1
|
Al-Toubah T, Schell MJ, Morse B, Haider M, Valone T, Strosberg J. Phase II study of pembrolizumab and lenvatinib in advanced well-differentiated neuroendocrine tumors. ESMO Open 2024; 9:102386. [PMID: 38507897 PMCID: PMC10966166 DOI: 10.1016/j.esmoop.2024.102386] [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: 11/11/2023] [Revised: 12/15/2023] [Accepted: 01/25/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (CPIs) have not been shown to be active in well-differentiated neuroendocrine tumors (NETs), with response rates <5%. Lenvatinib is a multitargeted tyrosine kinase inhibitor which binds to vascular endothelial growth factor and fibroblast growth factor receptors and has demonstrated efficacy in pancreatic and gastrointestinal NETs [44% and 16% objective radiographic response rate (ORR), respectively]. The combination of antiangiogenic and CPI therapies can be synergistic. We therefore evaluated the combination of lenvatinib and pembrolizumab in well-differentiated gastrointestinal (GI) and thoracic NETs. PATIENTS AND METHODS A prospective, phase II trial evaluated patients with advanced GI/thoracic NETs (pancreatic NETs were excluded due to high response rate of lenvatinib monotherapy in this patient population), with evidence of progression within 8 months of study entry and at least two prior lines of systemic therapy. Patients received lenvatinib 20 mg daily and pembrolizumab 200 mg intravenously every 3 weeks until unacceptable toxicity or progression of disease. Primary endpoint was objective response rate, and an interim analysis was planned once 20 patients were enrolled. Four ORRs were required to continue enrollment. RESULTS Twenty patients were enrolled on protocol from April 2021 to January 2022 (nine small intestine, five lung, two thymic, two unknown primary, one cecal, one presacral primaries). Two patients (10%) achieved a partial response (atypical lung and small intestinal primaries). Median progression-free survival (PFS) was 8 months (95% confidence interval 5.8-10.2 months). Twelve (60%) patients experienced probably or definitely associated grade 3 adverse events (10 hypertension). Fourteen patients (70%) required dose reductions or discontinued one of the medications. Two patients discontinued treatment before radiographic assessment. CONCLUSIONS The combination of pembrolizumab and lenvatinib did not show sufficient response in patients with NETs to warrant continued enrollment on trial.
Collapse
Affiliation(s)
- T Al-Toubah
- Department of GI Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - M J Schell
- Department of Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - B Morse
- Department of Diagnostic Imaging, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - M Haider
- Department of GI Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - T Valone
- Department of GI Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - J Strosberg
- Department of GI Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA.
| |
Collapse
|
2
|
Yang JK, Kwon H, Kim S. Recent advances in light-triggered cancer immunotherapy. J Mater Chem B 2024; 12:2650-2669. [PMID: 38353138 DOI: 10.1039/d3tb02842a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Light-triggered phototherapies, such as photodynamic therapy (PDT) and photothermal therapy (PTT), have shown strong therapeutic efficacy with minimal invasiveness and systemic toxicity, offering opportunities for tumor-specific therapies. Phototherapies not only induce direct tumor cell killing, but also trigger anti-tumor immune responses by releasing various immune-stimulating factors. In recent years, conventional phototherapies have been combined with cancer immunotherapy as synergistic therapeutic modalities to eradicate cancer by exploiting the innate and adaptive immunity. These combined photoimmunotherapies have demonstrated excellent therapeutic efficacy in preventing tumor recurrence and metastasis compared to phototherapy alone. This review covers recent advancements in combined photoimmunotherapy, including photoimmunotherapy (PIT), PDT-combined immunotherapy, and PTT-combined immunotherapy, along with their underlying anti-tumor immune response mechanisms. In addition, the challenges and future research directions for light-triggered cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Jin-Kyoung Yang
- Department of Chemical Engineering, Dong-eui University, Busan, 47340, Republic of Korea.
| | - Hayoon Kwon
- Chemical & Biological integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Sehoon Kim
- Chemical & Biological integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
3
|
Carretta M, Thorseth ML, Schina A, Agardy DA, Johansen AZ, Baker KJ, Khan S, Rømer AMA, Fjæstad KY, Linder H, Kuczek DE, Donia M, Grøntved L, Madsen DH. Dissecting tumor microenvironment heterogeneity in syngeneic mouse models: insights on cancer-associated fibroblast phenotypes shaped by infiltrating T cells. Front Immunol 2024; 14:1320614. [PMID: 38259467 PMCID: PMC10800379 DOI: 10.3389/fimmu.2023.1320614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Murine syngeneic tumor models have been used extensively for cancer research for several decades and have been instrumental in driving the discovery and development of cancer immunotherapies. These tumor models are very simplistic cancer models, but recent reports have, however, indicated that the different inoculated cancer cell lines can lead to the formation of unique tumor microenvironments (TMEs). To gain more knowledge from studies based on syngeneic tumor models, it is essential to obtain an in-depth understanding of the cellular and molecular composition of the TME in the different models. Additionally, other parameters that are important for cancer progression, such as collagen content and mechanical tissue stiffness across syngeneic tumor models have not previously been reported. Here, we compare the TME of tumors derived from six common syngeneic tumor models. Using flow cytometry and transcriptomic analyses, we show that strikingly unique TMEs are formed by the different cancer cell lines. The differences are reflected as changes in abundance and phenotype of myeloid, lymphoid, and stromal cells in the tumors. Gene expression analyses support the different cellular composition of the TMEs and indicate that distinct immunosuppressive mechanisms are employed depending on the tumor model. Cancer-associated fibroblasts (CAFs) also acquire very different phenotypes across the tumor models. These differences include differential expression of genes encoding extracellular matrix (ECM) proteins, matrix metalloproteinases (MMPs), and immunosuppressive factors. The gene expression profiles suggest that CAFs can contribute to the formation of an immunosuppressive TME, and flow cytometry analyses show increased PD-L1 expression by CAFs in the immunogenic tumor models, MC38 and CT26. Comparison with CAF subsets identified in other studies shows that CAFs are skewed towards specific subsets depending on the model. In athymic mice lacking tumor-infiltrating cytotoxic T cells, CAFs express lower levels of PD-L1 and lower levels of fibroblast activation markers. Our data underscores that CAFs can be involved in the formation of an immunosuppressive TME.
Collapse
Affiliation(s)
- Marco Carretta
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Marie-Louise Thorseth
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Aimilia Schina
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Dennis Alexander Agardy
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Astrid Zedlitz Johansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Kevin James Baker
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Shawez Khan
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Anne Mette Askehøj Rømer
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Klaire Yixin Fjæstad
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Hannes Linder
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Dorota Ewa Kuczek
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Daniel Hargbøl Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Del Prete A, Salvi V, Soriani A, Laffranchi M, Sozio F, Bosisio D, Sozzani S. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell Mol Immunol 2023; 20:432-447. [PMID: 36949244 PMCID: PMC10203372 DOI: 10.1038/s41423-023-00990-6] [Citation(s) in RCA: 254] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/14/2023] [Indexed: 03/24/2023] Open
Abstract
Dendritic cells (DCs) exhibit a specialized antigen-presenting function and play crucial roles in both innate and adaptive immune responses. Due to their ability to cross-present tumor cell-associated antigens to naïve T cells, DCs are instrumental in the generation of specific T-cell-mediated antitumor effector responses in the control of tumor growth and tumor cell dissemination. Within an immunosuppressive tumor microenvironment, DC antitumor functions can, however, be severely impaired. In this review, we focus on the mechanisms of DC capture and activation by tumor cell antigens and the role of the tumor microenvironment in shaping DC functions, taking advantage of recent studies showing the phenotype acquisition, transcriptional state and functional programs revealed by scRNA-seq analysis. The therapeutic potential of DC-mediated tumor antigen sensing in priming antitumor immunity is also discussed.
