potential for therapeutic targeting of the immune system through cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death-1 (PD-1) immune-checkpoint pathway blockade. MATERIALS AND METHODS We searched PubMed and presented abstracts for publications describing the clinical benefit of checkpoint blockade in NSCLC. RESULTS Antibody-mediated checkpoint molecule blockade is being investigated in NSCLC, and of these approaches, the anti-CTLA-4 antibody ipilimumab has undergone the most extensive clinical study. By targeting the immune system rather than specific antigens, checkpoint blockade agents differ from vaccine therapy. In a phase II study in advanced NSCLC, phased ipilimumab with chemotherapy demonstrated the greatest efficacy in squamous NSCLC. A phase I study of nivolumab, an anti-PD-1 antibody, has suggested that this agent is also active against squamous and non-squamous NSCLC. Ongoing phase III studies are evaluating the therapeutic potential of these agents. CONCLUSIONS Although treatment options for NSCLC are limited, a better understanding of the immune profile of this disease has facilitated the development of immunotherapeutics that target checkpoint blockade molecules, and clinical evaluation to date supports combining checkpoint blockade with chemotherapy for squamous NSCLC.
Collapse
Affiliation(s)
- C Zielinski
- Central European Cooperative Oncology Group (CECOG), Vienna, Vienna General Hospital, Vienna, Austria.
| | | | | | | |
Collapse
|
38
|
Villaflor VM, Salgia R. Targeted agents in non-small cell lung cancer therapy: What is there on the horizon? J Carcinog 2013; 12:7. [PMID: 23599689 PMCID: PMC3622362 DOI: 10.4103/1477-3163.109253] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 01/10/2013] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is a heterogeneous group of diseases. There has been much research in lung cancer over the past decade which has advanced our ability to treat these patients with a more personalized approach. The scope of this paper is to review the literature and give a broad understanding of the current molecular targets for which we currently have therapies as well as other targets for which we may soon have therapies. Additionally, we will cover some of the issues of resistance with these targeted therapies. The molecular targets we intend to discuss are epidermal growth factor receptor (EGFR), Vascular endothelial growth factor (VEGF), anaplastic large-cell lymphoma kinase (ALK), KRAS, C-MET/RON, PIK3CA. ROS-1, RET Fibroblast growth factor receptor (FGFR). Ephrins and their receptors, BRAF, and immunotherapies/vaccines. This manuscript only summarizes the work which has been done to date and in no way is meant to be comprehensive.
Collapse
Affiliation(s)
- Victoria M Villaflor
- Department of Medicine, Section of Hematology/Oncology University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
39
|
Brahmer JR. Harnessing the immune system for the treatment of non-small-cell lung cancer. J Clin Oncol 2013; 31:1021-8. [PMID: 23401435 DOI: 10.1200/jco.2012.45.8703] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Over the last several years, new therapeutic targets have emerged in immunotherapy, particularly the immune checkpoint pathways. Blocking inhibitory pathways via monoclonal antibodies, such as the anti-cytotoxic T-lymphocyte antigen-4 antibody (ipilimumab), anti-programmed cell death-1 antibody (BMS-936558), and anti-programmed cell death-1 ligand antibody (BMS-936559), has the ability to break down the shield that tumors co-opt for their defense. Vaccines are able to help the immune system develop immune memory that can have long-lasting, tumor-specific effects. Newer vaccines, particularly the tumor cell vaccine, belagenpumatucel-L, and the antigen-specific vaccines, melanoma-associated antigen-A3, liposomal BLP-25, TG4010, and recombinant human epidermal growth factor, are being evaluated in some of the largest trials ever attempted in lung cancer therapy. These therapies alone or in combination may hold the key to making immunotherapy a reality in the treatment of lung cancer.
Collapse
Affiliation(s)
- Julie R Brahmer
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| |
Collapse
|
40
|
[Review on immunotherapies for lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2012; 15:606-11. [PMID: 23075686 PMCID: PMC5999828 DOI: 10.3779/j.issn.1009-3419.2012.10.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Lung cancer is a highly malignant disease with poor prognosis, most cases are diagnosed at a very late stage. More effective medications or therapies should be developed to improve its prognosis. The advancement of tumor immunity and tumor immunosuppression facilitated the feasibility of immunotherapies for lung cancer. Ipilimumab, antibody to Programmed death-1 (PD-1), Toll-like receptor agonists, liposomal BLP25 (L- BLP25), belagenpumatucel-L, melanoma-associated antigen A3 (MAGE-A3) vaccine and talactoferrin have been proved to be effective for lung cancer through early clinical trials, most of the drugs have moved forward to phase III trials, so as to collect much higher level evidence to support the immunotherapies incorporated into the multidisciplinary treatment of lung cancer. The selection of target patients at appropriate stages, breaking down of tumor immunosuppression as well as the objective measurement of tumor response to the therapy are major challenges for the development of immunotherapies for lung cancer. The clarifying of the mechanism of immune escape led to the above drug development, and immune-senescence has already become the hotspot in this field.
