51
|
Malhotra J, Jabbour SK. Patient-related outcomes with the use of checkpoint inhibitors for the treatment of metastatic non-small cell lung cancer. Transl Lung Cancer Res 2018; 7:S138-S141. [PMID: 29782572 DOI: 10.21037/tlcr.2018.03.09] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Jyoti Malhotra
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Salma K Jabbour
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
52
|
Okuma Y, Wakui H, Utsumi H, Sagawa Y, Hosomi Y, Kuwano K, Homma S. Soluble Programmed Cell Death Ligand 1 as a Novel Biomarker for Nivolumab Therapy for Non-Small-cell Lung Cancer. Clin Lung Cancer 2018; 19:410-417.e1. [PMID: 29859759 DOI: 10.1016/j.cllc.2018.04.014] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/06/2018] [Accepted: 04/24/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Biomarkers for predicting the effect of anti-programmed cell death 1 (PD-1) monoclonal antibody against non-small-cell lung cancer (NSCLC) are urgently required. Although it is known that the blood levels of soluble programmed cell death ligand 1 (sPD-L1) are elevated in various malignancies, the nature of sPD-L1 has not been thoroughly elucidated. We investigated the significance of plasma sPD-L1 levels as a biomarker for anti-PD-1 monoclonal antibody, nivolumab therapy. PATIENTS AND METHODS The present prospective study included 39 NSCLC patients. The patients were treated with nivolumab at the dose of 3 mg/kg every 2 weeks, and the effects of nivolumab on NSCLC were assessed according to the change in tumor size, time to treatment failure (TTF), and overall survival (OS). The baseline plasma sPD-L1 concentration was determined using an enzyme-linked immunosorbent assay. RESULTS The area under the curve of the receiver operating characteristic curve was 0.761. The calculated optimal cutoff point for sPD-L1 in the plasma samples was 3.357 ng/mL. Of the 39 patients, 59% with low plasma sPD-L1 levels achieved a complete response or partial response and 25% of those with high plasma sPD-L1 levels did so. In addition, 22% of the patients with low plasma sPD-L1 levels developed progressive disease compared with 75% of those with high plasma sPD-L1 levels. The TTF and OS were significantly longer for those patients with low plasma sPD-L1 levels compared with the TTF and OS for those with high plasma sPD-L1 levels. CONCLUSION The clinical benefit from nivolumab therapy was significantly associated with the baseline plasma sPD-L1 levels. Plasma sPD-L1 levels might represent a novel biomarker for the prediction of the efficacy of nivolumab therapy against NSCLC.
Collapse
Affiliation(s)
- Yusuke Okuma
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan; Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Hiroshi Wakui
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Hirofumi Utsumi
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Yukiko Sagawa
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Kazuyoshi Kuwano
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Sadamu Homma
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
53
|
Clinical efficacy of percutaneous cryoablation combined with allogenic NK cell immunotherapy for advanced non-small cell lung cancer. Immunol Res 2018; 65:880-887. [PMID: 28508945 DOI: 10.1007/s12026-017-8927-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this study, the safety and clinical efficacy of cryosurgery combined with allogenic NK cell immunotherapy for the treatment of advanced non-small cell lung cancer (NSCLC) were evaluated. From July 2016 to March 2017, we enrolled 60 patients who met the enrollment criteria and divided them into two groups: (1) the simple cryoablation group (n = 30) and (2) the cryoablation combined with allogenic NK cell group (n = 30). The changes in immune function, quality of life, and clinical response were evaluated. We found that allogenic NK cells combined with cryosurgical treatment for advanced NSCLC have a synergistic effect, which not only enhancing the immune function of patients, improving the quality of life, and significantly increasing the response rate (RR) and disease control rate (DCR) compared to cryoablation group. This study is the first clinical trial of allogenic NK cells combined with cryosurgery for the treatment of advanced NSCLC and preliminaily its safety and efficacy.
Collapse
|
54
|
Tian X, Ma J, Wang T, Tian J, Zhang Y, Mao L, Xu H, Wang S. Long Non-Coding RNA HOXA Transcript Antisense RNA Myeloid-Specific 1-HOXA1 Axis Downregulates the Immunosuppressive Activity of Myeloid-Derived Suppressor Cells in Lung Cancer. Front Immunol 2018; 9:473. [PMID: 29662483 PMCID: PMC5890118 DOI: 10.3389/fimmu.2018.00473] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/22/2018] [Indexed: 02/06/2023] Open
Abstract
HOXA transcript antisense RNA myeloid-specific 1 (HOTAIRM1) is a long non-coding RNA that has been shown to be a key regulator of myeloid cell development by targeting HOXA1. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that possess immunosuppressive function. However, the impact of HOTAIRM1 on the development of MDSCs remains unknown. In this study, we demonstrated that HOTAIRM1 was expressed in MDSCs and that overexpression of HOTAIRM1 could downregulate the expression of suppressive molecules in MDSCs. In addition, HOTAIRM1 levels were observed to be decreased in the peripheral blood cells of lung cancer patients compared with those of healthy controls. By analyzing HOTAIRM1 expression levels in different types of lung cancer, we found that HOTAIRM1 was mainly expressed in lung adenocarcinoma. Finally, it was confirmed that HOTAIRM1 could enhance the expression of HOXA1 in MDSCs and that high levels of HOXA1, the target gene of HOTAIRM1, could delay tumor progression and enhance the antitumor immune response by downregulating the immunosuppression of MDSCs. Taken together, this study illustrates that HOTAIRM1/HOXA1 downregulates the immunosuppressive function of MDSCs and may be a potential therapeutic target in lung cancer.
Collapse
Affiliation(s)
- Xinyu Tian
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jie Ma
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ting Wang
- Department of Laboratory Medicine, Jiangsu Cancer Hospital, Nanjing, China
| | - Jie Tian
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yue Zhang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Lingxiang Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Huaxi Xu
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
55
|
Shariatpanahi SP, Shariatpanahi SP, Madjidzadeh K, Hassan M, Abedi-Valugerdi M. Mathematical modeling of tumor-induced immunosuppression by myeloid-derived suppressor cells: Implications for therapeutic targeting strategies. J Theor Biol 2018; 442:1-10. [PMID: 29337259 DOI: 10.1016/j.jtbi.2018.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 01/04/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) belong to immature myeloid cells that are generated and accumulated during the tumor development. MDSCs strongly suppress the anti-tumor immunity and provide conditions for tumor progression and metastasis. In this study, we present a mathematical model based on ordinary differential equations (ODE) to describe tumor-induced immunosuppression caused by MDSCs. The model consists of four equations and incorporates tumor cells, cytotoxic T cells (CTLs), natural killer (NK) cells and MDSCs. We also provide simulation models that evaluate or predict the effects of anti-MDSC drugs (e.g., l-arginine and 5-Fluorouracil (5-FU)) on the tumor growth and the restoration of anti-tumor immunity. The simulated results obtained using our model were in good agreement with the corresponding experimental findings on the expansion of splenic MDSCs, immunosuppressive effects of these cells at the tumor site and effectiveness of l-arginine and 5-FU on the re-establishment of antitumor immunity. Regarding this latter issue, our predictive simulation results demonstrated that intermittent therapy with low-dose 5-FU alone could eradicate the tumors irrespective of their origins and types. Furthermore, at the time of tumor eradication, the number of CTLs prevailed over that of cancer cells and the number of splenic MDSCs returned to the normal levels. Finally, our predictive simulation results also showed that the addition of l-arginine supplementation to the intermittent 5-FU therapy reduced the time of the tumor eradication and the number of iterations for 5-FU treatment. Thus, the present mathematical model provides important implications for designing new therapeutic strategies that aim to restore antitumor immunity by targeting MDSCs.
Collapse
Affiliation(s)
- Seyed Peyman Shariatpanahi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran; Breast Cancer Research Center, ACECR, Tehran, Iran.
| | | | | | - Moustapha Hassan
- Experimental Cancer Medicine, Clinical Research Center, Novum, Karolinska Institutet, Huddinge, 141 86 Stockholm, Sweden; Clinical Research Center, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden.
| | - Manuchehr Abedi-Valugerdi
- Experimental Cancer Medicine, Clinical Research Center, Novum, Karolinska Institutet, Huddinge, 141 86 Stockholm, Sweden.
| |
Collapse
|
56
|
Immunotherapy for cervical cancer: Can it do another lung cancer? Curr Probl Cancer 2018; 42:148-160. [PMID: 29500076 DOI: 10.1016/j.currproblcancer.2017.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 12/10/2017] [Indexed: 02/04/2023]
Abstract
Cervical cancer, although preventable, is still the second most common cancer among women worldwide. In developing countries like India, where screening for cervical cancer is virtually absent, most women seek treatment only at advanced stages of the disease. Although standard treatment is curative in more than 90% of women during the early stages, for stage IIIb and above this rate drops to 50% or less. Hence, novel therapeutic adjuvants are required to improve survival at advanced stages. Lung cancer has shown the way forward with the use of Immunotherapeutic interventions as standard line of treatment in advanced stages. In this review, we provide an overview of mechanisms of immune evasion, strategies that can be employed to boost the immune system in order to improve the overall survival of the patients and summarize briefly the clinical trials that have been completed or that are underway to bring therapeutic vaccines for cervical cancer to the clinics.
Collapse
|
57
|
Liu X, Ran R, Shao B, Rugo HS, Yang Y, Hu Z, Wei Z, Wan F, Kong W, Song G, Jiang H, Liang X, Zhang R, Yan Y, Xu G, Li H. Combined peripheral natural killer cell and circulating tumor cell enumeration enhance prognostic efficiency in patients with metastatic triple-negative breast cancer. Chin J Cancer Res 2018; 30:315-326. [PMID: 30046226 DOI: 10.21147/j.issn.1000-9604.2018.03.04] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Objective Triple-negative breast cancer (TNBC) is a heterogeneous disease with poor prognosis. Circulating tumor cells (CTCs) are a promising predictor for breast cancer prognoses but their reliability regarding progression-free survival (PFS) is controversial. We aim to verify their predictive value in TNBC. Methods In present prospective cohort study, we used the Pep@MNPs method to enumerate CTCs in baseline blood samples from 75 patients with TNBC (taken at inclusion in this study) and analyzed correlations between CTC numbers and outcomes and other clinical parameters. Results Median PFS was 6.0 (range: 1.0-25.0) months for the entire cohort, in whom we found no correlations between baseline CTC status and initial tumor stage (P=0.167), tumor grade (P=0.783) or histological type (P=0.084). However, among those getting first-line treatment, baseline CTC status was positively correlated with ratio of peripheral natural killer (NK) cells (P=0.032), presence of lung metastasis (P=0.034) and number of visceral metastatic site (P=0.037). Baseline CTC status was predictive for PFS in first-line TNBC (P=0.033), but not for the cohort as a whole (P=0.118). This prognostic limitation of CTC could be ameliorated by combining CTC and NK cell enumeration (P=0.049). Conclusions Baseline CTC status was predictive of lung metastasis, peripheral NK cell ratio and PFS in TNBC patients undergoing first-line treatment. We have developed a combined CTC-NK enumeration strategy that allows us to predict PFS in TNBC without any preconditions.