Collapse
Affiliation(s)
- Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Humanitas Clinical and Research Center-IRCCS Rozzano, Milano, Italy
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessandra Soriani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mattia Laffranchi
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Sozio
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Silvano Sozzani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
- IRCCS Neuromed, Pozzilli, IS, Italy.
| |
Collapse
|
5
|
Li H, Deng C, Tan Y, Dong J, Zhao Y, Wang X, Yang X, Luo J, Gao H, Huang Y, Zhang ZR, Gong T. Chondroitin sulfate-based prodrug nanoparticles enhance photodynamic immunotherapy via Golgi apparatus targeting. Acta Biomater 2022; 146:357-369. [PMID: 35577045 DOI: 10.1016/j.actbio.2022.05.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/28/2022] [Accepted: 05/09/2022] [Indexed: 01/02/2023]
Abstract
Photodynamic therapy (PDT) is an emerging therapeutic approach that can inhibit tumor growth by destroying local tumors and activating systemic antitumor immune responses. However, PDT can be ineffective because of photosensitizer aggregation, tumor-induced dendritic cells (DCS) dysfunction and PDT-mediated immunosuppression. Therefore, we designed chondroitin sulfate-based prodrug nanoparticles for the co-delivery of the photosensitizer chlorin e6 (Ce6) and retinoic acid (RA), which can reduce PDT-mediated immunosuppression by disrupting the Golgi apparatus and blocking the production of immunosuppressive cytokines. Moreover, CpG oligodeoxynucleotide was combined as immunoadjuvant to promote the maturation of DCs. As expected, the strategy of Golgi apparatus targeting immunotherapy combined PDT was confirmed to relieve PDT-induced immunosuppression, showed excellent PDT antitumor efficacy in B16F10-subcutaneous bearing mice model. Thus, our finding offers a promising approach for photodynamic immunotherapy of advanced cancers. STATEMENT OF SIGNIFICANCE: Golgi apparatus has been shown to be a potential target of immunosuppression for producing several immunosuppressive cytokines. In this work, a Golgi apparatus-targeted prodrug nanoparticle was developed to enhance the immune response in photodynamic immunotherapy. The nanoparticle can target and disrupt the Golgi apparatus in tumor cells, which reduced PDT-mediated immunosuppression by blocking the production of immunosuppressive cytokines. This work provides an effective strategy of PDT in combination with the Golgi apparatus-targeted nanovesicle for enhanced cancer therapy.
Collapse
Affiliation(s)
- Haohuan Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China; Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Caifeng Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China; Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yulu Tan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Jianxia Dong
- Department of Clinical Pharmacy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuanhao Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Xiaorong Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Xingyue Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Jingwen Luo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology and Hubei Collaborative Innovation Center for Green Transformation of Bio-resources, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Zhi-Rong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
6
|
Drocaş I, Crăiţoiu Ş, Stepan AE, Simionescu CE, Marinescu D. VEGF Immunoexpression in Endometrioid Endometrial Carcinomas. CURRENT HEALTH SCIENCES JOURNAL 2022; 48:155-161. [PMID: 36320870 PMCID: PMC9590369 DOI: 10.12865/chsj.48.02.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/20/2022] [Indexed: 11/05/2022]
Abstract
Vascular endothelial growth factor (VEGF) is the most important stimulator of endometrial tumor angiogenesis, a mechanism that may be a therapeutic target in the context of an incidence and persistent mortality of endometrial endometrial carcinomas (EEC). In this study, VEGF immunoexpression was analyzed for 50 cases of EEC in relation to the histopathological parameters of tumor aggressiveness. High VEGF scores have been associated with the high grade and advanced stage of EEC, but unrelated to the depth of myometrial invasion, the pattern of tumor invasion, or vascular invasion. VEGF may be useful for assessing EEC aggression, but also for tumor angiogenic potential, which recommends it as a possible mark for specific antitumor therapy.
Collapse
Affiliation(s)
- Ileana Drocaş
- PhD Student, Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Ştefania Crăiţoiu
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Alex Emilian Stepan
- Department of Pathology, University of Medicine and Pharmacy of Craiova, Romania
| | | | - Daniela Marinescu
- Department of General Surgery, University of Medicine and Pharmacy of Craiova
| |
Collapse
|
7
|
LENG X, HUANG G, MA F, DING J. Correlation between the characteristics of ultrasonic contrast and the regional distribution of tumor vascular heterogeneity in breast cancer. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.40320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Xiaoling LENG
- The Affiliated Tumor Hospital of Xinjiang Medical University, China
| | - Guofu HUANG
- The Fifth Affiliated Hospital of Xinjiang Medical University, China
| | - Fucheng MA
- The Affiliated Tumor Hospital of Xinjiang Medical University, China
| | - Jianbing DING
- College of Basic Medicine, Xinjiang Medical University, China
| |
Collapse
|
8
|
Yan Z, Ma J, Yao S, Yao Z, Wang H, Chu J, Zhao S, Liu Y. Anti-Angiogenic Agent Combined with Anti-PD-1 Immunotherapy Showed Activity in Patients With Classical Hodgkin Lymphoma Who Have Failed Immunotherapy: A Retrospective Case Report Study. Front Immunol 2021; 12:727464. [PMID: 34899689 PMCID: PMC8664390 DOI: 10.3389/fimmu.2021.727464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/12/2021] [Indexed: 11/18/2022] Open
Abstract
Background PD-1/PD-L1 inhibitor immunotherapy has showed impressive activity in various cancers, especially relapsed/refractory (r/r) classical Hodgkin lymphoma (cHL). However, acquired resistance is inevitable for most patients. Sometimes severe side effects also lead to treatment termination. When immunotherapy failed, alternative treatment options are limited. In the past few years, we have used the anti-angiogenic agent apatinib and PD-1 inhibitor camrelizumab to treat cHL patients who failed prior immunotherapy. In this study, we analyzed the data of these patients. Patients and Methods Patients with r/r cHL who had failed immunotherapy and subsequently received apatinib-camrelizumab (AC) combination therapy were included in this study. Patient data were collected from medical records and follow-up system. The efficacy and safety of AC therapy were analyzed. Results Seven patients who failed immunotherapy were identified in our database, of which five patients acquired immunotherapy resistance and two patients experienced severe side effects. They received a combination of camrelizumab (200 mg every four weeks) and apatinib (425 mg or 250 mg per day). As of the cut-off date, these patients had received a median of 4 cycles (range, 2 - 31) of treatment. Two (2/7) patients achieved complete response, four (4/7) partial response, and one (1/7) stable disease. The median progression-free survival was 10.0 months (range, 2.0 – 27.8). Low-dose apatinib (250 mg) plus camrelizumab was well tolerated and had no unexpected side effects. Besides, no reactive cutaneous capillary endothelial proliferation was observed in AC-treated patients. Conclusions Low dose apatinib plus camrelizumab might be a promising treatment option for r/r cHL patients who have failed immunotherapy. This combination treatment is worthy of further investigation in more patients including solid cancer patients who have failed immunotherapy.