Collapse
|
41
|
Brahmer JR, Tykodi SS, Chow LQM, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF, Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366:2455-65. [PMID: 22658128 PMCID: PMC3563263 DOI: 10.1056/nejmoa1200694] [Citation(s) in RCA: 6243] [Impact Index Per Article: 480.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Programmed death 1 (PD-1) protein, a T-cell coinhibitory receptor, and one of its ligands, PD-L1, play a pivotal role in the ability of tumor cells to evade the host's immune system. Blockade of interactions between PD-1 and PD-L1 enhances immune function in vitro and mediates antitumor activity in preclinical models. METHODS In this multicenter phase 1 trial, we administered intravenous anti-PD-L1 antibody (at escalating doses ranging from 0.3 to 10 mg per kilogram of body weight) to patients with selected advanced cancers. Anti-PD-L1 antibody was administered every 14 days in 6-week cycles for up to 16 cycles or until the patient had a complete response or confirmed disease progression. RESULTS As of February 24, 2012, a total of 207 patients--75 with non-small-cell lung cancer, 55 with melanoma, 18 with colorectal cancer, 17 with renal-cell cancer, 17 with ovarian cancer, 14 with pancreatic cancer, 7 with gastric cancer, and 4 with breast cancer--had received anti-PD-L1 antibody. The median duration of therapy was 12 weeks (range, 2 to 111). Grade 3 or 4 toxic effects that investigators considered to be related to treatment occurred in 9% of patients. Among patients with a response that could be evaluated, an objective response (a complete or partial response) was observed in 9 of 52 patients with melanoma, 2 of 17 with renal-cell cancer, 5 of 49 with non-small-cell lung cancer, and 1 of 17 with ovarian cancer. Responses lasted for 1 year or more in 8 of 16 patients with at least 1 year of follow-up. CONCLUSIONS Antibody-mediated blockade of PD-L1 induced durable tumor regression (objective response rate of 6 to 17%) and prolonged stabilization of disease (rates of 12 to 41% at 24 weeks) in patients with advanced cancers, including non-small-cell lung cancer, melanoma, and renal-cell cancer. (Funded by Bristol-Myers Squibb and others; ClinicalTrials.gov number, NCT00729664.).
Collapse
Affiliation(s)
- Julie R Brahmer
- Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Belalcazar A, Raez LE, Santos ES. Immunotherapy for nonsmall-cell lung cancer. MEMO-MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2012. [DOI: 10.1007/s12254-012-0019-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
43
|
Wang J, Zou ZH, Xia HL, He JX, Zhong NS, Tao AL. Strengths and weaknesses of immunotherapy for advanced non-small-cell lung cancer: a meta-analysis of 12 randomized controlled trials. PLoS One 2012; 7:e32695. [PMID: 22403699 PMCID: PMC3293858 DOI: 10.1371/journal.pone.0032695] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 01/30/2012] [Indexed: 12/31/2022] Open
Abstract
Background Lung cancer is one of the leading causes of cancer death worldwide. Non-small-cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancers. Immunotherapy has yielded no consistent benefit to date for those patients. Assessing the objective efficacy and safety of immunotherapy for advanced NSCLC patients will help to instruct the future development of immunotherapeutic drugs. Methodology and Principal Findings We performed a meta-analysis of 12 randomized controlled trials including 3134 patients (1570 patients in the immunotherapy group and 1564 patients in the control group) with histologically confirmed stage IIIA, IIIB, or IV NSCLC. The analysis was executed with efficacy end points regarding overall survival (OS), progression-free survival (PFS), complete response (CR), partial response (PR), and total effective rate. Overall unstratified OS, PFS, PR, and total effective rate were significantly improved in advanced NSCLC patients in the immunotherapy group (P = 0.0007, 0.0004, 0.002, 0.003, respectively), whereas CR was not improved (P = 0.97). Subgroup analysis showed that monoclonal antibody (mAb) immunotherapy significantly improved the PFS, PR, and total effective rate and showed a trend of improving OS of advanced NSCLC patients compared with the control group, with one kind of adverse event being significantly dominant. Compared with the control group, the vaccine subgroup showed no significant difference with regard to serious adverse events, whereas cytokine immunotherapy significantly induced three kinds of serious adverse events. Conclusions Immunotherapy works efficiently on advanced NSCLC patients. Of several immunotherapies, mAb therapy may be a potential immunotherapy for advanced NSCLC patients, and become a standard complementary therapeutic approach in the future if the issues concerning toxicity and allergenicity of mAbs have been overcome.
Collapse
Affiliation(s)
- Juan Wang
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ze-Hong Zou
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hong-Lin Xia
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian-Xing He
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nan-Shan Zhong
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ai-Lin Tao
- Guangzhou Municipal Key Laboratory of Allergy and Clinical Immunology, Allergy Research Branch of the State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- * E-mail:
| |
Collapse
|
44
|
De Pas T, Giovannini M, Rescigno M, Catania C, Toffalorio F, Spitaleri G, Delmonte A, Barberis M, Spaggiari L, Solli P, Veronesi G, De Braud F. Vaccines in non-small cell lung cancer: rationale, combination strategies and update on clinical trials. Crit Rev Oncol Hematol 2012; 83:432-43. [PMID: 22366114 DOI: 10.1016/j.critrevonc.2011.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 12/20/2011] [Accepted: 12/22/2011] [Indexed: 12/20/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains the leading cause of cancer related mortality worldwide and despite some advances in therapy the overall prognosis remains disappointing. New therapeutic approaches like vaccination have been proposed and several clinical trials are ongoing. Many tumor antigens have been identified so far and specific tumor vaccines targeting these antigens have been developed. Even if the ideal setting for vaccine therapy might be the adjuvant one, vaccines seem to be potentially beneficial also in advanced disease and combination therapy could be a promising treatment option. In the advanced setting anti-MUC-1 vaccine (belagenpumatucel) and anti-TGF-β(2) vaccine (BPL-25) have entered in phase III trials as maintenance therapy after first line chemotherapy. In the adjuvant setting the most relevant and promising vaccines are directed against MAGE-A3 and PRAME, respectively. We will review the key points for effective active immunotherapies and combination therapies, giving an update on the most promising vaccines developed in NSCLC.
Collapse
Affiliation(s)
- Tommaso De Pas
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|