Collapse
Affiliation(s)
- Xiaoran Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ran Ran
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Bin Shao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hope S Rugo
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco 94115, USA
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, the National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Zhiyuan Hu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, the National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Zewen Wei
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, the National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Fengling Wan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Weiyao Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Guohong Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hanfang Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xu Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ruyan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ying Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Guobing Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Clinical Laboratory, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Huiping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
58
|
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
|
59
|
Aerts JGJV, de Goeje PL, Cornelissen R, Kaijen-Lambers MEH, Bezemer K, van der Leest CH, Mahaweni NM, Kunert A, Eskens FALM, Waasdorp C, Braakman E, van der Holt B, Vulto AG, Hendriks RW, Hegmans JPJJ, Hoogsteden HC. Autologous Dendritic Cells Pulsed with Allogeneic Tumor Cell Lysate in Mesothelioma: From Mouse to Human. Clin Cancer Res 2017; 24:766-776. [PMID: 29233904 DOI: 10.1158/1078-0432.ccr-17-2522] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/01/2017] [Accepted: 12/04/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Mesothelioma has been regarded as a nonimmunogenic tumor, which is also shown by the low response rates to treatments targeting the PD-1/PD-L1 axis. Previously, we demonstrated that autologous tumor lysate-pulsed dendritic cell (DC) immunotherapy increased T-cell response toward malignant mesothelioma. However, the use of autologous tumor material hampers implementation in large clinical trials, which might be overcome by using allogeneic tumor cell lines as tumor antigen source. The purpose of this study was to investigate whether allogeneic lysate-pulsed DC immunotherapy is effective in mice and safe in humans.Experimental Design: First, in two murine mesothelioma models, mice were treated with autologous DCs pulsed with either autologous or allogeneic tumor lysate or injected with PBS (negative control). Survival and tumor-directed T-cell responses of these mice were monitored. Results were taken forward in a first-in-human clinical trial, in which 9 patients were treated with 10, 25, or 50 million DCs per vaccination. DC vaccination consisted of autologous monocyte-derived DCs pulsed with tumor lysate from five mesothelioma cell lines.Results: In mice, allogeneic lysate-pulsed DC immunotherapy induced tumor-specific T cells and led to an increased survival, to a similar extent as DC immunotherapy with autologous tumor lysate. In the first-in-human clinical trial, no dose-limiting toxicities were established and radiographic responses were observed. Median PFS was 8.8 months [95% confidence interval (CI), 4.1-20.3] and median OS not reached (median follow-up = 22.8 months).Conclusions: DC immunotherapy with allogeneic tumor lysate is effective in mice and safe and feasible in humans. Clin Cancer Res; 24(4); 766-76. ©2017 AACR.
Collapse
Affiliation(s)
- Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Pauline L de Goeje
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Robin Cornelissen
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Koen Bezemer
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Cor H van der Leest
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Niken M Mahaweni
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - André Kunert
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.,Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ferry A L M Eskens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Cynthia Waasdorp
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Eric Braakman
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Bronno van der Holt
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Arnold G Vulto
- Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Joost P J J Hegmans
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Henk C Hoogsteden
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
60
|
CD14+ HLA-DR-/low MDSCs are elevated in the periphery of early-stage breast cancer patients and suppress autologous T cell proliferation. Breast Cancer Res Treat 2017; 168:401-411. [PMID: 29230664 DOI: 10.1007/s10549-017-4594-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/22/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE Despite the recent expansion in the use of immunotherapy for many cancer types, it is still not a standard treatment for breast cancer. Identifying differences in the immune systems of breast cancer patients compared to healthy women might provide insight into potential targets for immunotherapy and thus may assist its clinical implementation. METHODS Multi-colour flow cytometry was used to investigate myeloid and lymphoid populations in the peripheral blood of breast cancer patients (n = 40) and in the blood of healthy age-matched women (n = 25). We additionally performed functional testing to identify immune suppressive mechanisms used by circulating CD14+ myeloid cells from breast cancer patients. RESULTS Our results show that breast cancer patients have significantly elevated frequencies of cells with the monocytic myeloid-derived suppressor cell (mMDSC) phenotype CD14+ HLA-DR-/low compared with healthy women (p < 0.01). We also observed higher levels of earlier differentiated T cells and correspondingly lower levels of T cells in later stages of differentiation (p < 0.05). These disease-associated differences could already be detected in early-stage breast cancer patients in stages 1 and 2 (n = 33 of 40) (p < 0.05). Levels of circulating T cells correlated with certain clinical features and with patient age (p < 0.05). Functional tests showed that CD14+ myeloid cells from breast cancer patients more potently suppressed autologous T cell proliferation than CD14+ cells from healthy women (p < 0.01). Subsequent investigation determined that suppression was mediated in part by reactive oxygen species, because inhibiting this pathway partially restored T cell proliferation (p < 0.01). CONCLUSION Our results highlight the potential importance of cells with mMDSC phenotypes in breast cancer, identifiable already at early stages of disease. This may provide a basis for identifying possible new therapeutic targets to enhance anti-cancer immunity.
Collapse
|
61
|
Lin M, Liang S, Wang X, Liang Y, Zhang M, Chen J, Niu L, Xu K. Percutaneous irreversible electroporation combined with allogeneic natural killer cell immunotherapy for patients with unresectable (stage III/IV) pancreatic cancer: a promising treatment. J Cancer Res Clin Oncol 2017; 143:2607-2618. [PMID: 28871458 DOI: 10.1007/s00432-017-2513-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE This study was attempted to investigate the safety and clinical efficacy of percutaneous irreversible electroporation combined with allogeneic natural killer cell therapy for treating stage III/IV pancreatic cancer, evaluate median progression-free survival (PFS), and overall survival (OS). METHODS Between March 2016 and February 2017, we enrolled 67 patients who met the enrollment criteria. According to the latest NCCN Guidelines, the patients were divided into stage III (35 patients, 16 patients received only irreversible electroporation (IRE) and 19 patients received IRE-NK: 8 patients underwent one course NK and 11 patients underwent ≥3 courses) and stage IV (32 patients, 14 patients received only IRE and 18 patients received IRE-NK: 8 patients underwent one course NK and 10 patients underwent ≥3 courses). The safety and short-term effects were evaluated first, then the median PFS, median OS, response rate (RR) and disease control rate (DCR) were assessed. RESULTS Adverse events of all patients were limited to grades A and B, included local (mainly cough 12.7%, nausea and emesis 6.8%, pain of puncture point 25.3% and duodenum and gastric retention 5.9%) and systemic (mainly fatigue 21.5, fever 33.5%, and blood pressure intraoperative transient reduction 27.4% and white cell count reduction 22.6%) reactions, fever was most frequent. The serum amylase level at 24 h and 7 d after IRE was not significantly changed compared to those before IRE (P > 0.05). CA19-9 value was lower in IRE-NK group than in IRE at 1 month after treatment (P < 0.05). After a median follow-up of 7.9 months (3.8-12.1 months): in stage III group, median PFS was higher in IRE-NK group (9.1 months) than in IRE group (7.9 months, P = 0.0432), median OS was higher in IRE-NK (13.6 months) than in IRE (12.2 months; P = 0.0327), and median PFS was higher in who received multiple NK than single NK (9.9 vs. 8.2 months; P = 0.0387, respectively), median OS who received multiple NK was higher than single NK (13.7 vs. 12.1 months; P = 0.0451, respectively), the RR in IRE-NK (63.2%) was higher than in IRE (50.0%; P < 0.05); in stage IV group, median OS was higher in IRE-NK (10.2 months) than in IRE (9.1 months; P = 0.0367), the DCR in IRE-NK (66.7%) was higher than in IRE (42.9%; P < 0.05). CONCLUSION Percutaneous irreversible electroporation combined with allogeneic natural killer cell immunotherapy significantly increased median PFS and median OS in stage III pancreatic cancer and extended the median OS of stage IV pancreatic cancer. Multiple allogeneic natural killer cells infusion was associated with better prognosis to stage III pancreatic cancer.
Collapse
Affiliation(s)
- Mao Lin
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School, Guangzhou, China
- Fuda Cancer Institute, Guangzhou, China
| | - Shuzhen Liang
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School, Guangzhou, China
| | - Xiaohua Wang
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School, Guangzhou, China
| | - Yinqing Liang
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School, Guangzhou, China
| | | | - Jibing Chen
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School, Guangzhou, China.
- Fuda Cancer Institute, Guangzhou, China.
| | - Lizhi Niu
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School, Guangzhou, China.
- Fuda Cancer Institute, Guangzhou, China.
- Department of Oncology, Fuda Cancer Hospital, Jinan University School, Guangzhou, China.
| | - Kecheng Xu
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School, Guangzhou, China
- Fuda Cancer Institute, Guangzhou, China
| |
Collapse
|
62
|
Domagala-Kulawik J, Raniszewska A. How to evaluate the immune status of lung cancer patients before immunotherapy. Breathe (Sheff) 2017; 13:291-296. [PMID: 29225707 PMCID: PMC5715360 DOI: 10.1183/20734735.001917] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Nowadays, cancer immunotherapy is a promising strategy in solid tumour treatment. It has become a breakthrough in achieving long-term survival in many advanced cases. The essence of modern immunotherapy is to improve the host antitumour immune defence. Currently, it is critically important to determine the biomarkers that could be helpful in planning this type of individual therapy. It has turned out that an important prognostic factor is the evaluation of inflammatory infiltration of the tumour mass, including the characteristics of populations of lymphocytes and macrophages, and the expression of suppressive and regulatory molecules. For lung cancer, <30% of the tumours are resectable and available for a complete microscopic examination. In other cases, the material for the study of inflammatory infiltration may be a tumour biopsy, but this is of limited importance. A valuable way to evaluate the microenvironment of tumour growth is a bronchoalveolar lavage (BAL) fluid examination. In the BAL fluid, the cellular and noncellular components determine the specific type of inflammatory response in an environment of developing cancer. BAL fluid analysis may be a valuable addition to peripheral blood analysis during qualification for modern immunomodulatory therapy. Moreover, it is important material to seek biomarkers of clinical significance. Bronchoalveolar lavage may be used to evaluate the immune status of lung cancer patients before immunotherapyhttp://ow.ly/lXRi30dzmGY
Collapse
Affiliation(s)
- Joanna Domagala-Kulawik
- Dept of Internal Medicine, Pulmonology and Allergology, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
63
|
Hu Z. The future of immune checkpoint blockade immunotherapy: towards personalized therapy or towards combination therapy. J Thorac Dis 2017; 9:4226-4229. [PMID: 29268478 DOI: 10.21037/jtd.2017.10.31] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Zhiwei Hu
- Department of Surgery Division of Surgical Oncology, The Ohio State University College of Medicine, James Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
64
|
The Significance of the PD-L1 Expression in Non-Small-Cell Lung Cancer: Trenchant Double Swords as Predictive and Prognostic Markers. Clin Lung Cancer 2017; 19:120-129. [PMID: 29153898 DOI: 10.1016/j.cllc.2017.10.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 10/12/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022]
Abstract
Lung cancer is the leading cause of death due to cancer worldwide. Surgery, chemotherapy, and radiotherapy have been the standard treatment for lung cancer, and targeted molecular therapy has greatly improved the clinical course of patients with non-small-cell lung cancer (NSCLC) harboring driver mutations, such as in epidermal growth factor receptor and anaplastic lymphoma kinase genes. Despite advances in such therapies, the prognosis of patients with NSCLC without driver oncogene mutations remains poor. Immunotherapy targeting programmed cell death-1 (PD-1) and programmed cell death-ligand 1 (PD-L1) has recently been shown to improve the survival in advanced NSCLC. The PD-L1 expression on the surface of tumor cells has emerged as a potential biomarker for predicting responses to immunotherapy and prognosis after surgery in NSCLC. However, the utility of PD-L1 expression as a predictive and prognostic biomarker remains controversial because of the existence of various PD-L1 antibodies, scoring systems, and positivity cutoffs. In this review, we summarize the data from representative clinical trials of PD-1/PD-L1 immune checkpoint inhibitors in NSCLC and previous reports on the association between PD-L1 expression and clinical outcomes in patients with NSCLC. Furthermore, we discuss the future perspectives of immunotherapy and immune checkpoint factors.