Collapse
Affiliation(s)
- Zheng Yan
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jialin Ma
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Shuna Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Zhihua Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Haiying Wang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Junfeng Chu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Shuang Zhao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yanyan Liu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
9
|
Dyhl-Polk A, Mikkelsen MK, Ladekarl M, Nielsen DL. Clinical Trials of Immune Checkpoint Inhibitors in Hepatocellular Carcinoma. J Clin Med 2021; 10:2662. [PMID: 34208788 PMCID: PMC8234948 DOI: 10.3390/jcm10122662] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Introduction: Several immune checkpoint inhibitors (CPIs) are under clinical development in hepatocellular carcinoma (HCC) and the field is advancing rapidly. In this comprehensive review, we discuss published results and report on ongoing clinical trials. Methods: A literature search was carried out using PubMed and EMBASE; data reported at international meetings and clinicaltrials.gov were included as well. The search was updated 5 March 2021. We evaluated studies with monotherapy CPI's, combinations of CPI's and combinations of CPI's with other treatment modalities separately. Only studies with at least 10 included patients were considered. Results: We identified 2649 records published in the English language literature. After review, 29 studies remained, including 12 studies with preliminary data only. The obtained overall response rate of PD-1/PDL-1 monotherapy in phase II studies in the second-line setting was 15-20% with disease control in approximately 60% of patients. The responses were of long duration in a subset of patients. Furthermore, the safety profiles were manageable. However, a phase III study comparing nivolumab with sorafenib in the first-line setting and a phase III study evaluating pembrolizumab versus best supportive care in the second-line setting did not meet their prespecified endpoints. More recently, a phase I/II study of nivolumab and ipilimumab has resulted in a response rate of approximately 30% with a median OS of 22 months in the second-line setting. Multiple trials have been initiated to evaluate CPIs in combination with molecularly targeted drugs, especially anti-angiogenic drugs or local therapy. A phase III study investigating atezolizumab plus bevacizumab versus sorafenib in the first-line setting showed significantly increased survival in the combination arm. Conclusions: The combination of atezolizumab and bevacizumab represents a new standard of care in the first-line setting for fit patients with preserved liver function. CPIs can produce durable tumor remission and induce long-standing anti-tumor immunity in a subgroup of patients with advanced HCC. Although phase III trials of CPI monotherapy have been negative, the combination of PD-1/PD-L1 inhibitors with other anti-angiogenic drugs, CTLA-4 inhibitors or other modalities may result in new treatment options for patients with HCC. Research on predictive biomarkers is crucial for further development of CPIs in HCC.
Collapse
Affiliation(s)
- Anne Dyhl-Polk
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; (M.K.M.); (D.L.N.)
| | - Marta Kramer Mikkelsen
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; (M.K.M.); (D.L.N.)
| | - Morten Ladekarl
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Hobrovej 19-22, 9000 Aalborg, Denmark;
| | - Dorte Lisbet Nielsen
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; (M.K.M.); (D.L.N.)
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
10
|
Heeran AB, Dunne MR, Morrissey ME, Buckley CE, Clarke N, Cannon A, Donlon NE, Nugent TS, Durand M, Dunne C, Larkin JO, Mehigan B, McCormick P, Lynam-Lennon N, O’Sullivan J. The Protein Secretome Is Altered in Rectal Cancer Tissue Compared to Normal Rectal Tissue, and Alterations in the Secretome Induce Enhanced Innate Immune Responses. Cancers (Basel) 2021; 13:571. [PMID: 33540635 PMCID: PMC7867296 DOI: 10.3390/cancers13030571] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Locally advanced rectal cancer is treated with neoadjuvant-chemoradiotherapy; however, only ~22% of patients achieve a complete response, and resistance mechanisms are poorly understood. The role of inflammation and immune cell biology in this setting is under-investigated. In this study, we profiled the inflammatory protein secretome of normal (non-cancer) (n = 8) and malignant rectal tissue (n = 12) pre- and post-radiation in human ex vivo explant models and examined the influence of these untreated and treated secretomes on dendritic cell biology (n = 8 for cancer and normal). These resultant profiles were correlated with patient clinical characteristics. Nineteen factors were secreted at significantly higher levels from the rectal cancer secretome when compared to the normal rectal secretome; Flt-1, P1GF, IFN-γ, IL-6, IL-10, CCL20, CCL26, CCL22, CCL3, CCL4, CCL17, GM-CSF, IL-12/IL-23p40, IL-17A, IL-1α, IL-17A/F, IL-1RA, TSLP and CXCL10 (p < 0.05). Radiation was found to have differential effects on normal rectal tissue and rectal cancer tissue with increased IL-15 and CCL22 secretion following radiation from normal rectal tissue explants (p < 0.05), while no significant alterations were observed in the irradiated rectal cancer tissue. Interestingly, however, the irradiated rectal cancer secretome induced the most potent effect on dendritic cell maturation via upregulation of CD80 and PD-L1. Patient's visceral fat area correlated with secreted factors including CCL20, suggesting that obesity status may alter the tumour microenvironment (TME). These results suggest that radiation does not have a negative effect on the ability of the rectal cancer TME to induce an immune response. Understanding these responses may unveil potential therapeutic targets to enhance radiation response and mitigate normal tissue injury. Tumour irradiation in this cohort enhances innate immune responses, which may be harnessed to improve patient treatment outcome.
Collapse
Affiliation(s)
- Aisling B. Heeran
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Margaret R. Dunne
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Maria E. Morrissey
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Croí E. Buckley
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Niamh Clarke
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Aoife Cannon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Noel E. Donlon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Timothy S. Nugent
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Michael Durand
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - Cara Dunne
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - John O. Larkin
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - Brian Mehigan
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - Paul McCormick
- GEMS, St. James’s Hospital, D08 NHY1 Dublin 8, Ireland; (M.D.); (C.D.); (J.O.L.); (B.M.); (P.M.)
| | - Niamh Lynam-Lennon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| | - Jacintha O’Sullivan
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James’s Hospital, D08 W9RT Dublin 8, Ireland; (A.B.H.); (M.R.D.); (M.E.M.); (C.E.B.); (N.C.); (A.C.); (N.E.D.); (T.S.N.); (N.L.-L.)
| |
Collapse
|
11
|
Wooster AL, Girgis LH, Brazeale H, Anderson TS, Wood LM, Lowe DB. Dendritic cell vaccine therapy for colorectal cancer. Pharmacol Res 2021; 164:105374. [PMID: 33348026 PMCID: PMC7867624 DOI: 10.1016/j.phrs.2020.105374] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths in the United States despite an array of available treatment options. Current standard-of-care interventions for this malignancy include surgical resection, chemotherapy, and targeted therapies depending on the disease stage. Specifically, infusion of anti-vascular endothelial growth factor agents in combination with chemotherapy was an important development in improving the survival of patients with advanced colorectal cancer, while also helping give rise to other forms of anti-angiogenic therapies. Yet, one approach by which tumor angiogenesis may be further disrupted is through the administration of a dendritic cell (DC) vaccine targeting tumor-derived blood vessels, leading to cytotoxic immune responses that decrease tumor growth and synergize with other systemic therapies. Early generations of such vaccines exhibited protection against various forms of cancer in pre-clinical models, but clinical results have historically been disappointing. Sipuleucel-T (Provenge®) was the first, and to-date, only dendritic cell-based therapy to receive FDA approval after significantly increasing overall survival in prostate cancer patients. The unparalleled success of Sipuleucel-T has helped revitalize the clinical development of dendritic cell vaccines, which will be examined in this review. We also highlight the promise of these vaccines to instill anti-angiogenic immunity for individuals with advanced colorectal cancer.
