1
|
Nel AE, Pavlisko EN, Roggli VL. The Interplay Between the Immune System, Tumor Suppressor Genes, and Immune Senescence in Mesothelioma Development and Response to Immunotherapy. J Thorac Oncol 2024; 19:551-564. [PMID: 38000500 DOI: 10.1016/j.jtho.2023.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/30/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023]
Abstract
Despite efforts to ban asbestos mining and manufacturing, mesothelioma deaths in the United States have remained stable at approximately 2500 cases annually. This trend is not unique to the United States but is also a global phenomenon, associated with increased aging of populations worldwide. Although geoeconomic factors such as lack of regulations and continued asbestos manufacturing in resource-poor countries play a role, it is essential to consider biological factors such as immune senescence and increased genetic instability associated with aging. Recognizing that mesothelioma shares genetic instability and immune system effects with other age-related cancers is crucial because the impact of aging on mesothelioma is frequently assessed in the context of disease latency after asbestos exposure. Nevertheless, the long latency period, often cited as a reason for mesothelioma's elderly predominance, should not overshadow the shared mechanisms. This communication focuses on the role of immune surveillance in mesothelioma, particularly exploring the impact of immune escape resulting from altered TSG function during aging, contributing to the phylogenetic development of gene mutations and mesothelioma oncogenesis. The interplay between the immune system, TSGs, and aging not only shapes the immune landscape in mesothelioma but also contributes to the development of heterogeneous tumor microenvironments, significantly influencing responses to immunotherapy approaches and survival rates. By understanding the complex interplay between aging, TSG decline, and immune senescence, health care professionals can pave the way for more effective and personalized immunotherapies, ultimately offering hope for better outcomes in the fight against mesothelioma.
Collapse
Affiliation(s)
- Andre E Nel
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California; Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.
| | | | - Victor L Roggli
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
2
|
Zhou Y, Ju H, Hu Y, Li T, Chen Z, Si Y, Sun X, Shi Y, Fang H. Tregs dysfunction aggravates postoperative cognitive impairment in aged mice. J Neuroinflammation 2023; 20:75. [PMID: 36932450 PMCID: PMC10022212 DOI: 10.1186/s12974-023-02760-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/09/2023] [Indexed: 03/19/2023] Open
Abstract
OBJECTIVES Enhanced neuroinflammation is an important mechanism underlying perioperative neurocognitive disorders. Regulatory T cells (Tregs) play a crucial role in regulating systemic immune responses. The present study was aimed to investigate the participation of Tregs in the development of postoperative cognitive dysfunction (POCD). METHODS Surgery-associated neurocognitive disorder was induced in 18-month-old mice subjected to internal fixation of tibial fracture. Morris water maze was used to examine mice cognitive function. Splenic Tregs were collected for RNA sequencing and flow cytometry. Levels of inflammatory factors in the circulation and hippocampus were measured by enzyme-linked immunosorbent assay. Protein presences of tight junction proteins were detected by immunofluorescence. RESULTS Surgery of internal fixation of tibial fracture induced cognitive impairment in aged mice, accompanied by elevated plasma levels of inflammatory factors and increased circulating Tregs. Transfusion of Tregs from young mice partially restored the structure of the blood-brain barrier and alleviated POCD in aged mice. Compared with young Tregs, differentially expressed genes in aged Tregs were enriched in tumor necrosis factor (TNF) signaling pathway and cytokine-cytokine receptor interaction. Flow cytometry revealed that aged Tregs had blunted functions under basal and stimulated conditions. Blockade of the CD25 epitope protected the blood-brain barrier structure, reduced TNF-α levels in the hippocampus, and improved surgery-associated cognition in aged mice. CONCLUSIONS Blocking peripheral regulatory T cells improves surgery-induced cognitive function in aged mice. Therefore, aged Tregs play an essential role in the occurrence of POCD.
Collapse
Affiliation(s)
- Yile Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huihui Ju
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tingting Li
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhouyi Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Si
- Department of Anesthesiology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xia Sun
- Department of Anesthesiology, Shanghai Cancer Center, Fudan University, Shanghai, China.
| | - Yi Shi
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Anesthesiology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Clopton B, Long W, Santos M, Asarian A, Genato R, Xiao P. Sarcomatoid mesothelioma: unusual findings and literature review. J Surg Case Rep 2022; 2022:rjac512. [PMID: 36415726 PMCID: PMC9675762 DOI: 10.1093/jscr/rjac512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 10/16/2022] [Indexed: 10/10/2023] Open
Abstract
Sarcomatoid mesothelioma is an aggressive disease secondary to its propensity to undergo rapid growth, show inconsistent expression of tumor markers and invade surrounding tissues. Therefore, there are numerous obstacles that clinical researchers face as they look for new methods to diagnose and treat the malignancy. We present a case of sarcomatoid mesothelioma, originally thought to be metastasis from renal cell carcinoma.
Collapse
Affiliation(s)
- Brittni Clopton
- American University of the Caribbean School of Medicine, Cupecoy, St. Maarten
| | - Winnie Long
- Department of Surgery, the Brooklyn Hospital Center, Icahn School of Medicine, Brooklyn, NY, USA
| | - Monica Santos
- Department of Surgery, the Brooklyn Hospital Center, Icahn School of Medicine, Brooklyn, NY, USA
| | - Armand Asarian
- Department of Surgery, the Brooklyn Hospital Center, Icahn School of Medicine, Brooklyn, NY, USA
| | - Romulo Genato
- Department of Surgery, the Brooklyn Hospital Center, Icahn School of Medicine, Brooklyn, NY, USA
| | - Philip Xiao
- Department of Pathology, the Brooklyn Hospital Center, Icahn School of Medicine, Brooklyn, NY, USA
| |
Collapse
|
4
|
Regulatory T Cell Depletion Using a CRISPR Fc-Optimized CD25 Antibody. Int J Mol Sci 2022; 23:ijms23158707. [PMID: 35955841 PMCID: PMC9369266 DOI: 10.3390/ijms23158707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Regulatory T cells (Tregs) are major drivers behind immunosuppressive mechanisms and present a major hurdle for cancer therapy. Tregs are characterized by a high expression of CD25, which is a potentially valuable target for Treg depletion to alleviate immune suppression. The preclinical anti-CD25 (αCD25) antibody, clone PC-61, has met with modest anti-tumor activity due to its capacity to clear Tregs from the circulation and lymph nodes, but not those that reside in the tumor. The optimization of the Fc domain of this antibody clone has been shown to enhance the intratumoral Treg depletion capacity. Here, we generated a stable cell line that produced optimized recombinant Treg-depleting antibodies. A genome engineering strategy in which CRISPR-Cas9 was combined with homology-directed repair (CRISPR-HDR) was utilized to optimize the Fc domain of the hybridoma PC-61 for effector functions by switching it from its original rat IgG1 to a mouse IgG2a isotype. In a syngeneic tumor mouse model, the resulting αCD25-m2a (mouse IgG2a isotype) antibody mediated the effective depletion of tumor-resident Tregs, leading to a high effector T cell (Teff) to Treg ratio. Moreover, a combination of αCD25-m2a and an αPD-L1 treatment augmented tumor eradication in mice, demonstrating the potential for αCD25 as a cancer immunotherapy.