Collapse
|
65
|
Arrieta O, Montes-Servín E, Hernandez-Martinez JM, Cardona AF, Casas-Ruiz E, Crispín JC, Motola D, Flores-Estrada D, Barrera L. Expression of PD-1/PD-L1 and PD-L2 in peripheral T-cells from non-small cell lung cancer patients. Oncotarget 2017; 8:101994-102005. [PMID: 29254220 PMCID: PMC5731930 DOI: 10.18632/oncotarget.22025] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 09/03/2017] [Indexed: 12/11/2022] Open
Abstract
Binding of programmed death-1 (PD-1) with its ligands (PD-L1/2) transmits a co-inhibitory signal in activated T-cells that promotes T-cell exhaustion, leading to tumor immune evasion. The efficacy of antibodies targeting PD-1 and PD-L1 has led to a paradigm shift in lung cancer treatment but the prognostic and predictive value of tumor PD-L1 expression remains controversial. Evaluating PD-1, PD-L1/2 expression in peripheral blood cells may serve as a potential biomarker for prognosis and response to therapy. In this prospective observational study, plasma cytokine levels and PD-1, PD-L1 and PD-L2 expression was evaluated in circulating CD3+, CD3+CD4+ and CD3+CD8+ cells from 70 treatment-naïve patients with advanced NSCLC (Stage IIIB and IV) and from 10 healthy donors. The primary objective was to assess OS according to PD-1, PD-L1, PD-L2 expression status on PBMCs and lymphocyte subsets. Our results indicate that the percentage of PD-L1+CD3+, PD-L1+CD3+CD8+ PD-L2+PBMCs, PD-L2+CD3+, PD-L2+CD3+CD4+ cells was higher in patients than in healthy donors. Survival was decreased among patients with a high percentage of either PD-1+PBMCs, PD-1+CD3+, PD-L1+CD3+, PD-L1+CD3+CD8+, PD-L2+CD3+, PD-L2+CD3+CD4+, or PD-L2+CD3+CD8+ cells. IL-2 and TNF-α showed the strongest association with PD-L1 and PD-L2 expression on specific subsets of T-lymphocytes. Our findings suggest that increased PD-1/PD-L1/PDL-2 expression in PBMCs, particularly in T-cells, may be an additional mechanism leading to tumor escape from immune control. This study is registered with ClinicalTrials.gov, number NCT02758314.
Collapse
Affiliation(s)
- Oscar Arrieta
- Functional Unit of Thoracic Oncology and Laboratory of Personalized Medicine, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Edgar Montes-Servín
- Functional Unit of Thoracic Oncology and Laboratory of Personalized Medicine, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Juan-Manuel Hernandez-Martinez
- Functional Unit of Thoracic Oncology and Laboratory of Personalized Medicine, Instituto Nacional de Cancerología, Mexico City, Mexico.,CONACyT-Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Andrés F Cardona
- Clinical and Translational Oncology Group, Institute of Oncology, Clínica del Country, Bogotá, Colombia
| | - Eibar Casas-Ruiz
- Functional Unit of Thoracic Oncology and Laboratory of Personalized Medicine, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - José C Crispín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Daniel Motola
- Centro Oncológico, Hospital Médica Sur, Mexico City, Mexico
| | - Diana Flores-Estrada
- Functional Unit of Thoracic Oncology and Laboratory of Personalized Medicine, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Lourdes Barrera
- Functional Unit of Thoracic Oncology and Laboratory of Personalized Medicine, Instituto Nacional de Cancerología, Mexico City, Mexico
| |
Collapse
|
66
|
Lin M, Alnaggar M, Liang S, Wang X, Liang Y, Zhang M, Chen J, Niu L, Xu K. An important discovery on combination of irreversible electroporation and allogeneic natural killer cell immunotherapy for unresectable pancreatic cancer. Oncotarget 2017; 8:101795-101807. [PMID: 29254205 PMCID: PMC5731915 DOI: 10.18632/oncotarget.21974] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/23/2017] [Indexed: 12/11/2022] Open
Abstract
Purpose To study the safety and clinical efficacy on combination of irreversible electroporation and allogeneic natural killer cell therapy for treating Stage III/IV pancreatic cancer, evaluating median progression free survival (PFS), and overall survival (OS). Results Adverse events of all patients were limited to grades 1 and 2, including local (mainly tussis 13.4%, nausea and emesis 7.1%, pain of puncture point 29.6% and duodenum and gastric retention 4.3%) and systemic (mainly fatigue 22.3%, fever 31.6%, and transient reduction of intraoperative blood pressure 25.1% and white cell count reduction 18.3%) reactions, fever was the most frequent. The serum amylase level at 24 h and 7 d after IRE was not significantly changed compared to those before IRE (P > 0.05). CA19–9 value was lower in IRE-NK group than in IRE at 1 month after treatment (P < 0.05). After a median follow-up of 7.4 months (3.6–11.2 months): in stage III group, median PFS was higher in IRE-NK group (9.3 months) than in IRE group (8.1 months, P = 0.0465), median OS was higher in IRE-NK (13.2 months) than in IRE (11.4 months, P = 0.0411), and median PFS was higher in who received multiple NK than single NK (9.8 months vs.8.1 months, P = 0.0423, respectively), median OS who received multiple NK was higher than single NK (13.9 months vs.12.3 months, P = 0.0524, respectively), the RR in IRE-NK (63.2%) was higher than in IRE (50.0%, P < 0.05); in stage IV group, median OS was higher in IRE-NK (9.8 months) than in IRE (8.7 months, P = 0.0397), the DCR in IRE-NK (66.7%) was higher than in IRE (42.9%, P < 0.05). Materials and Methods Between July 2016 and May 2017, we enrolled 71 patients who met the enrollment criteria. The patients were divided into stage III (32 patients, 17 patients received only IRE and 15 patients received IRE-NK (Irreversible electroporation- natural killer): 8 patients underwent a course of NK and 7 patients underwent ≥ 3 courses) and stage IV (39 patients, 22 patients received only IRE and 17 patients received IRE-NK: 9 patients underwent a course of NK and 8 patients underwent ≥ 3 courses). The safety and short-term effects were evaluated firstly, then the median PFS, median OS, response rate (RR) and disease control rate (DCR) were assessed. Conclusions Combination of irreversible electroporation and allogeneic natural killer cell immunotherapy significantly increased median PFS and median OS in stage III pancreatic cancer and extended the median OS of stage IV pancreatic cancer. Multiple allogeneic natural killer cells infusion was associated with better prognosis to stage III pancreatic cancer.
Collapse
Affiliation(s)
- Mao Lin
- Fuda Cancer Hospital, School of Medicine, Jinan University, Guangzhou, China.,Fuda Cancer Institute, Guangzhou, China
| | - Mohammed Alnaggar
- Fuda Cancer Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Shuzhen Liang
- Fuda Cancer Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Xiaohua Wang
- Fuda Cancer Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Yinqing Liang
- Fuda Cancer Hospital, School of Medicine, Jinan University, Guangzhou, China
| | | | - Jibing Chen
- Fuda Cancer Hospital, School of Medicine, Jinan University, Guangzhou, China.,Fuda Cancer Institute, Guangzhou, China
| | - Lizhi Niu
- Fuda Cancer Hospital, School of Medicine, Jinan University, Guangzhou, China.,Fuda Cancer Institute, Guangzhou, China
| | - Kecheng Xu
- Fuda Cancer Hospital, School of Medicine, Jinan University, Guangzhou, China.,Fuda Cancer Institute, Guangzhou, China
| |
Collapse
|
67
|
THE RELATIONSHIP BETWEEN NEUTROPHİL / LYMPHOCYTES RATIO AND PLATELET / LYMPHOCYTES RATIO WITH PROGNOSIS IN OPERATED STAGE 1-2 OF NON- SMALL CELL LUNG CANCER DISEASE: ONE CENTRAL EXPERIENCE. ANADOLU KLINIĞI TIP BILIMLERI DERGISI 2017. [DOI: 10.21673/anadoluklin.315155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
68
|
Circulating tumor cell as a biomarker for evaluating allogenic NK cell immunotherapy on stage IV non-small cell lung cancer. Immunol Lett 2017; 191:10-15. [PMID: 28916277 DOI: 10.1016/j.imlet.2017.09.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/28/2017] [Accepted: 09/11/2017] [Indexed: 01/08/2023]
Abstract
In this study, we determined the number of peripheral blood circulating tumor cells (CTCs) pre- and post-NK in patients with stage IV non- small cell lung cancer (NSCLC) as a reference for understanding the relevance of any changes to the efficacy of NK cells therapy. The patients were given one to three courses of immunotherapy. CTC numbers and CTC-related gene expression were measured in the peripheral blood of 31 patients with stage IV NSCLC at 1day before and 7 and 30d after NK cells therapy using magnetic activated cell sorting (MACS) and fluorescence activated cell sorting (FACS) combined with real-time quantitative PCR (RT-qPCR). Throughout the research, fever was the most common reaction (34.6%). The number of CTCs was 18.11±5.813, 15.13±5.984 and 10.32±5.623, respectively, and this decreased significantly over time. ΔCt values for the CTC-related genes CEA, MAGE-3 and CK18 increased significantly after NK cells infusion. The expression of CEA, CK18 and MAGE-3 decreased significantly with time after NK. CTC was a useful biomarker for evaluating the efficacy of NK cells therapy on stage IV NSCLC.
Collapse
|
69
|
Elevated Foxp3/CD8 Ratio in Lung Adenocarcinoma Metastatic Lymph Nodes Resected by Transcervical Extended Mediastinal Lymphadenectomy. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5185034. [PMID: 28831395 PMCID: PMC5558641 DOI: 10.1155/2017/5185034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/20/2017] [Accepted: 07/05/2017] [Indexed: 12/26/2022]
Abstract
A balance between tumor invasion and immune defence system is widely investigated. Objective. The aim of this study was to evaluate lymphocyte phenotype in lymph nodes (LNs) of patients with lung cancer in relation to the presence of metastases. Methods. We investigated 364 LNs resected by transcervical extended mediastinal lymphadenectomy (TEMLA) of 49 patients with squamous cell carcinoma (SCC) or adenocarcinoma (AD) with (A) and without metastases (B). Expression of CD4, CD8, CD25, CTLA-4, and Foxp3 was assessed by immunohistochemical staining. Results. We observed a strong nuclear staining for Foxp3 in lymphocytes and cancer cells and strong membranous/cytoplasmatic reaction for CD4 and CD8, but low for CD25 and CTLA-4. There were significantly higher proportions of CD8+ cells in AD (B) versus AD (A) LNs (80% versus 52.5%, p < 0.05). The Foxp3/CD8 ratio was higher in AD (A) versus AD (B) LNs (0.4 versus 0.25, p < 0.05). No significant differences in the cell markers expression in SCC LNs were found. Conclusion. Significant differences in lymphocyte phenotype in AD may indicate an exceptional biology of this type of lung cancer. TEMLA resected LNs may serve as valuable samples for evaluation of immune status in lung cancer patients.