Collapse
Affiliation(s)
- Amanda L Wooster
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Lydia H Girgis
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Hayley Brazeale
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Trevor S Anderson
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Laurence M Wood
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Devin B Lowe
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States.
| |
Collapse
|
12
|
Melanoma Cell Resistance to Vemurafenib Modifies Inter-Cellular Communication Signals. Biomedicines 2021; 9:biomedicines9010079. [PMID: 33467521 PMCID: PMC7830125 DOI: 10.3390/biomedicines9010079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/15/2022] Open
Abstract
The therapeutic success of BRAF inhibitors (BRAFi) and MEK inhibitors (MEKi) in BRAF-mutant melanoma is limited by the emergence of drug resistance, and several lines of evidence suggest that changes in the tumor microenvironment can play a pivotal role in acquired resistance. The present study focused on secretome profiling of melanoma cells sensitive or resistant to the BRAFi vemurafenib. Proteomic and cytokine/chemokine secretion analyses were performed in order to better understand the interplay between vemurafenib-resistant melanoma cells and the tumor microenvironment. We found that vemurafenib-resistant melanoma cells can influence dendritic cell (DC) maturation by modulating their activation and cytokine production. In particular, human DCs exposed to conditioned medium (CM) from vemurafenib-resistant melanoma cells produced higher levels of pro-inflammatory cytokines—that potentially facilitate melanoma growth—than DCs exposed to CM derived from parental drug-sensitive cells. Bioinformatic analysis performed on proteins identified by mass spectrometry in the culture medium from vemurafenib-sensitive and vemurafenib-resistant melanoma cells suggests a possible involvement of the proteasome pathway. Moreover, our data confirm that BRAFi-resistant cells display a more aggressive phenotype compared to parental ones, with a significantly increased production of interferon-γ, interleukin-8, vascular-endothelial growth factor, CD147/basigin, and metalloproteinase 2 (MMP-2). Plasma levels of CD147/basigin and MMP-2 were also measured before the start of therapy and at disease progression in a small group of melanoma patients treated with vemurafenib or vemurafenib plus cobimetinib. A significant increment in CD147/basigin and MMP-2 was observed in all patients at the time of treatment failure, strengthening the hypothesis that CD147/basigin might play a role in BRAFi resistance.
Collapse
|
13
|
Liu M, Cai L, Li Q, Chen X, Gao L, Jiang L. The Expression of VEGF and CD31 in Endometrial Lesions and Its Associations with Blood Flow Parameters of Transvaginal 3D Power Doppler Ultrasonography: A Preliminary Study. Cancer Manag Res 2020; 12:11211-11218. [PMID: 33177872 PMCID: PMC7649237 DOI: 10.2147/cmar.s277274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/09/2020] [Indexed: 11/23/2022] Open
Abstract
Aim To investigate the association of the blood flow parameters measured by transvaginal three-dimensional power Doppler ultrasound and the angiogenesis of endometrial cancer. Material and Methods The expressions of vascular endothelial growth factor (VEGF) and CD31 in benign and malignant endometrial lesions, and in malignant lesions with different clinical and pathological features were analyzed. The correlations of the blood flow parameters (vascularization index [VI], blood flow index [FI], and vascularization-blood flow index [VFI]) of transvaginal 3D power Doppler ultrasound, and VEGF expressions, microvessel density (MVD) were also evaluated. Results The VEGF-positive rates and the MVD values in benign and malignant endometrial lesions were significantly different (both P<0.001). The differences of VEGF-positive rates (P < 0.001) and MVD values (P = 0.021) between type I and type II lesions of endometrial cancer were statistically significant. There was no significant difference in the VEGF-positive rate and MVD value between stage IA and IB (P=0.443, P=0.311). The difference of VEGF expression and MVD in stage IA, stage IB and stage II and above was statistically significant (P=0.003, P=0.017). The VEGF-positive rate and MVD value were not significantly different in IAG1 and IAG2 lesions of endometrioid adenocarcinoma (P=0.709, P=0.792). There was no significant correlation between VI, FI, VFI and VEGF expression and MVD in endometrial cancer. Conclusion The VEGF-positive rates and MVD values were relatively high in malignant endometrial lesions, type II and stage II and above lesions of type I endometrial cancer, indicating that the angiogenesis of endometrial cancer tissues might play a crucial role in the tumor classification, and pelvic metastasis.
Collapse
Affiliation(s)
- Meijuan Liu
- Department of Ultrasound, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| | - Li Cai
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| | - Qifan Li
- Department of Ultrasound, RongCheng Maternal and Child Care Service Centre, Weihai, People's Republic of China
| | - Xiaoran Chen
- Department of Ultrasound, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| | - Lingyun Gao
- Department of Ultrasound, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| | - Lei Jiang
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, People's Republic of China
| |
Collapse
|
14
|
Kennedy SA, Morrissey ME, Dunne MR, O'Connell F, Butler CT, Cathcart MC, Buckley AM, Mehigan BJ, Larkin JO, McCormick P, Kennedy BN, O'Sullivan J. Combining 1,4-dihydroxy quininib with Bevacizumab/FOLFOX alters angiogenic and inflammatory secretions in ex vivo colorectal tumors. BMC Cancer 2020; 20:952. [PMID: 33008336 PMCID: PMC7532092 DOI: 10.1186/s12885-020-07430-y] [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: 05/18/2020] [Accepted: 09/16/2020] [Indexed: 11/17/2022] Open
Abstract
Background Colorectal cancer (CRC) is the second most common cause of cancer-related mortality worldwide with one in every five patients diagnosed with metastatic CRC (mCRC). In mCRC cases, the 5-year survival rate remains at approximately 14%, reflecting the lack of effectiveness of currently available treatments such as the anti-VEGF targeting antibody Bevacizumab combined with the chemotherapy folinic acid, fluorouracil and oxaliplatin (FOLFOX). Approximately 60% of patients do not respond to this combined treatment. Furthermore, Bevacizumab inhibits dendritic cell (DC) maturation in poor responders, a key process for tumor eradication. Method Following drug treatment, secreted expression levels of angiogenic and inflammatory markers in tumor conditioned media generated from human ex vivo colorectal tumors were measured by ELISA. Dendritic cell phenotypic and maturation markers were assessed by flow cytometry. Results Our novel compound, 1,4-dihydroxy quininib, acts in an alternative pathway compared to the approved therapy Bevacizumab. 1,4-dihydroxy quininib alone, and in combination with Bevacizumab or FOLFOX significantly reduced TIE-2 expression which is involved in the promotion of tumor vascularization. Combination treatment with 1,4-dihydroxy quininib significantly increased the expression level of DC phenotypic and maturation markers. Conclusion Our results indicate the anti-angiogenic small molecule 1,4-dihydroxy quininib could be an alternative novel treatment in combination therapy for CRC patients.