Collapse
|
5
|
Cheng B, Ding K, Chen P, Ji J, Luo T, Guo X, Qiu W, Ma C, Meng X, Wang J, Yu J, Liu Y. Anti-PD-L1/TGF-βR fusion protein (SHR-1701) overcomes disrupted lymphocyte recovery-induced resistance to PD-1/PD-L1 inhibitors in lung cancer. Cancer Commun (Lond) 2022; 42:17-36. [PMID: 34981670 PMCID: PMC8753312 DOI: 10.1002/cac2.12244] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/23/2021] [Accepted: 11/25/2021] [Indexed: 12/22/2022] Open
Abstract
Background Second‐generation programmed cell death‐protein 1/programmed death‐ligand 1 (PD‐1/PD‐L1) inhibitors, such as bintrafusp alfa (M7824), SHR‐1701, and YM101, have been developed to simultaneously block PD‐1/PD‐L1 and transforming growth factor‐beta/transforming growth factor‐beta receptor (TGF‐β/TGF‐βR). Consequently, it is necessary to identify predictive factors of lung cancer patients who are not only resistant to PD‐1/PD‐L1 inhibitors but also sensitive to bifunctional drugs. The purpose of this study was to search for such predictors. Methods Multivariable Cox regression was used to study the association between the clinical outcome of treatment with PD‐1/PD‐L1 inhibitors and lymphocyte recovery after lymphopenia in lung cancer patients. Murine CMT167 lung cancer cells were engineered to express the firefly luciferase gene and implanted orthotopically in the lung of syngeneic mice. Bioluminescence imaging, flow cytometry, and immunohistochemistry were employed to determine response to immunotherapy and function of tumor‐infiltrating immune cells. Results For lung cancer patients treated with anti‐PD‐1/PD‐L1 antibodies, poor lymphocyte recovery was associated with a shorter progression‐free survival (PFS; P < 0.001), an accumulation of regulatory T cells (Tregs), and an elimination of CD8+ T cells in the peripheral blood. Levels of CD8+ T cells and Treg cells were also imbalanced in the tumors and peripheral immune organs of mice with poor lymphocyte recovery after chemotherapy. Moreover, these mice failed to respond to anti‐PD‐1 antibodies but remained sensitive to the anti‐PD‐L1/TGF‐βR fusion protein (SHR‐1701). Consistently, SHR‐1701 but not anti‐PD‐1 antibodies, markedly enhanced IFN‐γ production and Ki‐67 expression in peripheral CD8+ T cells from patients with impaired lymphocyte recovery. Conclusions Lung cancer patients with poor lymphocyte recovery and suffering from persistent lymphopenia after previous chemotherapy are resistant to anti‐PD‐1/PD‐L1 antibodies but might be sensitive to second‐generation agents such as SHR‐1701.
Collapse
Affiliation(s)
- Bo Cheng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China.,Shandong Key Laboratory of Brain Functional Remodeling, Jinan, Shandong, 250012, P. R. China
| | - Kaikai Ding
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China.,Shandong Key Laboratory of Brain Functional Remodeling, Jinan, Shandong, 250012, P. R. China
| | - Pengxiang Chen
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Jianxiong Ji
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China.,Shandong Key Laboratory of Brain Functional Remodeling, Jinan, Shandong, 250012, P. R. China.,Department of Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, P. R. China
| | - Tao Luo
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China.,Shandong Key Laboratory of Brain Functional Remodeling, Jinan, Shandong, 250012, P. R. China
| | - Xiaofan Guo
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China.,Department of Neurology, Loma Linda University Health, Loma Linda, CA, 92354, USA
| | - Wei Qiu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China.,Shandong Key Laboratory of Brain Functional Remodeling, Jinan, Shandong, 250012, P. R. China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Medicine, Jinan, Shandong, 250012, P. R. China
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China.,Shandong Key Laboratory of Brain Functional Remodeling, Jinan, Shandong, 250012, P. R. China.,Department of Biomedicine, University of Bergen, Bergen, 5009, Norway
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Yuan Liu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China.,Shandong Key Laboratory of Brain Functional Remodeling, Jinan, Shandong, 250012, P. R. China
| |
Collapse
|
6
|
Harber J, Kamata T, Pritchard C, Fennell D. Matter of TIME: the tumor-immune microenvironment of mesothelioma and implications for checkpoint blockade efficacy. J Immunother Cancer 2021; 9:e003032. [PMID: 34518291 PMCID: PMC8438820 DOI: 10.1136/jitc-2021-003032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an incurable cancer with a dismal prognosis and few effective treatment options. Nonetheless, recent positive phase III trial results for immune checkpoint blockade (ICB) in MPM herald a new dawn in the fight to advance effective treatments for this cancer. Tumor mutation burden (TMB) has been widely reported to predict ICB in other cancers, but MPM is considered a low-TMB tumor. Similarly, tumor programmed death-ligand 1 (PD-L1) expression has not been proven predictive in phase III clinical trials in MPM. Consequently, the precise mechanisms that determine response to immunotherapy in this cancer remain unknown. The present review therefore aimed to synthesize our current understanding of the tumor immune microenvironment in MPM and reflects on how specific cellular features might impact immunotherapy responses or lead to resistance. This approach will inform stratified approaches to therapy and advance immunotherapy combinations in MPM to improve clinical outcomes further.