Collapse
|
70
|
Shackelford RE, Ansari JM, Wei EX, Alexander JS, Cotelingam J. Anaplastic lymphoma kinase rearrangements in non-small-cell lung cancer: novel applications in diagnostics and treatment. Pharmacogenomics 2017; 18:1179-1192. [PMID: 28745554 DOI: 10.2217/pgs-2017-0098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The ALK gene, first identified as an anaplastic large cell lymphoma driver mutation, is dysregulated in nearly 20 different human malignancies, including 3-7% of non-small-cell lung cancers (NSCLC). In NSCLC, ALK commonly fuses with the EML4, forming a constitutively active tyrosine kinase that drives oncogenic progression. Recently, several ALK-inhibiting drugs have been developed that are more effective than standard chemotherapeutic regimens in treating advanced ALK-positive NSCLC. For this reason, molecular diagnostic testing for dysregulated ALK expression is a necessary part of identifying optimal NSCLC treatment options. Here, we review the molecular pathology of ALK-positive NSCLC, ALK molecular diagnostic techniques, ALK-targeted NSCLC treatments, and drug resistance mechanisms to ALK-targeted therapies.
Collapse
Affiliation(s)
| | - Junaid M Ansari
- Department of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, LA 71130, USA
| | - Eric X Wei
- Department of Pathology, LSU Health Shreveport, Shreveport, LA, USA
| | - Jonathan S Alexander
- Department of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, LA 71130, USA
| | - James Cotelingam
- Department of Pathology, LSU Health Shreveport, Shreveport, LA, USA
| |
Collapse
|
71
|
Calabrò L, Ceresoli GL, D'Incecco A, Scherpereel A, Aerts J, Maio M. Immune checkpoint therapy of mesothelioma: Pre-clinical bases and clinical evidences. Cytokine Growth Factor Rev 2017; 36:25-31. [PMID: 28736182 DOI: 10.1016/j.cytogfr.2017.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 12/22/2022]
Abstract
Treatment with immune-checkpoint blocking monoclonal antibody (mAb) is demonstrating a significant efficacy in different tumor types. Here, we discuss the impact of this promising approach in malignant mesothelioma (MM), a still dreadful disease in which medical treatment has been set on platinum based chemotherapy for decades with unsatisfactory results.
Collapse
Affiliation(s)
- Luana Calabrò
- Medical Oncology and Immunotherapy, Center for Immuno-Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy.
| | - Giovanni Luca Ceresoli
- Thoracic & GU Oncology Unit Department of Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Armida D'Incecco
- Medical Oncology and Immunotherapy, Center for Immuno-Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Arnaud Scherpereel
- Pulmonary and Thoracic Oncology, CHU de Lille, Univ Lille, Mesoclin Network, F59000 Lille, France
| | - Joachim Aerts
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Michele Maio
- Medical Oncology and Immunotherapy, Center for Immuno-Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| |
Collapse
|
72
|
Kuznetsova M, Lopatnikova J, Khantakova J, Maksyutov R, Maksyutov A, Sennikov S. Generation of populations of antigen-specific cytotoxic T cells using DCs transfected with DNA construct encoding HER2/neu tumor antigen epitopes. BMC Immunol 2017. [PMID: 28633645 PMCID: PMC5479015 DOI: 10.1186/s12865-017-0219-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Recent fundamental and clinical studies have confirmed the effectiveness of utilizing the potential of the immune system to remove tumor cells disseminated in a patient's body. Cytotoxic T lymphocytes (CTLs) are considered the main effectors in cell-mediated antitumor immunity. Approaches based on antigen presentation to CTLs by dendritic cells (DCs) are currently being intensively studied, because DCs are more efficient in tumor antigen presentation to T cells through their initiation of strong specific antitumor immune responses than other types of antigen-presenting cells. Today, it has become possible to isolate CTLs specific for certain antigenic determinants from heterogeneous populations of mononuclear cells. This enables direct and specific cell-mediated immune responses against cells carrying certain antigens. The aim of the present study was to develop an optimized protocol for generating CTL populations specific for epitopes of tumor-associated antigen HER2/neu, and to assess their cytotoxic effects against the HER2/neu-expressing MCF-7 tumor cell line. METHODS The developed protocol included sequential stages of obtaining mature DCs from PBMCs from HLA A*02-positive healthy donors, magnet-assisted transfection of mature DCs with the pMax plasmid encoding immunogenic peptides HER2 p369-377 (E75 peptide) and HER2 p689-697 (E88 peptide), coculture of antigen-activated DCs with autologous lymphocytes, magnetic-activated sorting of CTLs specific to HER2 epitopes, and stimulation of isolated CTLs with cytokines (IL-2, IL-7, and IL-15). RESULTS The resulting CTL populations were characterized by high contents of CD8+ cells (71.5% in cultures of E88-specific T cells and 90.2% in cultures of E75-specific T cells) and displayed strong cytotoxic effects against the MCF-7 cell line (percentages of damaged tumor cells in samples under investigation were 60.2 and 65.7% for E88- and E75-specific T cells, respectively; level of spontaneous death of target cells was 17.9%). CONCLUSIONS The developed protocol improves the efficiency of obtaining HER2/neu-specific CTLs and can be further used to obtain cell-based vaccines for eradicating targeted tumor cells to prevent tumor recurrence after the major tumor burden has been eliminated and preventing metastasis in patients with HER2-overexpressing tumors.
Collapse
Affiliation(s)
- Maria Kuznetsova
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Yadrintsevskaya str., 14, Novosibirsk, 630099, Russia
| | - Julia Lopatnikova
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Yadrintsevskaya str., 14, Novosibirsk, 630099, Russia
| | - Julia Khantakova
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Yadrintsevskaya str., 14, Novosibirsk, 630099, Russia
| | - Rinat Maksyutov
- State Research Center of Virology and Biotechnology "VECTOR", Koltsovo, Novosibirsk Region, 630559, Russia
| | - Amir Maksyutov
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Yadrintsevskaya str., 14, Novosibirsk, 630099, Russia.,State Research Center of Virology and Biotechnology "VECTOR", Koltsovo, Novosibirsk Region, 630559, Russia
| | - Sergey Sennikov
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology", Yadrintsevskaya str., 14, Novosibirsk, 630099, Russia.
| |
Collapse
|
73
|
Short-term clinical efficacy of percutaneous irreversible electroporation combined with allogeneic natural killer cell for treating metastatic pancreatic cancer. Immunol Lett 2017; 186:20-27. [DOI: 10.1016/j.imlet.2017.03.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 02/07/2023]
|
74
|
Lin M, Liang S, Wang X, Liang Y, Zhang M, Chen J, Niu L, Xu K. Cryoablation combined with allogenic natural killer cell immunotherapy improves the curative effect in patients with advanced hepatocellular cancer. Oncotarget 2017; 8:81967-81977. [PMID: 29137237 PMCID: PMC5669863 DOI: 10.18632/oncotarget.17804] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 04/29/2017] [Indexed: 12/17/2022] Open
Abstract
In this study, the clinical efficacy of cryosurgery combined with allogenic natural killer cell immunotherapy for advanced hepatocellular cancer was evaluated. From October 2015 to March 2017, we enrolled 61 patients who met the enrollment criteria and divided them into two groups: 1) the simple cryoablation group (Cryo group, n = 26); and 2) the cryoablation combined with allogenic natural killer cells group (Cryo-NK group, n = 35), the safety and short-term effects were evaluated firstly, then the median progression-free survival, response rate and disease control rate were assessed. All adverse events experienced by the patients were recorded, and included local (e.g., pain, pleural effusion, and ascites) and systemic (e.g., chills, fatigue, and fever) reactions, fever was more frequent. Other possible seriously side effects (e.g., blood or bone marrow changes) were not detected. Combining allogeneic natural killer cells with cryoablation had a synergistic effect, not only enhancing the immune function, improving the quality of life of the patients, but also reducing the expression of AFP and significantly exhibiting good clinical efficacy of the patients. After a median follow-up of 8.7 months (3.9 -15.1months), median progression-free survival was higher in Cryo-NK (9.1 months) than in Cryo (7.6 months, P = 0.0107), median progression-free survival who received multiple natural killer was higher than who just received single natural killer (9.7 months vs.8.4 months, P = 0.0011, respectively), the response rate in Cryo-NK (60.0%) was higher than in Cryo (46.1%, P < 0.05), the disease control rate in Cryo-NK (85.7%) was higher than in Cryo group (69.2%, P < 0.01). Percutaneous cryoablation combined with allogeneic natural killer cell immunotherapy significantly increased median progression-free survival of advanced hepatocellular cancer patients. Multiple allogeneic natural killer cells infusion was associated with better prognosis to advanced hepatocellular cancer.
Collapse
Affiliation(s)
- Mao Lin
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China.,Fuda Cancer Institute, Guangzhou, China
| | - Shuzhen Liang
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China
| | - Xiaohua Wang
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China
| | - Yinqing Liang
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China
| | | | - Jibing Chen
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China.,Fuda Cancer Institute, Guangzhou, China
| | - Lizhi Niu
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China.,Fuda Cancer Institute, Guangzhou, China.,Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China
| | - Kecheng Xu
- Department of Biological Treatment Center, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China.,Fuda Cancer Institute, Guangzhou, China
| |
Collapse
|
75
|
Korczak-Kowalska G, Stelmaszczyk-Emmel A, Bocian K, Kiernozek E, Drela N, Domagała-Kulawik J. Expanding Diversity and Common Goal of Regulatory T and B Cells. II: In Allergy, Malignancy, and Transplantation. Arch Immunol Ther Exp (Warsz) 2017; 65:523-535. [PMID: 28470464 PMCID: PMC5688211 DOI: 10.1007/s00005-017-0471-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/19/2017] [Indexed: 01/03/2023]
Abstract
Regulation of immune response was found to play an important role in the course of many diseases such as autoimmune diseases, allergy, malignancy, organ transplantation. The studies on immune regulation focus on the role of regulatory cells (Tregs, Bregs, regulatory myeloid cells) in these disorders. The number and function of Tregs may serve as a marker of disease activity. As in allergy, the depletion of Tregs is observed and the results of allergen-specific immunotherapy could be measured by an increase in the population of IL-10+ regulatory cells. On the basis of the knowledge of anti-cancer immune response regulation, new directions in therapy of tumors are introduced. As the proportion of regulatory cells is increased in the course of neoplasm, the therapeutic action is directed at their inhibition. The depletion of Tregs may be also achieved by an anti-check-point blockade, anti-CD25 agents, and inhibition of regulatory cell recruitment to the tumor site by affecting chemokine pathways. However, the possible favorable role of Tregs in cancer development is considered and the plasticity of immune regulation should be taken into account. The new promising direction of the treatment based on regulatory cells is the prevention of transplant rejection. A different way of production and implementation of classic Tregs as well as other cell types such as double-negative cells, Bregs, CD4+ Tr1 cells are tested in ongoing trials. On the basis of the results of current studies, we could show in this review the significance of therapies based on regulatory cells in different disorders.