Collapse
Affiliation(s)
- Susan A Kennedy
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Maria E Morrissey
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Margaret R Dunne
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Fiona O'Connell
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Clare T Butler
- UCD Conway Institute & UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| | - Mary-Clare Cathcart
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | - Amy M Buckley
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland
| | | | | | | | - Breandán N Kennedy
- UCD Conway Institute & UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| | - Jacintha O'Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin 8, Ireland.
| |
Collapse
|
15
|
Qi X, Yang M, Ma L, Sauer M, Avella D, Kaifi JT, Bryan J, Cheng K, Staveley-O'Carroll KF, Kimchi ET, Li G. Synergizing sunitinib and radiofrequency ablation to treat hepatocellular cancer by triggering the antitumor immune response. J Immunother Cancer 2020; 8:e001038. [PMID: 33115942 PMCID: PMC7594543 DOI: 10.1136/jitc-2020-001038] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Minimally invasive radiofrequency ablation (RFA) is used as a first-line treatment option for hepatocellular cancer (HCC) with the weaknesses of incomplete ablation, tumor recurrence, and inferior outcomes. To overcome this limitation, we proposed to develop sunitinib-RFA integrated therapy with a potential of activating anti-HCC immune response. METHODS Using our unique murine model, we developed a novel RFA platform with a modified human cardiac RF generator. Therapeutic efficacy of sunitinib-RFA combined treatment in HCC was tested in this platform. Tumor progression was monitored by MRI; tumor necrosis and apoptosis were detected by H&E and terminal deoxynucleotidyl transferase dUTP nick end labeling; immune reaction was defined by flow cytometry; and signaling molecules were examined with real-time PCR (qPCR), western blot, and immunohistochemical staining. RESULTS A significantly reduced tumor growth and extended lift span were observed in the mice receiving combined treatment with RFA and sunitinib. This combined treatment significantly increased the frequency of CD8+ T cell, memory CD8+ T cell, and dendritic cells (DCs); decreased the frequency of regulatory T cells; and activated tumor-specific antigen (TSA) immune response in tumor microenvironment. We found that RFA caused PD-1 upregulation in tumor-infiltrated T cells by boosting hepatocyte growth factor (HGF) expression, which was suppressed by sunitinib treatment. We have also demonstrated that sunitinib suppressed VEGF's effect in enhancing PD-L1 expression in DCs and attenuated heat-sink effect. The results indicate that RFA induced tumor destruction and release of in situ TSAs which can activate a tumoricidal immune response in sunitinib-treated mice, significantly improving anti-HCC therapeutic efficacy. CONCLUSIONS Sunitinib enables RFA-released in situ TSA to ignite an effective anti-tumor immune response by suppressing HGF and VEGF signaling pathways. Sunitinib-RFA as a synergistic therapeutic approach significantly suppresses HCC growth.
Collapse
Affiliation(s)
- Xiaoqiang Qi
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
| | - Lixin Ma
- Department of Radiology, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial VA Hospital, Columbia, Missouri, USA
| | - Madeline Sauer
- School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Diego Avella
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, Missouri, USA
| | - Jussuf T Kaifi
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, Missouri, USA
| | - Jeffrey Bryan
- Department of Veterinary Oncology, University of Missouri, Columbia, Missouri, USA
| | - Kun Cheng
- Pharmacology and Pharmaceutical Sciences, University of Missouri Kansas City, Kansas City, Missouri, USA
| | - Kevin F Staveley-O'Carroll
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial VA Hospital, Columbia, Missouri, USA
| | - Eric T Kimchi
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial VA Hospital, Columbia, Missouri, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri, Columbia, Missouri, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
16
|
Yan Z, Yao ZH, Yao SN, Wang HY, Chu JF, Song M, Zhao S, Liu YY. Camrelizumab plus apatinib successfully treated a patient with advanced esophageal squamous cell carcinoma. Immunotherapy 2020; 12:1161-1166. [PMID: 32814482 DOI: 10.2217/imt-2020-0197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: Advanced esophageal squamous cell carcinoma (ESCC) is a lethal disease with poor response to conventional chemotherapy. Immunotherapy showed better activity than chemotherapy in late-line treatment. However, the rate and duration of response are far from satisfactory. The efficacy of an anti-angiogenic agent combined with immunotherapy for ESCC is unknown. Results: A patient with ESCC experienced disease relapse after chemo-radiotherapy. The disease progressed after combined chemotherapy. A combination regimen of the PD-1 inhibitor camrelizumab and the anti-angiogenic agent apatinib was administered. The patient achieved a PET/CT-confirmed durable complete response with mild toxicity. Conclusion: The PD-1 inhibitor combined with the anti-angiogenic agent is effective and safe for the treatment of ESCC. This regimen is worth investigation in clinical trials.
Collapse
Affiliation(s)
- Zheng Yan
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Zhi-Hua Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Shu-Na Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Hai-Ying Wang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Jun-Feng Chu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Ming Song
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Shuang Zhao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| | - Yan-Yan Liu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, 27 Dongming Road, Zhengzhou, Henan 450008, China
| |
Collapse
|
17
|
Hameed S, Mo S, Mustafa G, Bajwa SZ, Khan WS, Dai Z. Immunological Consequences of Nanoparticle‐Mediated Antitumor Photoimmunotherapy. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sadaf Hameed
- Department of Biomedical EngineeringCollege of EngineeringPeking University Beijing 100871 China
| | - Shanyan Mo
- Department of Biomedical EngineeringCollege of EngineeringPeking University Beijing 100871 China
| | - Ghulam Mustafa
- Department of SciencesBahria University Lahore Lahore 54000 Pakistan
| | - Sadia Z. Bajwa
- Nanobiotech GroupNational Institute for Biotechnology and Genetic Engineering (NIBGE) P.O. Box No. 577, Jhang Road Faisalabad 44000 Pakistan
| | - Waheed S. Khan
- Nanobiotech GroupNational Institute for Biotechnology and Genetic Engineering (NIBGE) P.O. Box No. 577, Jhang Road Faisalabad 44000 Pakistan
| | - Zhifei Dai
- Department of Biomedical EngineeringCollege of EngineeringPeking University Beijing 100871 China
| |
Collapse
|
18
|
Ovarian Cancer Immunotherapy: Turning up the Heat. Int J Mol Sci 2019; 20:ijms20122927. [PMID: 31208030 PMCID: PMC6628106 DOI: 10.3390/ijms20122927] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the leading cause of death among gynecological malignancies. Despite surgery and chemotherapy, 5-years survival rates have improved only modestly over the past few decades remaining at 45% for advanced stages. Therefore, novel therapies are urgently needed. The presence of tumor-infiltrating lymphocytes (TILs) in OC tumor microenvironment (TME) has already proved to be correlated with overall survival (OS), while immune evasion mechanisms are associated with poor prognosis. Although these data indicate that immunotherapy has a strong rationale in OC, single agent immune-checkpoints inhibitors (ICIs) have shown only modest results in this malignancy. In this review, we will discuss immune-targeting combination therapies and adoptive cell therapy (ACT), highlighting the challenges represented by these strategies, which aim at disrupting the stroma-tumor barrier to boost immune system against ovarian cancer.
Collapse
|
19
|
Saxena V, Fitch J, Ketz J, White P, Wetzel A, Chanley MA, Spencer JD, Becknell B, Pierce KR, Arregui SW, Nelson RD, Schwartz GJ, Velazquez V, Walker LA, Chen X, Yan P, Hains DS, Schwaderer AL. Whole Transcriptome Analysis of Renal Intercalated Cells Predicts Lipopolysaccharide Mediated Inhibition of Retinoid X Receptor alpha Function. Sci Rep 2019; 9:545. [PMID: 30679625 PMCID: PMC6345901 DOI: 10.1038/s41598-018-36921-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 11/23/2018] [Indexed: 01/14/2023] Open
Abstract
The renal collecting duct consists of intercalated cells (ICs) and principal cells (PCs). We have previously demonstrated that collecting ducts have a role in the innate immune defense of the kidney. Transcriptomics is an important tool used to enhance systems-level understanding of cell biology. However, transcriptomics performed on whole kidneys provides limited insight of collecting duct cell gene expression, because these cells comprise a small fraction of total kidney cells. Recently we generated reporter mouse models to enrich collecting duct specific PC and ICs and reported targeted gene expression of anti-microbial peptide genes. Here we report transcriptomics on enriched ICs and PCs and performed a pilot study sequencing four single ICs. We identified 3,645 genes with increased relative expression in ICs compared to non-ICs. In comparison to non-PCs, 2,088 genes had higher relative expression in PCs. IC associated genes included the innate interleukin 1 receptor, type 1 and the antimicrobial peptide(AMP) adrenomedullin. The top predicted canonical pathway for enriched ICs was lipopolysaccharide/Interleukin 1 mediated inhibition of Retinoid X Receptor alpha function and decreased Retinoid X Receptor expression was confirmed to occur 1-hour post experimental murine UTI in ICs but not in non-ICs.