Collapse
Affiliation(s)
- James Harber
- Cancer Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| | - Tamihiro Kamata
- Cancer Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| | - Catrin Pritchard
- Cancer Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| | - Dean Fennell
- Cancer Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| |
Collapse
|
7
|
Rajaratinam H, Rasudin NS, Al Astani TAD, Mokhtar NF, Yahya MM, Wan Zain WZ, Asma-Abdullah N, Mohd Fuad WE. Breast cancer therapy affects the expression of antineonatal Nav1.5 antibodies in the serum of patients with breast cancer. Oncol Lett 2021; 21:108. [PMID: 33376541 PMCID: PMC7751336 DOI: 10.3892/ol.2020.12369] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/05/2020] [Indexed: 11/06/2022] Open
Abstract
Neonatal Nav1.5 (nNav1.5) is the alternative splice variant of Nav1.5 and it has been widely associated with the progression of breast cancer. The immunological context of nNav1.5 with respect to breast cancer metastases remains unexplored. The presence of antibodies against nNav1.5 may highlight the immunogenicity of nNav1.5. Hence, the aim of the present study was to detect the presence of antineonatal Nav1.5 antibodies (antinNav1.5-Ab) in the serum of patients with breast cancer and to elucidate the effects of breast cancer therapy on its expression. A total of 32 healthy female volunteers and 64 patients with breast cancer were randomly recruited into the present study as the control and breast cancer group, respectively. Patients with breast cancer were divided equally based on their pre- and ongoing-treatment status. Serum samples were tested with in-house indirect enzyme-linked immunosorbent assay (ELISA) to detect antinNav1.5-Ab, CD25 (T regulatory cell marker) using an ELISA kit and Luminex assay to detect the expression of metastasis-associated cytokines, such as vascular endothelial growth factor (VEGF), interleukin (IL)-6, IL-10, IL-8, chemokine (C-C motif) ligand 2 and tumor necrosis factor-alpha (TNF-α) The mean difference in the expression of antinNav1.5-Ab among the three groups (control, pretreatment and ongoing-treatment) was significant (P=0.0005) and the pretreatment breast cancer group exhibited the highest expression. The concentration of CD25 was highest in the pretreatment breast cancer group compared with the control and ongoing-treatment groups. There was a significant positive correlation between antinNav1.5-Ab and IL-6 in the pretreatment group (r=0.7260; P=0.0210) and a significant negative correlation between antinNav1.5-Ab and VEGF in the ongoing-treatment group (r=-0.842; P-value=0.0040). The high expression of antinNav1.5-Ab in the pretreatment group was in accordance with the uninterrupted presence of metastasis and highlighted the immunogenicity of nNav1.5 whereas the low expression of antinNav1.5-Ab in the ongoing-treatment group reflected the efficacy of breast cancer therapy in eliminating metastases. The augmented manifestation of T regulatory cells in the pretreatment group highlighted the functional role of nNav1.5 in promoting metastasis. The parallel expression of antinNav1.5-Ab with the imbalanced expression of cytokines promoting metastasis (IL-8, IL-6 and TNF-α) and cytokines that prevent metastasis (IL-10) indicated the role of nNav1.5 in breast cancer growth. The expression of antinNav1.5-Ab in accordance to the metastatic microenvironment indicates the immunogenicity of the protein and highlights the influence of breast cancer therapy on its expression level.
Collapse
Affiliation(s)
- Harishini Rajaratinam
- School of Health Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Nur Syahmina Rasudin
- School of Health Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Tengku Ahmad Damitri Al Astani
- Department of Chemical Pathology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
- Breast Cancer Awareness and Research (BestARi) Unit, Hospital Universiti Sains Malaysia (HUSM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine (INFORMM), Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Maya Mazuwin Yahya
- Department of Surgery, School of Medical Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Wan Zainira Wan Zain
- Department of Surgery, School of Medical Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Nurul Asma-Abdullah
- School of Health Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| | - Wan Ezumi Mohd Fuad
- School of Health Sciences, Health Campus, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan 16150, Malaysia
| |
Collapse
|
8
|
Duong BTV, Wu L, Green BJ, Bavaghar-Zaeimi F, Wang Z, Labib M, Zhou Y, Cantu FJP, Jeganathan T, Popescu S, Pantea J, de Perrot M, Kelley SO. A liquid biopsy for detecting circulating mesothelial precursor cells: A new biomarker for diagnosis and prognosis in mesothelioma. EBioMedicine 2020; 61:103031. [PMID: 33045471 PMCID: PMC7553233 DOI: 10.1016/j.ebiom.2020.103031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Malignant pleural mesothelioma (MPM) is an aggressive cancer related to asbestos exposure. Early diagnosis is challenging due to generic symptoms and a lack of biomarkers. We previously demonstrated that mesothelial precursor cells (MPC) characterized by mesothelin (MSLN)+CD90+CD34+ could be implicated in the development of mesothelioma after asbestos exposure. Here, we aimed to determine the clinical significance of detecting MPC in blood for early-stage diagnosis and prognosis of mesothelioma. METHODS Due to the rarity of MPC in blood, it is challenging to identify this cell population using conventional techniques. Hence, we have developed a microfluidic liquid biopsy platform called MesoFind that utilizes an immunomagnetic, mesothelin capture strategy coupled with immunofluorescence to identify rare populations of cells at high sensitivity and precision. To validate our technique, we compared this approach to flow cytometry for the detection of MPC in murine blood and lavage samples. Upon successful validation of the murine samples, we then proceeded to examine circulating MPC in 23 patients with MPM, 23 asbestos-exposed individuals (ASB), and 10 healthy donors (HD) to evaluate their prognostic and diagnostic value. FINDING MPC were successfully detected in the blood of murine samples using MesoFind but were undetectable with flow cytometry. Circulating MPC were significantly higher in patients with epithelioid MPM compared to HD and ASB. The MPC subpopulation, MSLN+ and CD90+, were upregulated in ASB compared to HD suggesting an early role in pleural damage from asbestos. The MPC subpopulation, MSLN+ and CD34+, in contrast, were detected in advanced MPM and associated with markers of poor prognosis, suggesting a predominant role during cancer progression. INTERPRETATION The identification of circulating MPC presents an attractive solution for screening and early diagnosis of epithelioid mesothelioma. The presence of different subtypes of MPC have a prognostic value that could be of assistance with clinical decisions in patients with MPM. FUNDING Princess Margaret Hospital Foundation Mesothelioma Research Fund, Toronto General & Western Hospital Foundation.
Collapse
Affiliation(s)
- Bill T V Duong
- Department of Chemistry, University of Toronto, 80 St George St., Toronto, Ontario M5S 3H6, Canada
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, 101 College St., Toronto, Ontario M5G 1L7, Canada
| | - Brenda J Green
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario M5S 3G9, Canada
| | - Fatemeh Bavaghar-Zaeimi
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, 101 College St., Toronto, Ontario M5G 1L7, Canada; Division of Thoracic Surgery, Toronto General Hospital and Princess Margaret Cancer Centre, University Health Network, 200 Elizabeth St., Toronto, Ontario M5G 2C4, Canada
| | - Zongjie Wang
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario M5S 3G9, Canada; The Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, 10 King's College Rd., Toronto, Ontario M5S 3G4, Canada
| | - Mahmoud Labib
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Yuxiao Zhou
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Fernando J P Cantu
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Thurgaa Jeganathan
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Sandra Popescu
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Jennifer Pantea
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, 101 College St., Toronto, Ontario M5G 1L7, Canada; Division of Thoracic Surgery, Toronto General Hospital and Princess Margaret Cancer Centre, University Health Network, 200 Elizabeth St., Toronto, Ontario M5G 2C4, Canada; Department of Immunology, University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada.