Collapse
Affiliation(s)
- Grażyna Korczak-Kowalska
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Warsaw, Poland
| | - Anna Stelmaszczyk-Emmel
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Bocian
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Ewelina Kiernozek
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Nadzieja Drela
- Department of Immunology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
76
|
Steendam CM, Dammeijer F, Aerts JGJV, Cornelissen R. Immunotherapeutic strategies in non-small-cell lung cancer: the present and the future. Immunotherapy 2017; 9:507-520. [DOI: 10.2217/imt-2016-0151] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is still the leading cause of cancer death worldwide, with a poor prognosis. In the era of immunotherapies, the field is rapidly changing, and the clinician needs to be aware of the current state and future perspectives of immunotherapeutic strategies. In this review, we discuss the current status of immune checkpoint inhibitors, cancer vaccines and cellular therapies specifically in NSCLC. Last but not least, we will discuss rational combination strategies that are promising for the near future.
Collapse
Affiliation(s)
- Christi M Steendam
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, ‘s-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
- Department of Pulmonary Medicine, Amphia Hospital, Molengracht 21, 4818 CK Breda, The Netherlands
| | - Floris Dammeijer
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, ‘s-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, ‘s-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
- Department of Pulmonary Medicine, Amphia Hospital, Molengracht 21, 4818 CK Breda, The Netherlands
| | - Robin Cornelissen
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, ‘s-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| |
Collapse
|
77
|
Malhotra J, Jabbour SK, Aisner J. Current state of immunotherapy for non-small cell lung cancer. Transl Lung Cancer Res 2017; 6:196-211. [PMID: 28529902 PMCID: PMC5420529 DOI: 10.21037/tlcr.2017.03.01] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/22/2017] [Indexed: 12/26/2022]
Abstract
Lung cancer is the leading cause of cancer mortality and non-small cell lung cancer (NSCLC) accounts for more than 85% of all lung cancers. Platinum-based doublet chemotherapy is the standard first-line treatment for metastatic NSCLC when genomic testing reveals no targetable alteration such as epidermal growth factor receptor (EGFR) mutations, anaplastic lymphoma kinase (ALK) or ROS1 translocation/re-arrangements. But, chemotherapy produces response rates ranging only between 15-30%. For patients whose disease progresses on first-line chemotherapy, second-line therapy historically consists of taxane-based salvage chemotherapy with a response rate of less than 25%. Recently, immunotherapy with checkpoint inhibitor agents have demonstrated responses in advanced NSCLC, with some patients exhibiting durable responses after discontinuing therapy. In 2015, two immune checkpoint inhibitors targeting programmed cell death-1 (PD-1), nivolumab and pembrolizumab were approved for second-line therapy of NSCLC. In 2016, another checkpoint inhibitor targeting program death-ligand 1 (PD-L1), atezolizumab was approved for the same indication. Moreover, pembrolizumab also received approval in 2016 for first-line NSCLC treatment in patients with high PD-L1 expressing tumors. Immunotherapy for NSCLC has therefore, recently evolved into a true treatment modality with the acceptance of PD-1 and PD-L1 inhibitors as the new standard of care for second-line treatment. However, it is still at the discretion of the treating physician whether to use PD-1 or PD-L1 inhibitor as data to compare these two pathways is lacking. Focus is now also on exploring their role in the adjuvant and consolidation settings for NSCLC as well as on exploring novel combinations combining these agents with chemotherapy or radiation. Research is also needed in the development of predictive and prognostic biomarkers for these agents. While vaccine therapy trials in NSCLC have so far failed to show significant clinical benefit, the demonstration of enhanced immune response in these trials suggest the vaccine therapy needs additional evaluation in combination with other therapeutic modalities especially checkpoint inhibition.
Collapse
Affiliation(s)
- Jyoti Malhotra
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Salma K Jabbour
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Joseph Aisner
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
78
|
Prospective study of percutaneous cryoablation combined with allogenic NK cell immunotherapy for advanced renal cell cancer. Immunol Lett 2017; 184:98-104. [PMID: 28274792 DOI: 10.1016/j.imlet.2017.03.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 11/23/2022]
Abstract
In this study, the clinical efficacy of cryosurgery combined with allogenic NK cell immunotherapy for advanced renal cell cancer was evaluated. From July to December 2016, we enrolled 60 patients who met the enrollment criteria and divided them into two groups: (1) the simple cryoablation group (n=30); and (2) the cryoablation combined with allogenic NK cells group (n=30). The clinical efficacy, quality of life, immune function, and other related indicators were evaluated. Combining allogeneic NK cells with cryoablation had a synergistic effect, not only enhancing the immune function and improving the quality of life of the patients, but also significantly exhibiting good clinical efficacy of the patients. This study is the first clinical trial that has evaluated the safety and efficacy of allogenic NK cells combined with cryosurgery for the treatment of renal cell cancer.
Collapse
|
79
|
Abstract
In vivo imaging, which enables us to peer deeply within living subjects, is producing tremendous opportunities both for clinical diagnostics and as a research tool. Contrast material is often required to clearly visualize the functional architecture of physiological structures. Recent advances in nanomaterials are becoming pivotal to generate the high-resolution, high-contrast images needed for accurate, precision diagnostics. Nanomaterials are playing major roles in imaging by delivering large imaging payloads, yielding improved sensitivity, multiplexing capacity, and modularity of design. Indeed, for several imaging modalities, nanomaterials are now not simply ancillary contrast entities, but are instead the original and sole source of image signal that make possible the modality's existence. We address the physicochemical makeup/design of nanomaterials through the lens of the physical properties that produce contrast signal for the cognate imaging modality-we stratify nanomaterials on the basis of their (i) magnetic, (ii) optical, (iii) acoustic, and/or (iv) nuclear properties. We evaluate them for their ability to provide relevant information under preclinical and clinical circumstances, their in vivo safety profiles (which are being incorporated into their chemical design), their modularity in being fused to create multimodal nanomaterials (spanning multiple different physical imaging modalities and therapeutic/theranostic capabilities), their key properties, and critically their likelihood to be clinically translated.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Stanford University , 3155 Porter Drive, #1214, Palo Alto, California 94304-5483, United States
| | - Sanjiv Sam Gambhir
- The James H. Clark Center , 318 Campus Drive, First Floor, E-150A, Stanford, California 94305-5427, United States
| |
Collapse
|
80
|
Chen YM, Lai CH, Rau KM, Huang CH, Chang HC, Chao TY, Tseng CC, Fang WF, Chung YH, Wang YH, Su MC, Huang KT, Liu SF, Chen HC, Chang YC, Chang YP, Wang CC, Lin MC. Impact of clinical parameters and systemic inflammatory status on epidermal growth factor receptor-mutant non-small cell lung cancer patients readministration with epidermal growth factor receptor tyrosine kinase inhibitors. BMC Cancer 2016; 16:868. [PMID: 27821111 PMCID: PMC5100346 DOI: 10.1186/s12885-016-2917-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 10/31/2016] [Indexed: 11/21/2022] Open
Abstract
Background Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI) readministration to lung cancer patients is common owing to the few options available. Impact of clinical factors on prognosis of EGFR-mutant non-small cell lung cancer (NSCLC) patients receiving EGFR-TKI readministration after first-line EGFR-TKI failure and a period of TKI holiday remains unclear. Through this retrospective study, we aimed to understand the impact of clinical factors in such patients. Methods Of 1386 cases diagnosed between December 2010 and December 2013, 80 EGFR-mutant NSCLC patients who were readministered TKIs after failure of first-line TKIs and intercalated with at least one cycle of cytotoxic agent were included. We evaluated clinical factors that may influence prognosis of TKI readministration as well as systemic inflammatory status in terms of neutrophil-to-lymphocyte ratio (NLR) and lymphocyte-to-monocyte ratio (LMR). Baseline NLR and LMR were estimated at the beginning of TKI readministration and trends of NLR and LMR were change amount from patients receiving first-Line TKIs to TKIs readministration. Results Median survival time since TKI readministration was 7.0 months. In the univariable analysis, progression free survival (PFS) of first-line TKIs, baseline NLR and LMR, and trend of LMR were prognostic factors in patients receiving TKIs readministration. In the multivariate analysis, only PFS of first-line TKIs (p < 0.001), baseline NLR (p = 0.037), and trend of LMR (p = 0.004) were prognostic factors. Conclusion Longer PFS of first-line TKIs, low baseline NLR, and high trend of LMR were good prognostic factors in EGFR-mutant NSCLC patients receiving TKI readministration. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2917-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu-Mu Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Chien-Hao Lai
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Kun-Ming Rau
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Cheng-Hua Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Huang-Chih Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Tung-Ying Chao
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Chia-Cheng Tseng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Wen-Feng Fang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan.,Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi Campus, Chiayi, Taiwan
| | - Yu-Hsiu Chung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Yi-Hsi Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Mao-Chang Su
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Kuo-Tung Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Shih-Feng Liu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Hung-Chen Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Ya-Chun Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Yu-Ping Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Chin-Chou Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, No. 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, Taiwan.
| |
Collapse
|
81
|
Cukic V. NEUTROPHIL/LYMPHOCYTE RATIO AND PLATELET/LYMPHOCYTE RATIO IN PATIENTS WITH NSCLC. Mater Sociomed 2016; 28:378-381. [PMID: 27999489 PMCID: PMC5149430 DOI: 10.5455/msm.2016.28.378-381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 10/08/2016] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE to compare neutrophil/lymphocyte ratio (NLR) and platelet/lymphocyte ratio (PLR) in patients with NSCLC (Non- Small- Cell Lung Cancer): with and without metastases at the time of diagnosis to find out if there is the importance of these cell ratios in the assessment of severity NSCLC. MATERIAL AND METHODS this is the retrospective analysis of NRL and PRL in patients with NSCLC at the time of the diagnosis of disease before any anti tumor treatment (chemotherapy, radiotherapy, surgery). 57 of patients with NSCLC treated in the first three months of 2016. year were chosen at random regardless of sex and age. We examined full blood count cells (FBC), calculated NLR and PLR in every patient and compared obtained values in patients with and patients without metastases. RESULTS In 57 patients with NSCLC there were 15 males with metastases, 28 without metastases, and 8 females with metastases, 6 without metastases. Since there was no regularity in the distribution of obtained values of NLR and PLR we made the Mann-Whitney U test. Mean values are presented with a median and interquartile percentiles. There was no significant difference in NLR between patients without and with metastases (p = 0.614; p = NS) as well as in PLR (p=0,068; p=NS). CONCLUSION There must be a link between the immune status of the organism and lung cancer development. Immune cells have become of interest in recent years and much work has been done to study their role in the genesis of cancer but it did not give satisfactory results. Further clinical studies on large number of patients and further laboratory examination of the role of immune cells in cancer development and suppression are required.