Collapse
Affiliation(s)
- Vijay Saxena
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States.
| | - James Fitch
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - John Ketz
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
| | - Peter White
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Amy Wetzel
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Melinda A Chanley
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
| | - John D Spencer
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Brian Becknell
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Keith R Pierce
- Innate Immunity Translational Research Center, Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, Tennessee, United States
| | - Sam W Arregui
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States
| | - Raoul D Nelson
- Division of Nephrology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - George J Schwartz
- University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, New York, United States
| | - Victoria Velazquez
- Research Institute at Nationwide Children's Hospital Flow Cytometry Core Laboratory, Columbus, Ohio, United States
| | - Logan A Walker
- Department of Physics, College of Arts and Sciences, The Ohio State University, Columbus, Ohio, United States
| | - Xi Chen
- Genomics Shared Resource, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Pearlly Yan
- Genomics Shared Resource, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - David S Hains
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States.
| | - Andrew L Schwaderer
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States.
| |
Collapse
|
20
|
Jung M, Dodsworth M, Thum T. Inflammatory cells and their non-coding RNAs as targets for treating myocardial infarction. Basic Res Cardiol 2018; 114:4. [PMID: 30523422 PMCID: PMC6290728 DOI: 10.1007/s00395-018-0712-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022]
Abstract
Myocardial infarction triggers infiltration of several types of immune cells that coordinate both innate and adaptive immune responses. These play a dual role in post-infarction cardiac remodeling by initiating and resolving inflammatory processes, which needs to occur in a timely and well-orchestrated way to ensure a reestablishment of normalized cardiac functions. Thus, therapeutic modulation of immune responses might have benefits for infarct patients. While such strategies have shown great potential in treating cancer, applications in the post-infarction context have been disappointing. One challenge has been the complexity and plasticity of immune cells and their functions in cardiac regulation and healing. The types appear in patterns that are temporally and spatially distinct, while influencing each other and the surrounding tissue. A comprehensive understanding of the immune cell repertoire and their regulatory functions following infarction is sorely needed. Processes of cardiac remodeling trigger additional genetic changes that may also play critical roles in the aftermath of cardiovascular disease. Some of these changes involve non-coding RNAs that play crucial roles in the regulation of immune cells and may, therefore, be of therapeutic interest. This review summarizes what is currently known about the functions of immune cells and non-coding RNAs during post-infarction wound healing. We address some of the challenges that remain and describe novel therapeutic approaches under development that are based on regulating immune responses through non-coding RNAs in the aftermath of the disease.
Collapse
Affiliation(s)
- Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Michael Dodsworth
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
21
|
Muccioli M, Nandigam H, Loftus T, Singh M, Venkatesh A, Wright J, Pate M, McCall K, Benencia F. Modulation of double-stranded RNA pattern recognition receptor signaling in ovarian cancer cells promotes inflammatory queues. Oncotarget 2018; 9:36666-36683. [PMID: 30613350 PMCID: PMC6291178 DOI: 10.18632/oncotarget.26378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/24/2018] [Indexed: 12/24/2022] Open
Abstract
Inflammation and cancer are inter-related, and both pro- and anti-tumorigenic effects are possible in different contexts, highlighting the importance of characterizing specific inflammatory pathways in distinct tumor types. Malignant cells and non-cancerous cells such as fibroblasts, infiltrating leukocytes (i.e., dendritic cells [DC], macrophages, or lymphocytes) and endothelial cells, in combination with the extracellular matrix, constitute the tumor microenvironment (TME). In the last decades, the role of the TME in cancer progression has gained increased attention and efforts directed at abrogating its deleterious effects on anti-cancer therapies have been ongoing. In this context, we investigated the potential of mouse and human ovarian cancer cells to produce inflammatory factors in response to pathogen recognition receptor (PRR) signaling, which might help to shape the biology of the TME. We determined that mouse ovarian tumors generate chemokines that are able to interact with receptors harbored by tumor-associated DCs. We also found that dsRNA triggers significant pro-inflammatory cytokine up-regulation in both human and mouse ovarian tumor cell lines, and that several PRR can simultaneously contribute to the stimulated inflammatory response displayed by these cells. Thus, dsRNA-activated PRRs may not only constitute potentially relevant drug targets for therapies aiming to prevent inflammation associated with leukocyte recruitment, or as co-adjuvants of therapeutic treatments, but also might have a role in development of nascent tumors, for example via activation of cancer cells by microbial molecules associated to pathogens, or with those appearing in circulation due to dysbiosis.
Collapse
Affiliation(s)
- Maria Muccioli
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH, 45701, USA
| | - Harika Nandigam
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.,Biomedical Engineering Program, Russ College of Engineering & Technology, Ohio University, Athens, OH, 45701, USA
| | - Tiffany Loftus
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Manindra Singh
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH, 45701, USA
| | - Amritha Venkatesh
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.,Biomedical Engineering Program, Russ College of Engineering & Technology, Ohio University, Athens, OH, 45701, USA
| | - Julia Wright
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Michelle Pate
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Kelly McCall
- Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH, 45701, USA.,Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.,Diabetes Institute at Ohio University, Ohio University, Athens, OH, 45701, USA.,Biomedical Engineering Program, Russ College of Engineering & Technology, Ohio University, Athens, OH, 45701, USA.,Translational Biomedical Sciences Doctoral Program, Ohio University, Athens, OH, 45701, USA
| | - Fabian Benencia
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH, 45701, USA.,Diabetes Institute at Ohio University, Ohio University, Athens, OH, 45701, USA.,Biomedical Engineering Program, Russ College of Engineering & Technology, Ohio University, Athens, OH, 45701, USA.,Translational Biomedical Sciences Doctoral Program, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
22
|
Švajger U, Rožman P. Induction of Tolerogenic Dendritic Cells by Endogenous Biomolecules: An Update. Front Immunol 2018; 9:2482. [PMID: 30416505 PMCID: PMC6212600 DOI: 10.3389/fimmu.2018.02482] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022] Open
Abstract
The importance of microenvironment on dendritic cell (DC) function and development has been strongly established during the last two decades. Although DCs with general tolerogenic characteristics have been isolated and defined as a particular sub-population, it is predominantly their unequivocal biological plasticity, which allows for unparalleled responsiveness to environmental ques and shaping of their tolerogenic characteristics when interacting with tolerance-inducing biomolecules. Dendritic cells carry receptors for a great number of endogenous factors, which, after ligation, can importantly influence the development of their activation state. For this there is ample evidence merely by observation of DC characteristics isolated from various anatomical niches, e.g., the greater immunosuppressive potential of DCs isolated from intestine compared to conventional blood DCs. Endogenous biomolecules present in these environments most likely play a major role as a determinant of their phenotype and function. In this review, we will concisely summarize in what way various, tolerance-inducing endogenous factors influence DC biology, the development of their particular tolerogenic state and their subsequent actions in context of immune response inhibition and induction of regulatory T cells.