| | - Shana O Kelley
- Department of Chemistry, University of Toronto, 80 St George St., Toronto, Ontario M5S 3H6, Canada; Institute for Biomaterials and Biomedical Engineering, University of Toronto, 164 College St., Toronto, Ontario M5S 3G9, Canada; Department of Pharmaceutical Sciences, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada; Department of Biochemistry, University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada.
| |
Collapse
|
9
|
Otsuka K, Mitsuhashi A, Goto H, Hanibuchi M, Koyama K, Ogawa H, Ogino H, Saijo A, Kozai H, Yoneda H, Tobiume M, Kishuku M, Ishizawa K, Nishioka Y. Anti-PD-1 antibody combined with chemotherapy suppresses the growth of mesothelioma by reducing myeloid-derived suppressor cells. Lung Cancer 2020; 146:86-96. [PMID: 32526602 DOI: 10.1016/j.lungcan.2020.05.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND The combination of anti-PD-1/PD-L1 antibody with chemotherapy has been approved for the first-line therapy of lung cancer. However, the effects against malignant mesothelioma (MPM) and the immunological mechanisms by which chemotherapy enhances the effect of targeting PD-1/PD-L1 in MPM are poorly understood. MATERIALS AND METHODS We utilized syngeneic mouse models of MPM and lung cancer and assessed the therapeutic effects of anti-PD-1 antibody and its combination with cisplatin (CDDP) and pemetrexed (PEM). An immunological analysis of tumor-infiltrating cells was performed with immunohistochemistry. RESULTS We observed significant therapeutic effects of anti-PD-1 antibody against MPM. Although the effect was associated with CD8+ and CD4+ T cells in tumors, the number of Foxp3+ cells was not reduced but rather increased. Consequently, combination with CDDP/PEM significantly enhanced the antitumor effects of anti-PD-1 antibody by decreasing numbers of intratumoral myeloid-derived suppressor cells (MDSCs) and vessels probably through suppression of VEGF expression by CDDP + PEM. CONCLUSIONS The combination of anti-PD-1 antibody with CDDP + PEM may be a promising therapy for MPM via inhibiting the accumulation of MDSCs and vessels in tumors.
Collapse
Affiliation(s)
- Kenji Otsuka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Atsushi Mitsuhashi
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hisatsugu Goto
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masaki Hanibuchi
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Kazuya Koyama
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hirohisa Ogawa
- Department of Pathology and Laboratory Medicine, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hirokazu Ogino
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Atsuro Saijo
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroyuki Kozai
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroto Yoneda
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Makoto Tobiume
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masatoshi Kishuku
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.
| |
Collapse
|
10
|
Klampatsa A, O'Brien SM, Thompson JC, Rao AS, Stadanlick JE, Martinez MC, Liousia M, Cantu E, Cengel K, Moon EK, Singhal S, Eruslanov EB, Albelda SM. Phenotypic and functional analysis of malignant mesothelioma tumor-infiltrating lymphocytes. Oncoimmunology 2019; 8:e1638211. [PMID: 31428531 DOI: 10.1080/2162402x.2019.1638211] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/27/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Given the growing interest and promising preliminary results of immunotherapy in malignant pleural mesothelioma (MPM), it has become important to more fully understand the immune landscape in this tumor. This may be especially relevant in deciding who might benefit most from checkpoint blockade or agonist antibody therapy. Since the phenotype of tumor infiltrating lymphocytes (TILs) in MPM has not been fully described and their function has not been carefully assessed, we collected fresh tumor and blood from 22 patients undergoing surgical resection and analysed single cell suspensions by flow cytometry. The functionality of TILs was assessed by measurement of cytokine expression (IFN-γ) following overnight stimulation ex vivo. Results showed low numbers of CD8+ TILs whose function was either moderately or severely suppressed. The degree of TIL hypofunction did not correlate with the presence of co-existing macrophages or neutrophils, nor with expression of the inhibitory receptors PD-1, CD39 and CTLA-4. Hypofunction was associated with higher numbers of CD4 regulatory T cells (Tregs) and with expression of the inhibitory receptor TIGIT. On the other hand, presence of tissue-resident memory (Trm) cells and expression of TIM-3 on CD8+ cells were positively associated with cytokine production. However, Trm function was partially suppressed when the transcription factor Eomesodermin (Eomes) was co-expressed. Understanding the function of TILs in malignant mesothelioma may have clinical implications for immunotherapy, especially in choosing the best immunotherapy targets. Our data suggests that Treg cell blocking agents or TIGIT inhibitor antibodies might be especially valuable in these patients.
Collapse
Affiliation(s)
- Astero Klampatsa
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Shaun M O'Brien
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey C Thompson
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Abhishek S Rao
- Division of Thoracic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jason E Stadanlick
- Division of Thoracic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Marina C Martinez
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Maria Liousia
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Cantu
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Keith Cengel
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Edmund K Moon
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Evgeniy B Eruslanov
- Division of Thoracic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Steven M Albelda
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Yang X, Zhao J, Duan S, Hou X, Li X, Hu Z, Tang Z, Mo F, Lu X. Enhanced cytotoxic T lymphocytes recruitment targeting tumor vasculatures by endoglin aptamer and IP-10 plasmid presenting liposome-based nanocarriers. Theranostics 2019; 9:4066-4083. [PMID: 31281532 PMCID: PMC6592167 DOI: 10.7150/thno.33383] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/24/2019] [Indexed: 12/16/2022] Open
Abstract
Background: Adequate recruitment of highly active tumor antigen-specific cytotoxic T lymphocytes (CTLs) remains a major challenge in cancer immunotherapy. Objective: To construct liposome (LP)-based nanocapsules with surface endoglin aptamer (ENG-Apt) encapsulating mouse interferon-inducible protein-10 (mIP-10), with the ability to target mouse tumor vascular endothelial cells (mTECs) and enhance CTLs targeting and recruitment to the tumor vasculature. Methods: ENG-Apt/mIP-10-LP nanocapsules were prepared by grafting DSPE-PEG2000-ENG-Apt on the surface of liposomes containing mIP-10 plasmids, characterized and assessed for the cell binding specificity in vitro. The tumor-targeting ability of ENG-Apt/mIP-10-LP nanocapsules was evaluated in vivo. The anti-tumor efficacy of ENG-Apt/mIP-10-LP nanocapsules treatment, as well as the combination treatment of ENG-Apt/mIP-10-LP nanocapsules and adoptive TRP2CD8+ T cells, were both tested in melanoma-bearing mice, by evaluation of the tumor volume and the mouse survival time. To discuss the anti-tumoral mechanism of ENG-Apt/mIP-10-LP nanocapsules-based therapies, IFN-γ secretion, proportion of TRP2CD8+ T cells among TILs, MDSCs in the tumor microenvironment and Tregs in the spleen, were determined after the