Collapse
Affiliation(s)
- Vesna Cukic
- Clinic for Pulmonary Diseases and TB "Podhrastovi", Clinical centre of Sarajevo University, Bosnia and Herzegovina
| |
Collapse
|
82
|
Lievense LA, Cornelissen R, Bezemer K, Kaijen-Lambers ME, Hegmans JP, Aerts JG. Pleural Effusion of Patients with Malignant Mesothelioma Induces Macrophage-Mediated T Cell Suppression. J Thorac Oncol 2016; 11:1755-64. [DOI: 10.1016/j.jtho.2016.06.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/24/2016] [Accepted: 06/11/2016] [Indexed: 11/25/2022]
|
83
|
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]
|
84
|
Mountzios G, Linardou H, Kosmidis P. Immunotherapy in non-small cell lung cancer: the clinical impact of immune response and targeting. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:268. [PMID: 27563655 PMCID: PMC4971380 DOI: 10.21037/atm.2016.06.24] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/20/2016] [Indexed: 12/21/2022]
Abstract
Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related death worldwide. In recent years, through a better understanding of the interactions between the immune system and tumor cells (TC), immunotherapy has emerged as a promising therapeutic strategy. Chemotherapy has long been reported to interfere with the immune response to the tumor and conversely, anti-tumor immunity may add to those effects. Anti-tumor vaccines, such as MAGE-A3, Tecetomide, TG4010, CIMAvax, tumor cell vaccines and dendritic cell (DC) vaccines emerged as potent inducers of the immune response against the tumor. More recently the approval of the anti-programmed cell death 1 (anti-PD-1) monoclonal antibodies nivolumab and pembrolizumab for previously treated advanced squamous and non-squamous NSCLC, as well as other immune checkpoint inhibitors delivering promising results, has radically transformed the therapeutic landscape of NSCLC. Combination strategies now appear as the next step. Notwithstanding these successes, immunotherapy still holds significant drawbacks and currently several improvements are needed before routine use in clinical practice, including identification of robust biomarkers for optimal patient selection, as well as defining the best way to evaluate response.
Collapse
Affiliation(s)
- Giannis Mountzios
- Department of Medical Oncology, University of Athens School of Medicine, Athens, Greece
| | - Helena Linardou
- 1 Oncology Department, Metropolitan Hospital, Piraeus, Greece
| | | |
Collapse
|
85
|
Khanna P, Blais N, Gaudreau PO, Corrales-Rodriguez L. Immunotherapy Comes of Age in Lung Cancer. Clin Lung Cancer 2016; 18:13-22. [PMID: 27461776 DOI: 10.1016/j.cllc.2016.06.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/07/2016] [Accepted: 06/13/2016] [Indexed: 11/25/2022]
Abstract
Lung carcinoma is the leading cause of death by cancer worldwide. When possible, surgery is the best treatment strategy for patients with non-small-cell lung cancer. However, even with curative-intent therapy, most patients will develop local or systemic recurrence and, ultimately, succumb to their disease. In recent years, evidence on the role of the antitumor activity of the immune system and the understanding of tumor immunosurveillance have resulted in the emergence of immunotherapy as a promising therapeutic approach in lung cancer. The main approaches are immune checkpoint inhibition, such as blockade of the cytotoxic T-lymphocyte antigen-4 and programmed cell death-1 receptors and the programmed cell death-1 ligand, and vaccine therapy, which elicits specific antitumor immunity against relevant tumor-associated antigens. We have reviewed recently reported results from clinical trials and the possible future role of vaccine therapy and immune checkpoint inhibition in the treatment of small cell lung cancer and non-small-cell lung cancer.
Collapse
Affiliation(s)
- Priyanka Khanna
- Centro de Investigación y Manejo del Cáncer (CIMCA), San Jose, Costa Rica
| | - Normand Blais
- Medical Oncology and Hematology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Pierre-Olivier Gaudreau
- Medical Oncology and Hematology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Luis Corrales-Rodriguez
- Medical Oncology, Centro de Investigación y Manejo del Cáncer (CIMCA), San Jose, Costa Rica.
| |
Collapse
|
86
|
Bremnes RM, Busund LT, Kilvær TL, Andersen S, Richardsen E, Paulsen EE, Hald S, Khanehkenari MR, Cooper WA, Kao SC, Dønnem T. The Role of Tumor-Infiltrating Lymphocytes in Development, Progression, and Prognosis of Non-Small Cell Lung Cancer. J Thorac Oncol 2016; 11:789-800. [PMID: 26845192 DOI: 10.1016/j.jtho.2016.01.015] [Citation(s) in RCA: 336] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/24/2016] [Accepted: 01/26/2016] [Indexed: 12/25/2022]
Abstract
A malignant tumor is not merely an accumulation of neoplastic cells, but constitutes a microenvironment containing endothelial cells, fibroblasts, structural components, and infiltrating immune cells that impact tumor development, invasion, metastasis, and outcome. Hence, the evolution of cancers reflects intricate cellular and molecular interactions between tumor cells and constituents of the tumor microenvironment. Recent studies have shed new light on this complex interaction between tumor and host immune cells and the resulting immune response. The composition of the immune microenvironment differs across patients as well as in cancers of the same type, including various populations of T cells, B cells, dendritic cells, natural killer cells, myeloid-derived suppressor cells, neutrophils, and macrophages. The type, density, location, and organization of immune cells within solid tumors define the immune contexture, which has proved to be a major determinant of tumor characteristics and patient outcome. Lung cancer consists mostly of non-small cell lung cancer (85%); it is our most deadly malignant disease, with the 5-year survival rate being merely 15%. This review focuses on the immune contexture; the tumor-suppressing roles of tumor-infiltrating lymphocytes; and the relevance of this immune contexture for cancer diagnostics, prognostication, and treatment allocation, with an emphasis on non-small cell lung cancer.
Collapse
Affiliation(s)
- Roy M Bremnes
- Institute of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway; Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway.
| | - Lill-Tove Busund
- Institute of Medical Biology, The Arctic University of Norway, Tromsø, Norway; Department of Pathology, University Hospital of Northern Norway, Tromsø, Norway
| | - Thomas L Kilvær
- Institute of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway; Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway
| | - Sigve Andersen
- Institute of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway; Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway
| | - Elin Richardsen
- Institute of Medical Biology, The Arctic University of Norway, Tromsø, Norway; Department of Pathology, University Hospital of Northern Norway, Tromsø, Norway
| | - Erna Elise Paulsen
- Institute of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway; Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway
| | - Sigurd Hald
- Institute of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway; Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway
| | | | - Wendy A Cooper
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia; School of Medicine, University of Western Sydney, New South Wales, Australia; Sydney Medical School, University of Sydney, New South Wales, Australia
| | - Steven C Kao
- Chris O'Brien Lifehouse, Sydney, New South Wales, Australia; Asbestos Diseases Research Institute, Sydney, New South Wales, Australia; University of Sydney, Sydney, New South Wales, Australia
| | - Tom Dønnem
- Institute of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway; Department of Oncology, University Hospital of Northern Norway, Tromsø, Norway
| |
Collapse
|
87
|
Sun H, Liu D. IL-15/sIL-15Rα gene transfer suppresses Lewis lung cancer growth in the lungs, liver and kidneys. Cancer Gene Ther 2016; 23:54-60. [PMID: 26742578 DOI: 10.1038/cgt.2015.67] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 11/26/2015] [Accepted: 12/02/2015] [Indexed: 11/09/2022]
Abstract
Nearly 40% of people with lung cancer have tumor growth in other organs at the time of diagnosis. Current treatment strategies for patients with late-stage lung cancer are primarily palliative and only showed modest efficacy. The current study takes advantage of the hydrodynamic gene delivery technique to evaluate the antitumor activity of interleukin (IL)-15/sIL-15Rα on lung tumors growing in the lungs, liver and kidneys. We demonstrate that hydrodynamic tail vein injection of 2 μg of AG209 DP muIL-15sRα+IL-15 plasmid resulted in serum IL-15/sIL-15Rα reaching a peak level of ~10 μg ml(-1) 1 day after the injection and gradually declined to ~5 ng ml(-1) within 3 days. Quantitative PCR analysis revealed that overexpression of IL-15/sIL-15Rα induced the activation of natural killer and T cells, evidenced by increased mRNA levels of marker genes including granzyme B, perforin, Ifn-γ, T-bet and Cd8 in the lungs, liver and kidneys. Importantly, transfer of the Il-15/sIl-15Rα gene alone, or in combination with gemcitabine chemotherapy, significantly inhibited the tumor growth in these three organs and prolonged median survival time of treated mice by 1.7- and 3.3-fold, respectively. The therapeutic benefits are principally blockade and elimination of tumor growth in the liver and kidneys. Taken together, these results suggest that IL-15/sIL-15Rα-based gene therapy could be an effective approach to treat late-stage lung cancer with metastases in other organs.
Collapse
Affiliation(s)
- H Sun
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - D Liu
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| |
Collapse
|
88
|
Abstract
BACKGROUND Treatment-related immunosuppression in organ transplant recipients has been linked to increased incidence and risk of progression for several malignancies. Using a population-based cancer cohort, we evaluated whether organ transplantation was associated with worse prognosis in elderly patients with non-small cell lung cancer (NSCLC). METHODS Using the Surveillance, Epidemiology, and End Results Registry linked to Medicare claims, we identified 597 patients aged 65 years or older with NSCLC who had received organ transplants (kidney, liver, heart, or lung) before cancer diagnosis. These cases were compared to 114,410 untransplanted NSCLC patients. We compared overall survival (OS) by transplant status using Kaplan-Meier methods and Cox regression. To account for an increased risk of non-lung cancer death (competing risks) in transplant recipients, we used conditional probability function (CPF) analyses. Multiple CPF regression was used to evaluate lung cancer prognosis in organ transplant recipients while adjusting for confounders. RESULTS Transplant recipients presented with earlier stage lung cancer (P = 0.002) and were more likely to have squamous cell carcinoma (P = 0.02). Cox regression analyses showed that having received a non-lung organ transplant was associated with poorer OS (P < 0.05), whereas lung transplantation was associated with no difference in prognosis. After accounting for competing risks of death using CPF regression, no differences in cancer-specific survival were noted between non-lung transplant recipients and nontransplant patients. CONCLUSIONS Non-lung solid organ transplant recipients who developed NSCLC had worse OS than nontransplant recipients due to competing risks of death. Lung cancer-specific survival analyses suggest that NSCLC tumor behavior may be similar in these 2 groups.
Collapse
|
89
|
Lievense L, Aerts J, Hegmans J. Immune Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 893:59-90. [PMID: 26667339 DOI: 10.1007/978-3-319-24223-1_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lung cancer has long been considered an unsuitable target for immunotherapy due to its proposed immunoresistant properties. However, recent evidence has shown that anti-tumor immune responses can occur in lung cancer patients, paving the way for lung cancer as a novel target for immunotherapy. In order to take full advantage of the potential of immunotherapy, research is focusing on the presence and function of various immunological cell types in the tumor microenvironment. Immune cells which facilitate or inhibit antitumor responses have been identified and their prognostic value in lung cancer has been established. Knowledge regarding these pro- and anti-tumor immune cells and their mechanisms of action has facilitated the identification of numerous potential immunotherapeutic strategies and opportunities for intervention. A plethora of immunotherapeutic approaches is currently being developed and studied in lung cancer patients and phase 3 clinical trials are ongoing. Many different immunotherapies have shown promising clinical effects in patients with limited and advanced stage lung cancer, however, future years will have to tell whether immunotherapy will earn its place in the standard treatment of lung cancer.