Collapse
Affiliation(s)
- Urban Švajger
- Department for Therapeutic Services, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Primož Rožman
- Department for Therapeutic Services, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| |
Collapse
|
23
|
Naderi-Meshkin H, Ahmadiankia N. Cancer metastasis versus stem cell homing: Role of platelets. J Cell Physiol 2018; 233:9167-9178. [PMID: 30105746 DOI: 10.1002/jcp.26937] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Abstract
One of the major obstacles in achieving a successful stem cell therapy is insufficient homing of transplanted cells. To overcome this obstacle, understanding the underlying mechanisms of stem cell homing is of obvious importance. Central to this review is the concept that cancer metastasis can be viewed as a role model to build up a comprehensive concept of stem cell homing. In this novel perspective, the prosurvival choices of the cancerous cells in the bloodstream, their arrest, extravasation, and proliferation at the secondary site can be exploited in favor of targeted stem cell homing. To date, tumor cells have been found to employ a wide variety of strategies to promote metastasis. One of these strategies is through their ability to activate platelets and subsequently activated platelets serve cancer cell survival and metastasis. Accordingly, in the first part of this review the roles of platelets in cancer metastasis as well as stem cell homing are discussed. Next, we provide some lessons learned from cancer metastasis in favor of developing strategies for improvement of stem cell homing with emphasis on the role of platelets. Based on direct or indirect evidence from metastasis, strategies such as manipulation of stem cells to enhance interaction with platelets, preconditioning-pretreatment of stem cells with platelets in vitro, and coinjection of both stem cells and platelets are proposed to improve stem cell homing.
Collapse
Affiliation(s)
- Hojjat Naderi-Meshkin
- Stem Cells and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Naghmeh Ahmadiankia
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Cancer Prevention Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
24
|
Gornati L, Zanoni I, Granucci F. Dendritic Cells in the Cross Hair for the Generation of Tailored Vaccines. Front Immunol 2018; 9:1484. [PMID: 29997628 PMCID: PMC6030256 DOI: 10.3389/fimmu.2018.01484] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/14/2018] [Indexed: 12/14/2022] Open
Abstract
Vaccines represent the discovery of utmost importance for global health, due to both prophylactic action to prevent infections and therapeutic intervention in neoplastic diseases. Despite this, current vaccination strategies need to be refined to successfully generate robust protective antigen-specific memory immune responses. To address this issue, one possibility is to exploit the high efficiency of dendritic cells (DCs) as antigen-presenting cells for T cell priming. DCs functional plasticity allows shaping the outcome of immune responses to achieve the required type of immunity. Therefore, the choice of adjuvants to guide and sustain DCs maturation, the design of multifaceted vehicles, and the choice of surface molecules to specifically target DCs represent the key issues currently explored in both preclinical and clinical settings. Here, we review advances in DCs-based vaccination approaches, which exploit direct in vivo DCs targeting and activation options. We also discuss the recent findings for efficient antitumor DCs-based vaccinations and combination strategies to reduce the immune tolerance promoted by the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Gornati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Ivan Zanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.,Division of Gastroenterology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Francesca Granucci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
25
|
Mo Z, Yu F, Han S, Yang S, Wu L, Li P, Jiao S. New peptide MY1340 revert the inhibition effect of VEGF on dendritic cells differentiation and maturation via blocking VEGF-NRP-1 axis and inhibit tumor growth in vivo. Int Immunopharmacol 2018; 60:132-140. [PMID: 29730556 DOI: 10.1016/j.intimp.2018.04.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/25/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
The development and clinical application of immunostimulatory therapy provides us a new and exciting strategy in cancer treatment of which the agents act on crucial receptors. Given the fact that Neuropilin-1(NRP-1) is essential for vascular endothelial growth factor (VEGF) to inhibit LPS-dependent maturation of dendritic cells (DCs), it may present a potentially meaningful target in cancer immunotherapy. To explore this hypothesis, we synthesized a novel polypeptide called MY1340 consist of 32 amino acids with the aim of targeting VEGF-NRP-1 axis. Pull-down assay coupled with liquid chromatography-tandem mass spectrometry analysis (LC-MS/MS) was firstly conducted to identify NRP-1 as a potential MY1340 interacting protein, and the interaction between them was further confirmed by western blot. The competitive enzyme-linked immunosorbent assay (ELISA) results revealed that MY1340 was able to inhibit the binding between NRP-1 and VEGF with IC50 7.42 ng/ml, better than that of Tuftsin, although a natural ligand reportedly specific for the NRP-1 receptor. The presence of VEGF significantly reduced the expression of human leukocyte antigen-DR (HLA-DR), CD86 and CD11C on DCs, and this effect was reverted by MY1340-augment p65 NF-κB and ERK1/2 phosphorylation. We also present evidence that MY1340 is remarkably efficacious in the treatment of mice bearing subcutaneous liver cancer and induced DC maturation in the tumor environment in vivo. Taken together, these results indicate that MY1340 may represent a potential efficient immune therapeutic compound within disease that are rich in VEGF.
Collapse
Affiliation(s)
- Zheng Mo
- PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, Beijing, People's Republic of China
| | - Fei Yu
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Su Han
- DC Bio Lab, Beijing, People's Republic of China
| | | | - Liangliang Wu
- PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, Beijing, People's Republic of China
| | - Peng Li
- PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, Beijing, People's Republic of China
| | - Shunchang Jiao
- PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, Beijing, People's Republic of China.
| |
Collapse
|
26
|
Zhang L, He D, Huang J, Deng Y, Weng R, Pan L, Deng N. The Immunogenicity and Immunoprotection of VBP3 Multi-epitope Vaccine Targeting Angiogenesis and Tumor Inhibition in Lung Cancer-Bearing Mice. Int J Pept Res Ther 2017. [DOI: 10.1007/s10989-017-9667-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
27
|
Liu JF, Deng WW, Chen L, Li YC, Wu L, Ma SR, Zhang WF, Bu LL, Sun ZJ. Inhibition of JAK2/STAT3 reduces tumor-induced angiogenesis and myeloid-derived suppressor cells in head and neck cancer. Mol Carcinog 2017; 57:429-439. [PMID: 29215754 DOI: 10.1002/mc.22767] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/19/2017] [Accepted: 11/20/2017] [Indexed: 12/14/2022]
Abstract
Angiogenesis is an essential event in tumor growth and metastasis, and immune system also contributes to the tumor evasion. Emerging evidences have suggested the bidirectional link between angiogenesis and immunosuppression. Myeloid-derived suppressor cell (MDSC) is a kind of immunosuppressive cells and plays an important role in this process. However, the actual regulatory mechanisms of angiogenesis and MDSCs in head and neck squamous cell carcinoma (HNSCC) were unclear. In this study, through analyzing the immunohistochemistry staining of human HNSCC tissue microarray, we found that the microvascular density (MVD) was significantly increased in HNSCC patients. We also characterized angiogenic factors p-STAT3, VEGFA, CK2, and MDSCs marker CD11b in HNSCC tissue array, and found the close expression correlation among these markers. To determine the role of JAK2/STAT3 pathway in tumor microenvironment of HNSCC, we utilized AG490 (an inhibitor of JAK2/STAT3) for further research. Results showed that inhibition of JAK2/STAT3 suppressed angiogenesis by decreasing VEGFA and HIF1-α both in vitro and vivo. Moreover, in HNSCC transgenic mouse model, inhibiting JAK2/STAT3 not only suppressed angiogenesis but also reduced MDSCs in the tumor microenvironment through suppressing VEGFA and CK2. Our findings demonstrated the close relationship between angiogenesis and MDSCs in HNSCC, and inhibition of JAK2/STAT3 could reduce tumor-induced angiogenesis and decrease MDSCs.