treatments. Proliferation and apoptosis of tumor cells, and tumor angiogenesis were also assessed. Results: The prepared ENG-Apt/mIP-10-LP nanocapsules possess an adequate nanometric size, good stability, high specificity to mTECs and tumor sites, along with the ability to induce mIP-10 expression in vitro and in vivo. Treatment of ENG-Apt/mIP-10-LP nanocapsules demonstrated CTLs enrichment into the tumor site, which inhibited tumor cell proliferation and angiogenesis, as well as promoted tumor-cell apoptosis, leading to a decrease in tumor progression and prolonged survival time in melanoma tumor-bearing mice. In addition, the proportion of MDSCs and Tregs was found to decrease. The combination of ENG-Apt/mIP-10-LP nanocapsules with adoptive TRP2CD8+ T cells, showed stronger abilities in inhibiting tumor growth and increasing animal survival time, thereby displayed an enhanced anti-melanoma tumor efficacy, due to the recruitment of both endogenous CD8+ T cells and exogenous TRP2CD8+ T cells in vivo. Conclusion: ENG-Apt/mIP-10-LP nanocapsules could enhance the recruitment of both endogenous and exogenous CTLs specifically targeting melanoma tumor vasculatures and exert anti-tumoral effect, therefore provides a potentially novel strategy for tumor immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiaoling Lu
- Nanobody Research Center/School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| |
Collapse
|
12
|
Englinger B, Pirker C, Heffeter P, Terenzi A, Kowol CR, Keppler BK, Berger W. Metal Drugs and the Anticancer Immune Response. Chem Rev 2018; 119:1519-1624. [DOI: 10.1021/acs.chemrev.8b00396] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Alessio Terenzi
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Wu L, Blum W, Zhu CQ, Yun Z, Pecze L, Kohno M, Chan ML, Zhao Y, Felley-Bosco E, Schwaller B, de Perrot M. Putative cancer stem cells may be the key target to inhibit cancer cell repopulation between the intervals of chemoradiation in murine mesothelioma. BMC Cancer 2018; 18:471. [PMID: 29699510 PMCID: PMC5921988 DOI: 10.1186/s12885-018-4354-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/09/2018] [Indexed: 12/29/2022] Open
Abstract
Background Cancer cell repopulation during chemotherapy or radiotherapy is a major factor limiting the efficacy of treatment. Cancer stem cells (CSC) may play critical roles during this process. We aim to demonstrate the role of mesothelioma stem cells (MSC) in treatment failure and eventually to design specific target therapies against MSC to improve the efficacy of treatment in malignant mesothelioma. Methods Murine mesothelioma AB12 and RN5 cells were used to compare tumorigenicity in mice. The expression of CSC-associated genes was evaluated by quantitative real-time PCR in both cell lines treated with chemo-radiation. Stemness properties of MSC-enriched RN5-EOS-Puro2 cells were characterized with flow cytometry and immunostaining. A MSC-specific gene profile was screened by microarray assay and confirmed thereafter. Gene Ontology analysis of the selected genes was performed by GOMiner. Results Tumor growth delay of murine mesothelioma AB12 cells was achieved after each cycle of cisplatin treatment, however, tumors grew back rapidly due to cancer cell repopulation between courses of chemotherapy. Strikingly, a 10-times lower number of irradiated cells in both cell lines led to a similar tumor incidence and growth rate as with untreated cells. The expression of CSC-associated genes such as CD24, CD133, CD90 and uPAR was dramatically up-regulated, while others did not change significantly after chemoradiation. Highly enriched MSC after selection with puromycin displayed an increasing GFP-positive population and showed typical properties of stemness. Comparatively, the proportion of MSC significantly increased after RN5-EOS parental cells were treated with either chemotherapy, γ-ray radiation, or a combination of the two, while MSC showed more resistance to the above treatments. A group of identified genes are most likely MSC-specific, and major pathways related to regulation of cell growth or apoptosis are involved. Upregulation of the gene transcripts Tnfsf18, Serpinb9b, Ly6a, and Nppb were confirmed. Conclusion Putative MSC possess the property of stemness showing more resistance to chemoradiation, suggesting that MSC may play critical roles in cancer cell repopulation. Further identification of selected genes may be used to design novel target therapies against MSC, so as to eliminate cancer cell repopulation in mesothelioma. Electronic supplementary material The online version of this article (10.1186/s12885-018-4354-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Licun Wu
- Division of Thoracic Surgery, Latner Thoracic Surgery Laboratories, University Health Network, Toronto, ON, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Walter Blum
- Department of Medicine, Unit of Anatomy, University of Fribourg, CH-1700, Fribourg, Switzerland.,INSERM, U1162, Génomique Fonctionnelle des Tumeurs Solides, 27 rue Juliette Dodu, 75010, Paris, France
| | - Chang-Qi Zhu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Zhihong Yun
- Division of Thoracic Surgery, Latner Thoracic Surgery Laboratories, University Health Network, Toronto, ON, Canada
| | - Laszlo Pecze
- Department of Medicine, Unit of Anatomy, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Mikihiro Kohno
- Division of Thoracic Surgery, Latner Thoracic Surgery Laboratories, University Health Network, Toronto, ON, Canada
| | - Mei-Lin Chan
- Division of Thoracic Surgery, Latner Thoracic Surgery Laboratories, University Health Network, Toronto, ON, Canada
| | - Yidan Zhao
- Division of Thoracic Surgery, Latner Thoracic Surgery Laboratories, University Health Network, Toronto, ON, Canada
| | - Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, University Hospital Zurich, University of Zurich, 8044, Zürich, Switzerland
| | - Beat Schwaller
- Department of Medicine, Unit of Anatomy, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Marc de Perrot
- Division of Thoracic Surgery, Latner Thoracic Surgery Laboratories, University Health Network, Toronto, ON, Canada. .,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Division of Thoracic Surgery, Toronto Mesothelioma Research Program, Toronto General Hospital, 9N-961, 200 Elizabeth St, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
14
|
Rehrauer H, Wu L, Blum W, Pecze L, Henzi T, Serre-Beinier V, Aquino C, Vrugt B, de Perrot M, Schwaller B, Felley-Bosco E. How asbestos drives the tissue towards tumors: YAP activation, macrophage and mesothelial precursor recruitment, RNA editing, and somatic mutations. Oncogene 2018; 37:2645-2659. [PMID: 29507420 PMCID: PMC5955862 DOI: 10.1038/s41388-018-0153-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/11/2017] [Accepted: 12/30/2017] [Indexed: 12/11/2022]
Abstract
Chronic exposure to intraperitoneal asbestos triggered a marked response in the mesothelium well before tumor development. Macrophages, mesothelial precursor cells, cytokines, and growth factors accumulated in the peritoneal lavage. Transcriptome profiling revealed YAP/TAZ activation in inflamed mesothelium with further activation in tumors, paralleled by increased levels of cells with nuclear YAP/TAZ. Arg1 was one of the highest upregulated genes in inflamed tissue and tumor. Inflamed tissue showed increased levels of single-nucleotide variations, with an RNA-editing signature, which were even higher in the tumor samples. Subcutaneous injection of asbestos-treated, but tumor-free mice with syngeneic mesothelioma tumor cells resulted in a significantly higher incidence of tumor growth when compared to naïve mice supporting the role of the environment in tumor progression.