Collapse
Affiliation(s)
- Lysanne Lievense
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, Rotterdam, 3015 GD, The Netherlands
| | - Joachim Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, Rotterdam, 3015 GD, The Netherlands
| | - Joost Hegmans
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Dr. Molewaterplein 50, Rotterdam, 3015 GD, The Netherlands.
| |
Collapse
|
90
|
Li J, Ai Y, Wang L, Bu P, Sharkey CC, Wu Q, Wun B, Roy S, Shen X, King MR. Targeted drug delivery to circulating tumor cells via platelet membrane-functionalized particles. Biomaterials 2015; 76:52-65. [PMID: 26519648 DOI: 10.1016/j.biomaterials.2015.10.046] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/15/2015] [Accepted: 10/18/2015] [Indexed: 12/11/2022]
Abstract
Circulating tumor cells (CTCs) are responsible for metastases in distant organs via hematogenous dissemination. Fundamental studies in the past decade have suggested that neutralization of CTCs in circulation could represent an effective strategy to prevent metastasis. Current paradigms of targeted drug delivery into a solid tumor largely fall into two main categories: unique cancer markers (e.g. overexpression of surface receptors) and tumor-specific microenvironment (e.g. low pH, hypoxia, etc.). While relying on a surface receptor to target CTCs can be greatly challenged by cancer heterogeneity, targeting of tumor microenvironments has the advantage of recognizing a broader spectrum of cancer cells regardless of genetic differences or tumor types. The blood circulation, however, where CTCs transit through, lacks the same tumor microenvironment as that found in a solid tumor. In this study, a unique "microenvironment" was confirmed upon introduction of cancer cells of different types into circulation where activated platelets and fibrin were physically associated with blood-borne cancer cells. Inspired by this observation, synthetic silica particles were functionalized with activated platelet membrane along with surface conjugation of tumor-specific apoptosis-inducing ligand cytokine, TRAIL. Biomimetic synthetic particles incorporated into CTC-associated micro-thrombi in lung vasculature and dramatically decreased lung metastases in a mouse breast cancer metastasis model. Our results demonstrate a "Trojan Horse" strategy of neutralizing CTCs to attenuate metastasis.
Collapse
Affiliation(s)
- Jiahe Li
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| | - Yiwei Ai
- Department of Biomedical Engineering, Duke University Durham, NC, 27708, USA
| | - Lihua Wang
- Department of Biomedical Engineering, Duke University Durham, NC, 27708, USA
| | - Pengcheng Bu
- Department of Biomedical Engineering, Duke University Durham, NC, 27708, USA
| | - Charles C Sharkey
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Qianhui Wu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Brittany Wun
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Sweta Roy
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Duke University Durham, NC, 27708, USA
| | - Michael R King
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
91
|
Dingemans AMC, Groen HJM, Herder GJM, Stigt JA, Smit EF, Bahce I, Burgers JA, van den Borne BEEM, Biesma B, Vincent A, van der Noort V, Aerts JG. A randomized phase II study comparing paclitaxel-carboplatin-bevacizumab with or without nitroglycerin patches in patients with stage IV nonsquamous nonsmall-cell lung cancer: NVALT12 (NCT01171170)†. Ann Oncol 2015; 26:2286-93. [PMID: 26347109 DOI: 10.1093/annonc/mdv370] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/18/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Nitroglycerin (NTG) increases tumor blood flow and oxygenation by inhibiting hypoxia-inducible-factor (HIF)-1. A randomized phase II study has shown improved outcome when NTG patches were added to vinorelbine/cisplatin in patients with advanced nonsmall-cell lung cancer (NSCLC). In addition, there is evidence that the combination of bevacizumab and HIF-1 inhibitors increases antitumor activity. PATIENTS AND METHODS In this randomized phase II trial, chemo-naive patients with stage IV nonsquamous NSCLC were randomized to four cycles of carboplatin (area under the curve 6)-paclitaxel (200 mg/m(2))-bevacizumab 15 mg/kg on day 1 every 3 weeks with or without NTG patches 15 mg (day -2 to +2) followed by bevacizumab with or without NTG until progression. Response was assessed every two cycles. Primary end point was progression-free survival (PFS). The study was powered (80%) to detect a decrease in the hazard of tumor progression of 33% at α = 0.05 with a two-sided log-rank test when 222 patients were enrolled and followed until 195 events were observed. RESULTS Between 1 January 2011 and 1 January 2013, a total of 223 patients were randomized; 112 control arm and 111 experimental arm; response rate was 54% in control arm and 38% in experimental arm. Median [95% confidence interval (CI)] PFS in control arm was 6.8 months (5.6-7.3) and 5.1 months (4.2-5.8) in experimental arm, hazard ratio (HR) 1.27 (95% CI 0.96-1.67). Overall survival (OS) was 11.6 months (8.8-13.6) in control arm and 9.4 months (7.8-11.3) in experimental arm, HR 1.02 (95% CI 0.71-1.46). In the experimental arm, no additional toxicity was observed except headache (6% versus 52% in patients treated with NTG). CONCLUSION Adding NTG to first-line carboplatin-paclitaxel-bevacizumab did not improve PFS and OS in patients with stage IV nonsquamous NSCLC.
Collapse
Affiliation(s)
- A-M C Dingemans
- Department of Respiratory Disease, Maastricht University Medical Center, Maastricht
| | - H J M Groen
- Department of Respiratory Disease, University Medical Center Groningen, Groningen
| | - G J M Herder
- Department of Respiratory Disease, Sint Antonius Hospital, Nieuwegein
| | - J A Stigt
- Department of Respiratory Disease, Isala Hospital, Zwolle
| | - E F Smit
- Department of Respiratory Disease, VU Medical Center, Amsterdam Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam
| | - I Bahce
- Department of Respiratory Disease, VU Medical Center, Amsterdam
| | - J A Burgers
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam
| | | | - B Biesma
- Department of Respiratory Disease, Jeroen Bosch Hospital's, Hertogenbosch
| | - A Vincent
- Department of Biostatistics, Netherlands Cancer Institute, Amsterdam
| | - V van der Noort
- Department of Biostatistics, Netherlands Cancer Institute, Amsterdam
| | - J G Aerts
- Department of Respiratory Disease, Amphia Hospital, Breda Department of Respiratory Disease, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | |
Collapse
|
92
|
Efficacy and safety of dendritic cells co-cultured with cytokine-induced killer cells immunotherapy for non-small-cell lung cancer. Int Immunopharmacol 2015; 28:22-8. [DOI: 10.1016/j.intimp.2015.05.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 05/05/2015] [Accepted: 05/13/2015] [Indexed: 11/19/2022]
|
93
|
Chen YM, Lai CH, Chang HC, Chao TY, Tseng CC, Fang WF, Wang CC, Chung YH, Wang YH, Su MC, Huang KT, Chen HC, Chang YC, Lin MC. Baseline and Trend of Lymphocyte-to-Monocyte Ratio as Prognostic Factors in Epidermal Growth Factor Receptor Mutant Non-Small Cell Lung Cancer Patients Treated with First-Line Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors. PLoS One 2015; 10:e0136252. [PMID: 26313661 PMCID: PMC4552380 DOI: 10.1371/journal.pone.0136252] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/02/2015] [Indexed: 11/18/2022] Open
Abstract
Background Patients with early-stage lung cancer who have a high baseline lymphocyte-to-monocyte ratio (LMR) have a favorable prognosis. However, the prognostic significance of LMR in patients with advanced-stage EGFR-mutant non-small cell lung cancer (NSCLC) receiving first-line epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) has not been established. We conducted a retrospective analysis to investigate the influence of LMR on clinical outcomes including progression-free survival (PFS) and overall survival (OS) in EGFR-mutant patients with NSCLC. Materials and Methods Of 1310 lung cancer patients diagnosed between January 2011 and October 2013, 253 patients receiving first-line EGFR-TKIs for EGFR-mutant NSCLC were included. The cut-off values for baseline and the 1-month-to-baseline ratio of LMR (MBR), determined by using receiver operating characteristic curves, were 3.29 and 0.63, respectively. Patients were divided into 3 prognostic groups: high LMR and MBR, high LMR or MBR, and low LMR and MBR. Results The mean patient age was 65.2 years, and 41% were men. The median PFS and OS were 10.3 and 22.0 months, respectively. The PFS in patients with high LMR and MBR, high LMR or MBR, and low LMR and MBR were 15.4, 7.1, and 2.0 months, respectively (p < 0.001), whereas the OS were 32.6, 13.7, and 5.1 months, respectively (p < 0.001). Conclusion A combination of baseline and trend of LMR can be used to identify patients with a high mortality risk in EGFR-mutant NSCLC patients receiving first-line EGFR-TKIs.
Collapse
Affiliation(s)
- Yu-Mu Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chien-Hao Lai
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Huang-Chih Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tung-Ying Chao
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Cheng Tseng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Feng Fang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Respiratory Care, Chang Gung Institute of Technology, Chiayi, Taiwan
| | - Chin-Chou Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Hsiu Chung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Hsi Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Mao-Chang Su
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kuo-Tung Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hung-Chen Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ya-Chun Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
94
|
Singha AK, Bhattacharjee B, Maiti D. Cytotoxic activity of T lymphocytes is induced upon stimulation with IL-3 plus GM-CSF in animal leukemia model. Leuk Res 2015; 39:S0145-2126(15)30364-7. [PMID: 26350142 DOI: 10.1016/j.leukres.2015.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 08/09/2015] [Accepted: 08/20/2015] [Indexed: 01/03/2023]
Abstract
Chemotherapy for leukemia has severe toxicity and bone marrow transplantation is both financially and logistically demanding. Therefore, immunotherapy is a feasible and promising approach to treat leukemia. For immunotherapy, cytotoxic T lymphocytes (CTL) against leukemic cells were induced. In BALB/c mice, leukemia was induced by N-ethyl-N'-nitrosourea (ENU). The mice were treated with recombinant IL-3 and GM-CSF - both 5μg/kg/day for four days to induce functional CTL. The IL-3+GM-CSF treatment increased total leukocyte counts, accompanied by significant increase in CTL activity, in the leukemic mice. The IL-3+GM-CSF treatment also enhanced the expression of both p40 and p35 isoforms of IL-12. Perforin and granzyme B expressions were increased in the treated group supporting the T lymphocyte-mediated cytotoxic killing of the target cells. The protein tyrosine kinase (PTK) activity was increased in leukemia but decreased after the treatment with IL-3 and GM-CSF. Interferon gamma (IFN-γ) production was decreased in leukemic condition but increased after the treatment with these colony stimulating factors. These data indicate the anti-leukemic potential of the IL-3 and GM-CSF combination therapy.
Collapse
Affiliation(s)
- Ashish Kumar Singha
- Immunology microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Bhaskar Bhattacharjee
- Immunology microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Debasish Maiti
- Immunology microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India.
| |
Collapse
|
95
|
Foster JG, Wong SCK, Sharp TV. The hypoxic tumor microenvironment: driving the tumorigenesis of non-small-cell lung cancer. Future Oncol 2015; 10:2659-74. [PMID: 25531051 DOI: 10.2217/fon.14.201] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Since the application of molecular biology in cancer biology, lung cancer research has classically focused on molecular drivers of disease. One such pathway, the hypoxic response pathway, is activated by reduced local oxygen concentrations at the tumor site. Hypoxia-driven gene and protein changes enhance epithelial-to-mesenchymal transition, remodel the extracellular matrix, drive drug resistance, support cancer stem cells and aid evasion from immune cells. However, it is not the tumor cells alone which drive this response to hypoxia, but rather their interaction with a complex milieu of supporting cells. This review will focus on recent advances in our understanding of how these cells contribute to the tumor response to hypoxia in non-small-cell lung cancer.