Collapse
Affiliation(s)
- Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057, Zurich, Switzerland
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital University Health Network, University of Toronto, Toronto, ON, Canada
| | - Walter Blum
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | - Lazslo Pecze
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | - Thomas Henzi
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | | | - Catherine Aquino
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057, Zurich, Switzerland
| | - Bart Vrugt
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital University Health Network, University of Toronto, Toronto, ON, Canada
| | - Beat Schwaller
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | - Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, Lungen- und Thoraxonkologie Zentrum, University Hospital Zürich, Sternwartstrasse 14, 8091, Zurich, Switzerland.
| |
Collapse
|
15
|
Wu L, de Perrot M. Radio-immunotherapy and chemo-immunotherapy as a novel treatment paradigm in malignant pleural mesothelioma. Transl Lung Cancer Res 2017; 6:325-334. [PMID: 28713677 DOI: 10.21037/tlcr.2017.06.03] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive neoplasm with poor outcome. Novel radical radiation techniques using intensity modulated radiation therapy (IMRT) have become an important component of therapy in mesothelioma. Immunotherapy also provides new therapeutic options. However, how best to integrate immunotherapy with standard therapy such as radiation, chemotherapy and surgery remains unknown. A change of paradigm from adjuvant normofractionation to induction accelerated hypofractionated hemithoracic radiation could provide a platform to combine immunotherapy due to the potential benefit of short course high dose radiation on the immune system. Immunotherapy can also be combined with chemotherapy. Although chemotherapy is generally considered immunosuppressive, some chemotherapeutic agents do induce cell death that can be immunogenic and stimulate a specific immune response against the tumor. Immunotherapy could also be used in between cycles of chemotherapy to limit tumor cell repopulation and optimize the results of both treatments. The integration of immunotherapy into a multimodality approach is opening new avenue of treatment for mesothelioma.
Collapse
Affiliation(s)
- Licun Wu
- Toronto Mesothelioma Research Program, Toronto General Hospital and Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Marc de Perrot
- Toronto Mesothelioma Research Program, Toronto General Hospital and Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
16
|
Marcq E, Siozopoulou V, De Waele J, van Audenaerde J, Zwaenepoel K, Santermans E, Hens N, Pauwels P, van Meerbeeck JP, Smits ELJ. Prognostic and predictive aspects of the tumor immune microenvironment and immune checkpoints in malignant pleural mesothelioma. Oncoimmunology 2016; 6:e1261241. [PMID: 28197385 DOI: 10.1080/2162402x.2016.1261241] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/27/2016] [Accepted: 11/10/2016] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with a poor prognosis and an increasing incidence, for which novel therapeutic strategies are urgently required. Since the immune system has been described to play a presumed role in the protection against MPM, characterization of its tumor immune microenvironment (TME) and immune checkpoints can identify new immunotherapeutic targets and their predictive and/or prognostic value. To characterize the TME and the immune checkpoint expression profile, we performed immunohistochemistry (IHC) on formalin-fixed paraffin embedded (FFPE) tissue sections from 54 MPM patients (40 at time of diagnosis; 14 treated with chemotherapy). We stained for PD-1, PD-L1, TIM-3, LAG-3, CD4, CD8, CD45RO, granzyme B, FoxP3 and CD68. Furthermore, we analyzed the relationship between the immunological parameters and survival, as well as response to chemotherapy. We found that TIM-3, PD-1 and PD-L1 were expressed on both immune and tumor cells. Strikingly, PD-1 and PD-L1 expression on tumor cells was only seen in unpretreated samples. No LAG-3 expression was observed. CD45RO expression in the stroma was an independent negative predictive factor for response on chemotherapy, while CD4 and TIM-3 expression in lymphoid aggregates were independent prognostic factors for better outcome. Our data propose TIM-3 as a promising new target in mesothelioma. Chemotherapy influences the expression of immune checkpoints and therefore further research on the best combination treatment schedule is required.
Collapse
Affiliation(s)
- Elly Marcq
- Center for Oncological Research, University of Antwerp , Antwerp, Belgium
| | - Vasiliki Siozopoulou
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Jorrit De Waele
- Center for Oncological Research, University of Antwerp , Antwerp, Belgium
| | | | - Karen Zwaenepoel
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Eva Santermans
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University , Diepenbeek, Belgium
| | - Niel Hens
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Diepenbeek, Belgium; Centre for Health Economics Research and Modeling Infectious Diseases, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Jan P van Meerbeeck
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Thoracic Oncology/MOCA, Antwerp University Hospital, Antwerp, Belgium
| | - Evelien L J Smits
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
17
|
Comar M, Zanotta N, Zanconati F, Cortale M, Bonotti A, Cristaudo A, Bovenzi M. Chemokines involved in the early inflammatory response and in pro-tumoral activity in asbestos-exposed workers from an Italian coastal area with territorial clusters of pleural malignant mesothelioma. Lung Cancer 2016; 94:61-7. [DOI: 10.1016/j.lungcan.2016.01.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 01/19/2016] [Accepted: 01/29/2016] [Indexed: 01/04/2023]
|
18
|
Coffelt SB, de Visser KE. Immune-mediated mechanisms influencing the efficacy of anticancer therapies. Trends Immunol 2015; 36:198-216. [PMID: 25857662 DOI: 10.1016/j.it.2015.02.006] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/18/2015] [Accepted: 02/18/2015] [Indexed: 01/26/2023]
Abstract
Conventional anticancer therapies, such as chemotherapy, radiotherapy, and targeted therapy, are designed to kill cancer cells. However, the efficacy of anticancer therapies is not only determined by their direct effects on cancer cells but also by off-target effects within the host immune system. Cytotoxic treatment regimens elicit several changes in immune-related parameters including the composition, phenotype, and function of immune cells. Here we discuss the impact of innate and adaptive immune cells on the success of anticancer therapy. In this context we examine the opportunities to exploit host immune responses to boost tumor clearing, and highlight the challenges facing the treatment of advanced metastatic disease.