Collapse
Affiliation(s)
- John G Foster
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | | | | |
Collapse
|
96
|
Abstract
Cancers can evade the host immune system by inducing upregulation of immune inhibitory signals. Anti-programmed cell death-1 (PD-1) monoclonal antibodies block these inhibitory signals allowing the host to mount an immune response against malignant cells. This class of drugs is active in solid tumours, where upregulation of cell-surface PD-1 ligand proteins is nearly uniform. Because lymphoma is a malignancy of immune system cells, the role of the PD-1 pathway in these neoplasms is more complex. However, early clinical trials using PD-1 inhibitors have shown significant clinical activity in various subtypes of relapsed lymphoma. In this Review, we assess the scientific literature on the role of the PD-1 pathway in lymphoma, the relevant clinical data for PD-1 inhibition, and future strategies for this next generation of anticancer agents.
Collapse
Affiliation(s)
- Eliza A Hawkes
- Department of Medical Oncology and Clinical Haematology, Olivia Newton John Cancer and Wellness Centre, Austin Hospital, Melbourne, VIC, Australia; Department of Medical Oncology, Eastern Health, Melbourne, VIC Australia; Monash University, Melbourne, VIC, Australia.
| | - Andrew Grigg
- Department of Medical Oncology and Clinical Haematology, Olivia Newton John Cancer and Wellness Centre, Austin Hospital, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Geoff Chong
- Department of Medical Oncology and Clinical Haematology, Olivia Newton John Cancer and Wellness Centre, Austin Hospital, Melbourne, VIC, Australia; Department of Oncology, Northern Hospital, Melbourne, VIC, Australia; Department of Medical Oncology, Ballarat Health Services, Ballarat, VIC, Australia
| |
Collapse
|
97
|
Li K, Zhang Q, Zhang Y, Yang J, Zheng J. T-cell-associated cellular immunotherapy for lung cancer. J Cancer Res Clin Oncol 2015; 141:1249-58. [PMID: 25381064 DOI: 10.1007/s00432-014-1867-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 10/27/2014] [Indexed: 12/29/2022]
Abstract
PURPOSE The aim of the present study was to discuss recent findings on the role of T cells in lung cancer to provide information on their potential application, especially in cellular immunotherapy. METHODS Data on the different types of T cells that are currently used for the treatment of lung cancer were obtained by searching the PUBMED database. RESULTS Cytotoxic T lymphocytes, natural killer T cells, γδ T cells, lymphokine-activated killer cells, tumor-infiltrating lymphocytes, cytokine-induced killer cells and gene-modified T cells were analyzed to determine the benefits and drawbacks of their application in the treatment of lung cancer. Advances in the study of their antitumor mechanisms and directions for future research were discussed. CONCLUSIONS T cells are critical for tumorigenesis and therefore important targets for the treatment of lung cancer. T-cell-associated cellular immunotherapy opens up a window of opportunity for the development of complementary methods to traditional lung cancer treatments, which warrants further investigation to improve the clinical outcomes of lung cancer patients.
Collapse
MESH Headings
- Cytokine-Induced Killer Cells/immunology
- Cytokine-Induced Killer Cells/transplantation
- Cytotoxicity, Immunologic/physiology
- Humans
- Immunotherapy, Adoptive/methods
- Killer Cells, Lymphokine-Activated/immunology
- Killer Cells, Lymphokine-Activated/transplantation
- Killer Cells, Natural/immunology
- Killer Cells, Natural/transplantation
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/transplantation
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
Collapse
Affiliation(s)
- Ke Li
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, West Huaihai Road 84#, Xuzhou, 221002, Jiangsu, China
| | | | | | | | | |
Collapse
|
98
|
Pennock GK, Chow LQM. The Evolving Role of Immune Checkpoint Inhibitors in Cancer Treatment. Oncologist 2015; 20:812-22. [PMID: 26069281 DOI: 10.1634/theoncologist.2014-0422] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 03/20/2015] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Traditional treatment modalities for advanced cancer (radiotherapy, chemotherapy, or targeted agents) act directly on tumors to inhibit or destroy them. Along with surgery, these modalities are predominantly palliative, with toxicity and only modest improvements in survival in patients with advanced solid tumors. Accordingly, long-term survival rates for most patients with advanced cancer remain low, thus there is a need for cancer treatments with favorable benefit and toxicity profiles that can potentially result in long-term survival. The immune system plays a critical role in the recognition and eradication of tumor cells ("immune surveillance"), and immunotherapies based on this concept have been used for decades with some success against a few tumor types; however, most immunotherapies were limited by a lack of either substantial efficacy or specificity, resulting in toxicity. We now have a greater understanding of the complex interactions between the immune system and tumors and have identified key molecules that govern these interactions. This information has revitalized the interest in immunotherapy as an evolving treatment modality using immunotherapeutics designed to overcome the mechanisms exploited by tumors to evade immune destruction. Immunotherapies have potentially complementary mechanisms of action that may allow them to be combined with other immunotherapeutics, chemotherapy, targeted therapy, or other traditional therapies. This review discusses the concepts and data behind immunotherapies, with a focus on the checkpoint inhibitors and their responses, toxicities, and potential for long-term survival, and explores promising single-agent and combination therapies in development. IMPLICATIONS FOR PRACTICE Immunotherapy is an evolving treatment approach based on the role of the immune system in eradicating cancer. An example of an immunotherapeutic is ipilimumab, an antibody that blocks cytotoxic T-lymphocyte antigen-4 (CTLA-4) to augment antitumor immune responses. Ipilimumab is approved for advanced melanoma and induced long-term survival in a proportion of patients. The programmed death-1 (PD-1) checkpoint inhibitors are promising immunotherapies with demonstrated sustained antitumor responses in several tumors. Because they harness the patient's own immune system, immunotherapies have the potential to be a powerful weapon against cancer.
Collapse
Affiliation(s)
- Gregory K Pennock
- Levine Cancer Institute, Charlotte, North Carolina, USA; University of Washington, Seattle, Washington, USA
| | - Laura Q M Chow
- Levine Cancer Institute, Charlotte, North Carolina, USA; University of Washington, Seattle, Washington, USA
| |
Collapse
|
99
|
Local and systemic XAGE-1b-specific immunity in patients with lung adenocarcinoma. Cancer Immunol Immunother 2015; 64:1109-21. [PMID: 26025564 PMCID: PMC4540777 DOI: 10.1007/s00262-015-1716-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 05/09/2015] [Indexed: 01/06/2023]
Abstract
XAGE-1b is a cancer/testis antigen aberrantly expressed in pulmonary adenocarcinoma. Systemic antibody and T cell responses have been demonstrated in adenocarcinoma patients, but so far, local antigen-specific immunity has not been reported. In this study, XAGE-1b expression by tumor cells as well as the presence of systemic and/or local XAGE-1b-specific immunity was assessed in peripheral blood, tumor tissue and tumor-draining lymph nodes of Caucasian patients with pulmonary adenocarcinoma. XAGE-1b protein expression was detected in 43.6% (17 of 39) of patients when at least two different parts of a resected tumor were assessed. In 20 patients, analysis of T cells isolated and expanded from the primary tumor and its draining lymph node demonstrated XAGE-1b-specific responses in two patients. XAGE-1b-specific immunoglobulin G antibodies were found in 3 of 40 patients. These three antibody-positive patients had also mounted a systemic T cell response to XAGE-1b, measured by proliferation, cytokine production and expression of T cell activation markers on peripheral blood mononuclear cells. The population of XAGE-1b-specific T cells comprised both CD4+ and CD8+ T cells secreting both type I and II cytokines. Epitope mapping showed that T cells predominantly targeted the N-terminal part of the XAGE-1b protein, while the B cell response was directed against the C-terminal domain. Our study for the first time provides evidence for the presence of XAGE-1b-specific T cells within adenocarcinoma tissue, which supports the concept that XAGE-1b acts as a genuine tumor antigen and, therefore, might form an attractive target for a vaccine-based approach of immunotherapy.
Collapse
|
100
|
Pan QZ, Tang Y, Wang QJ, Li YQ, Zhang L, Li XD, Zhao JJ, Weng DS, Liu Q, Huang LX, He J, Chen SP, Ke ML, Zeng YX, Xia JC. Adjuvant cellular immunotherapy in patients with resected primary non-small cell lung cancer. Oncoimmunology 2015; 4:e1038017. [PMID: 26405607 DOI: 10.1080/2162402x.2015.1038017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 03/30/2015] [Accepted: 04/01/2015] [Indexed: 01/04/2023] Open
Abstract
Postoperative non-small cell lung cancer (NSCLC) patients require adjuvant therapy to improve their prognosis. In this study, we investigated the efficacy of a sequential combination of autologous cellular immunotherapy (CIT) and chemotherapy for postoperative NSCLC. This retrospective study included 120 postoperative NSCLC patients: 60 cases received only chemotherapy; 33 cases received chemotherapy and sequential CIT with cytokine-induced killer (CIK) cells; and 27 cases received chemotherapy and sequential CIT with alternate CIK and natural killer (NK) cells. Survival analysis showed significantly higher overall survival rates in the CIT group compared with the control group. Overall survival was higher in patients who received CIT with alternate CIK and NK cells than those who received treatment with only CIK cells. Multivariate analysis showed that adjuvant CIT was an independent prognostic factor for overall survival of patients with NSCLC. In subgroup analyses, adjuvant CIT significantly improved the overall survival of patients with less than 60 y old and positive lymph node. In conclusions, these data indicate that adjuvant CIT, especially with alternate application of CIK and NK cells, is an effective therapeutic approach to prolong survival of patients with NSCLC, particularly for patients ≤60 y old with positive lymph nodes.
Collapse
Affiliation(s)
- Qiu-Zhong Pan
- Collaborative Innovation Center for Cancer Medicine; State Key Laboratory of Oncology in South China; Sun Yat-Sen University Cancer Center ; Guangzhou, China ; Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Yan Tang
- Collaborative Innovation Center for Cancer Medicine; State Key Laboratory of Oncology in South China; Sun Yat-Sen University Cancer Center ; Guangzhou, China ; Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Qi-Jing Wang
- Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Yong-Qiang Li
- Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Li Zhang
- Department of Medical Oncology; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Xiao-Dong Li
- Department of Thoracic Surgery; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Jing-Jing Zhao
- Collaborative Innovation Center for Cancer Medicine; State Key Laboratory of Oncology in South China; Sun Yat-Sen University Cancer Center ; Guangzhou, China ; Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - De-Sheng Weng
- Collaborative Innovation Center for Cancer Medicine; State Key Laboratory of Oncology in South China; Sun Yat-Sen University Cancer Center ; Guangzhou, China ; Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Qing Liu
- Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Li-Xi Huang
- Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Jia He
- Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Shi-Ping Chen
- Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Miao-La Ke
- Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Yi-Xin Zeng
- Collaborative Innovation Center for Cancer Medicine; State Key Laboratory of Oncology in South China; Sun Yat-Sen University Cancer Center ; Guangzhou, China ; Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| | - Jian-Chuan Xia
- Collaborative Innovation Center for Cancer Medicine; State Key Laboratory of Oncology in South China; Sun Yat-Sen University Cancer Center ; Guangzhou, China ; Department of Biotherapy; Sun Yat-Sen University Cancer Center ; Guangzhou, China
| |
Collapse
|