Collapse
Affiliation(s)
- Seth B Coffelt
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Karin E de Visser
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Zhang W, Wu X, Wu L, Zhang W, Zhao X. Advances in the diagnosis, treatment and prognosis of malignant pleural mesothelioma. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:182. [PMID: 26366399 DOI: 10.3978/j.issn.2305-5839.2015.07.03] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/03/2015] [Indexed: 12/18/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rare cancer originated from pleural mesothelial cells. MPM has been associated with long-term exposure to asbestos. The prognosis of MPM is poor due to the difficulty of making diagnosis in the early stage, the rapid progression, the high invasiveness and the lack of effective treatment. Although the incidence of MPM is low in China to date, it has a tendency to increase in the coming years. The variety of clinical features may cause the delay of diagnosis and high rate of misdiagnosis. The diagnosis of MPM is based on biopsy of the pleura and immunohistochemistry. As China has become the largest country in the consumption of asbestos, it would give rise to a new surge of MPM in the future. The current treatment of MPM is multimodality therapy including surgery, radiotherapy, chemotherapy and immunotherapy. Two surgical procedures are commonly applied: extrapleural pneumonectomy (EPP) and pleurectomy/decortication (P/D). Three dimensional conformal radiotherapy is used to denote a spectrum of radiation planning and delivery techniques that rely on the 3D imaging to define the tumor. Cisplatin combined with pemetrexed (PEM) is the first-line chemotherapy for MPM. The principal targets in immunotherapy include T cells (Treg), CTLA-4 and PD-1. The diagnosis, treatment and prognosis still remain a major challenge for clinical research and will do so for years to come.
Collapse
Affiliation(s)
- Weiquan Zhang
- 1 Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan 250033, China ; 2 Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Xinshu Wu
- 1 Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan 250033, China ; 2 Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Licun Wu
- 1 Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan 250033, China ; 2 Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Weidong Zhang
- 1 Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan 250033, China ; 2 Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Xiaogang Zhao
- 1 Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan 250033, China ; 2 Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
20
|
Giaccone G, Bazhenova LA, Nemunaitis J, Tan M, Juhász E, Ramlau R, van den Heuvel MM, Lal R, Kloecker GH, Eaton KD, Chu Q, Dunlop DJ, Jain M, Garon EB, Davis CS, Carrier E, Moses SC, Shawler DL, Fakhrai H. A phase III study of belagenpumatucel-L, an allogeneic tumour cell vaccine, as maintenance therapy for non-small cell lung cancer. Eur J Cancer 2015; 51:2321-9. [PMID: 26283035 DOI: 10.1016/j.ejca.2015.07.035] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 07/12/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Treatment options after first-line chemotherapy are limited in non-small cell lung cancer (NSCLC). Belagenpumatucel-L is a therapeutic vaccine comprised of 4 transforming growth factor (TGF)-β2-antisense gene-modified, irradiated, allogeneic NSCLC cell lines that may be useful for maintenance after initial treatment. METHODS Stage III/IV NSCLC patients who did not progress after platinum-based chemotherapy were randomised 1:1 to receive maintenance belagenpumatucel-L or placebo. Patients were eligible for randomisation between one and four months from the end of induction chemotherapy. The primary endpoint was overall survival. RESULTS This phase III trial enrolled 270 patients in the belagenpumatucel-L arm and 262 in the control arm. Belagenpumatucel-L was well tolerated with no serious safety concerns. There was no difference in survival between the arms (median survival 20.3 versus 17.8months with belagenpumatucel-L versus placebo, respectively; hazard ratio (HR) 0.94, p=0.594). There were also no differences in progression-free survival (4.3months versus 4.0 for belagenpumatucel-L vs placebo, respectively; HR 0.99, p=0.947). A prespecified Cox regression analysis demonstrated that the time elapsed between randomisation and the end of induction chemotherapy had a significant impact on survival (p=0.002) and that prior radiation was a positive prognostic factor (median survival 28.4months with belagenpumatucel-L versus 16.0months with placebo; HR 0.61, p=0.032). CONCLUSIONS Although the overall trial did not meet its survival endpoint, improved survival for belagenpumatucel-L is suggested in patients who were randomised within 12weeks of completion of chemotherapy and in those who had received prior radiation. Further studies of belagenpumatucel-L in NSCLC are warranted.
Collapse
Affiliation(s)
- G Giaccone
- National Cancer Institute, Bethesda, MD, USA.
| | - L A Bazhenova
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - J Nemunaitis
- Mary Crowley Cancer Research Centers, Dallas, TX, USA
| | - M Tan
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - E Juhász
- Korányi National Institute for TB and Pulmonology, Budapest, Hungary
| | - R Ramlau
- Wielkopolskie Centrum Pulmonologii i Torakochirurgii, Poznań University of Medical Sciences, Poznan, Poland
| | - M M van den Heuvel
- Netherlands Cancer Institute, Antoni Van Leeuwenhoek Hospital, Thoracic Oncology, Amsterdam, Netherlands
| | - R Lal
- Guy's Hospital, King's Health Partners, London, England, United Kingdom
| | - G H Kloecker
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - K D Eaton
- Seattle Cancer Care Alliance, University of Washington, Seattle, WA, USA
| | - Q Chu
- Cross Cancer Institute, Edmonton, Alberta, Canada
| | - D J Dunlop
- Royal Infirmary, Glasgow, Scotland, United Kingdom
| | - M Jain
- Noble Hospital, Pune, India
| | - E B Garon
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | - E Carrier
- NovaRx Corporation, San Diego, CA, USA
| | - S C Moses
- NovaRx Corporation, San Diego, CA, USA
| | | | - H Fakhrai
- NovaRx Corporation, San Diego, CA, USA
| |
Collapse
|
21
|
Wu L, Yun Z, Tagawa T, De la Maza L, Wu MO, Yu J, Zhao Y, de Perrot M. Activation of CD1d-restricted natural killer T cells can inhibit cancer cell proliferation during chemotherapy by promoting the immune responses in murine mesothelioma. Cancer Immunol Immunother 2014; 63:1285-96. [PMID: 25183171 PMCID: PMC11029433 DOI: 10.1007/s00262-014-1597-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 08/07/2014] [Indexed: 12/31/2022]
Abstract
We studied the impact of natural killer T (NKT) cell activation by alpha-galactocysylceramide (α-GalCer, α-GC) on cancer cell repopulation during chemotherapy in murine mesothelioma. The number of NKT cells was found to be increased during the development of murine mesothelioma. NKT cells specifically recognize α-GC through CD1d resulting in their activation and expansion. Tumor-bearing mice were treated with chemotherapy once weekly, and α-GC was followed after each cycle of chemotherapy. Anti-tumor effect was evaluated on wild-type (WT) and CD1d knockout (CD1dKO) mice. Cancer cell proliferation and apoptosis were evaluated by Ki67 and TUNEL immunohistochemistry. CD4(+) and CD8(+) T cell proportion and activation in tumor, spleen, draining lymph node and peripheral blood were determined by flow cytometry, and gene expression of activated T cell-related cytokines was quantified by reverse transcription PCR. NKT cells were identified by CD1d-α-GC-tetramer staining. In WT mice, tumor growth delay was achieved by cisplatin (Cis), and this effect was improved in combination with α-GC, but α-GC alone had little effect. Cancer cell proliferation during chemotherapy was significantly inhibited by α-GC, while cancer cell death was significantly upregulated. α-GC following chemotherapy resulted in NKT cell expansion and an increase of interferon-γ production in the draining lymph node, blood and spleen. Gene expression of immune-associated cytokines was upregulated. Strikingly, the percentage of inducible T cell co-stimulator(+)CD4 T cells, Th17/Tc17 cells increased in splenocytes. In CD1d KO mice, however, Cis alone was less effective and Cis + α-GC provided no additional benefit over Cis alone. α-GC alone had minimal effect in both mice. NKT activation between cycles of chemotherapy could improve the outcome of mesothelioma treatment.
Collapse
Affiliation(s)
- Licun Wu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Zhihong Yun
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Tetsuzo Tagawa
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Luis De la Maza
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Matthew Onn Wu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Julie Yu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Yidan Zhao
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON Canada
- Toronto Mesothelioma Research Program, Division of Thoracic Surgery, Toronto General Hospital, 9N-961, 200 Elizabeth St, Toronto, ON M5G 2C4 Canada
| |
Collapse
|
22
|
Depletion of regulatory T cells by targeting folate receptor 4 enhances the potency of a GM-CSF-secreting tumor cell immunotherapy. Clin Immunol 2013; 148:287-98. [DOI: 10.1016/j.clim.2013.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/05/2013] [Accepted: 05/13/2013] [Indexed: 12/24/2022]
|
23
|
Immunity and malignant mesothelioma: From mesothelial cell damage to tumor development and immune response-based therapies. Cancer Lett 2012; 322:18-34. [DOI: 10.1016/j.canlet.2012.02.034] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 02/24/2012] [Accepted: 02/24/2012] [Indexed: 11/22/2022]
|
24
|
Wu L, Yun Z, Tagawa T, Rey-McIntyre K, de Perrot M. CTLA-4 blockade expands infiltrating T cells and inhibits cancer cell repopulation during the intervals of chemotherapy in murine mesothelioma. Mol Cancer Ther 2012; 11:1809-19. [PMID: 22584123 DOI: 10.1158/1535-7163.mct-11-1014] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer immunotherapy has shown promising results when combined with chemotherapy. Blocking CTLA-4 signaling by monoclonal antibody between cycles of chemotherapy may inhibit cancer cell repopulation and enhance the antitumoral immune reaction, thus improve the efficacy of chemotherapy in mesothelioma. The impact of CTLA-4 blockade on the early stage of tumor development was evaluated in a subcutaneous murine mesothelioma model. CTLA-4 blocking antibody was administered following each cycle of chemotherapy, and monotherapy was included as controls. Antitumor effect was evaluated by tumor growth delay and survival of the animals. Tumor cell repopulation was quantified by bromodeoxyuridine incorporation and Ki67 by immunohistochemistry and/or flow cytometry. In vitro cell killing was determined by classic chromium-released assay, and reverse transcription PCR (RT-PCR) was carried out to determine the gene expression of associated cytokines. Anti-CTLA-4 monoclonal antibody was able to inhibit tumor growth at early stage of tumor development. Antitumor effect was achieved by administration of CTLA-4 blockade between cycles of chemotherapy. Tumor cell repopulation during the intervals of cisplatin was inhibited by CTLA-4 blockade. Anti-CTLA-4 therapy gave rise to an increased number of CD4 and CD8 T cells infiltrating the tumor. RT-PCR showed that the gene expression of interleukin IL-2, IFN-γ, granzyme B, and perforin increased in the tumor milieu. Blockade of CTLA-4 signaling showed effective anticancer effect, correlating with inhibiting cancer cell repopulation between cycles of chemotherapy and upregulating tumor-infiltrating T lymphocytes, cytokines, and cytolytic enzymes in a murine mesothelioma model.
Collapse
Affiliation(s)
- Licun Wu
- Toronto Mesothelioma Research Program, Division of Thoracic Surgery, Toronto General Hospital, 200 Elizabeth St, Toronto, ON M5G 2C4, Canada
| | | | | | | | | |
Collapse
|
25
|
Ireland DJ, Kissick HT, Beilharz MW. The Role of Regulatory T Cells in Mesothelioma. CANCER MICROENVIRONMENT 2012; 5:165-72. [PMID: 22302659 DOI: 10.1007/s12307-012-0100-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 01/18/2012] [Indexed: 12/13/2022]
Abstract
Malignant mesothelioma (MM) appears to be responsive to immunotherapy. The lack of complete tumour cure as a result of many immunotherapies tested to date suggests that the immune response to MM is complex and multi-parametric. Regulatory T (Treg) cells are prevalent within murine and human mesotheliomas with their removal shown to result in tumour growth inhibition and the release of anti-tumour effector T cells from immunosuppression. The targeting of immune checkpoints as treatments for various solid tumours has recently shown promise in clinical settings. In addition, synergy between chemotherapy and immunotherapy has been demonstrated for many cancers, including mesothelioma. Here we demonstrate Treg cells as critical mediators of the anti-tumour immune response to MM and potential targets for anti-tumour immunotherapy; though the timing and dosage of Treg cell manipulating immunotherapies need to be optimised.
Collapse
Affiliation(s)
- Demelza J Ireland
- School of Pathology and Laboratory Medicine (M502), Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA, 6009, Australia.
| | - Haydn T Kissick
- Department of Surgery, Beth Israel Deaconess Medical Centre, Harvard Medical School, 3 Blackfan Circle, CLS 4/430, Boston, MA, 02215, USA
| | - Manfred W Beilharz
- School of Pathology and Laboratory Medicine (M502), Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA, 6009, Australia
| |
Collapse
|
26
|
Bograd AJ, Suzuki K, Vertes E, Colovos C, Morales EA, Sadelain M, Adusumilli PS. Immune responses and immunotherapeutic interventions in malignant pleural mesothelioma. Cancer Immunol Immunother 2011; 60:1509-27. [PMID: 21913025 DOI: 10.1007/s00262-011-1103-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 08/19/2011] [Indexed: 12/20/2022]
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive, primary pleural malignancy with poor prognosis, hypothesized to originate from a chronic inflammatory state within the pleura. Similar to what has been observed in other solid tumors (melanoma, ovarian and colorectal cancer), clinical and pre-clinical MPM investigations have correlated anti-tumor immune responses with improved survival. As such, a better understanding of the complex MPM tumor microenvironment is imperative in strategizing successful immunotherapies. Herein, we review the immune responses vital to the development and progression of MPM, as well as assess the role of immunomodulatory therapies, highlighting recent pre-clinical and clinical immunotherapy investigations.
Collapse
Affiliation(s)
- Adam J Bograd
- Division of Thoracic Surgery, Memorial Sloan-Kettering Cancer Center, NY 10065, USA
| | | | | | | | | | | | | |
Collapse
|