Collapse
Affiliation(s)
- Jian-Feng Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei-Wei Deng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lei Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi-Cun Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lei Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Si-Rui Ma
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wen-Feng Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Hundsberger T, Reardon DA, Wen PY. Angiogenesis inhibitors in tackling recurrent glioblastoma. Expert Rev Anticancer Ther 2017; 17:507-515. [PMID: 28438066 DOI: 10.1080/14737140.2017.1322903] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Despite aggressive multimodality treatment of glioblastoma, outcome remains poor and patients mostly die of local recurrences. Besides reoperation and occasionally reirradiation, systemic treatment of recurrent glioblastoma consists of alkylating chemotherapy (lomustine, temozolomide), bevacizumab and combinations thereof. Unfortunately, antiangiogenic agents failed to improve survival either as a monotherapy or in combination treatments. This review provides current insights into tumor-derived escape mechanisms and other areas of treatment failure of antiangiogenic agents in glioblastoma. Areas covered: We summarize the current literature on antiangiogenic agents in the treatment of glioblastoma, with a focus on recurrent disease. A literature search was performed using the terms 'glioblastoma', 'bevacizumab', 'antiangiogenic', 'angiogenesis', 'resistance', 'radiotherapy', 'chemotherapy' and derivations thereof. Expert commentary: New insights in glioma neoangiogenesis, increasing understanding of vascular pathway escape mechanisms, and upcoming immunotherapy approaches might revitalize the therapeutic potential of antiangiogenic agents against glioblastoma, although with a different treatment intention. The combination of antiangiogenic approaches with or without radiotherapy might still hold promise to complement the therapeutic armamentarium of fighting glioblastoma.
Collapse
Affiliation(s)
- Thomas Hundsberger
- a Department of Neurology and Department of Hematology /Oncology , Cantonal hospital , St. Gallen , Switzerland
| | - David A Reardon
- b Center for Neuro-Oncology , Dana-Farber Cancer Institute /Brigham and Women's Cancer Center , Boston , MA , USA
| | - Patrick Y Wen
- b Center for Neuro-Oncology , Dana-Farber Cancer Institute /Brigham and Women's Cancer Center , Boston , MA , USA
| |
Collapse
|
29
|
Biophysical Properties and Motility of Human Mature Dendritic Cells Deteriorated by Vascular Endothelial Growth Factor through Cytoskeleton Remodeling. Int J Mol Sci 2016; 17:ijms17111756. [PMID: 27809226 PMCID: PMC5133777 DOI: 10.3390/ijms17111756] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs), the most potent antigen-presenting cells, play a central role in the initiation, regulation, and maintenance of the immune responses. Vascular endothelial growth factor (VEGF) is one of the important cytokines in the tumor microenvironment (TME) and can inhibit the differentiation and functional maturation of DCs. To elucidate the potential mechanisms of DC dysfunction induced by VEGF, the effects of VEGF on the biophysical characteristics and motility of human mature DCs (mDCs) were investigated. The results showed that VEGF had a negative influence on the biophysical properties, including electrophoretic mobility, osmotic fragility, viscoelasticity, and transmigration. Further cytoskeleton structure analysis by confocal microscope and gene expression profile analyses by gene microarray and real-time PCR indicated that the abnormal remodeling of F-actin cytoskeleton may be the main reason for the deterioration of biophysical properties, motility, and stimulatory capability of VEGF-treated mDCs. This is significant for understanding the biological behavior of DCs and the immune escape mechanism of tumors. Simultaneously, the therapeutic efficacies may be improved by blocking the signaling pathway of VEGF in an appropriate manner before the deployment of DC-based vaccinations against tumors.
Collapse
|
30
|
Zhang CM, Lv JF, Gong L, Yu LY, Chen XP, Zhou HH, Fan L. Role of Deficient Mismatch Repair in the Personalized Management of Colorectal Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13090892. [PMID: 27618077 PMCID: PMC5036725 DOI: 10.3390/ijerph13090892] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/01/2016] [Accepted: 09/05/2016] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) represents the third most common type of cancer in developed countries and one of the leading causes of cancer deaths worldwide. Personalized management of CRC has gained increasing attention since there are large inter-individual variations in the prognosis and response to drugs used to treat CRC owing to molecular heterogeneity. Approximately 15% of CRCs are caused by deficient mismatch repair (dMMR) characterized by microsatellite instability (MSI) phenotype. The present review is aimed at highlighting the role of MMR status in informing prognosis and personalized treatment of CRC including adjuvant chemotherapy, targeted therapy, and immune checkpoint inhibitor therapy to guide the individualized therapy of CRC.
Collapse
Affiliation(s)
- Cong-Min Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| | - Jin-Feng Lv
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China.
- Institute of Hospital Pharmacy, Central South University, Changsha 410008, China.
| | - Liang Gong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| | - Lin-Yu Yu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| | - Lan Fan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| |
Collapse
|
31
|
Duletić-Načinović A, Gačić V, Valković T, Lučin K, Fišić E, Žuvić-Butorac M, Seili-Bekafigo I, Jonjić N. Concurrent Elevations of VEGF, Osteopontin and MCP-1 Serum Levels Are Independent Predictors of Survival in Patients with Diffuse Large B-Cell Lymphoma. Acta Haematol 2016; 136:52-61. [PMID: 27160311 DOI: 10.1159/000444624] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 02/10/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Diffuse large B-cell lymphomas (DLBCL) are heterogeneous diseases, and the identification of additional DLBCL risk factors is especially important. METHODS In this pilot study, we determined pretreatment serum levels of vascular endothelial growth factor (VEGF), osteopontin (OPN) and macrophage chemotactic protein-1 (MCP-1) in 67 newly diagnosed DLBCL patients before treatment with standard chemoimmunotherapy and in 30 healthy persons. RESULTS Serum levels of all three cytokines were significantly elevated in untreated patients compared to controls. VEGF and OPN concentrations were higher in patients with advanced Ann Arbor stage, B symptoms, Eastern Cooperative Oncology Group score ≥2, International Prognostic Index (IPI) ≥3 and partial/no remission. A high MCP-1 level was associated with advanced stage, increased IPI and bone marrow infiltration. In univariate analysis, elevated OPN and VEGF, and concurrent elevation of all three biomarkers, were identified as significant predictors of poor survival. Multivariate Cox analysis revealed that elevated OPN combined with elevated VEGF levels was one of the best parameter subsets predicting poorest survival. CONCLUSION According to our preliminary results, serum levels of VEGF and OPN before treatment predict response to therapy and survival after chemoimmunotherapy, and may help to further stratify DLBCL patients into risk groups.
Collapse
|
32
|
Immunological aspects of antitumor photodynamic therapy outcome. Cent Eur J Immunol 2016; 40:481-5. [PMID: 26862314 PMCID: PMC4737746 DOI: 10.5114/ceji.2015.56974] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/16/2015] [Indexed: 12/03/2022] Open
Abstract
Photodynamic therapy (PDT) of cancer is an efficient and promising therapeutic modality approved for the treatment of several types of tumors and non-malignant diseases. It involves administration of a non-toxic photosensitizer followed by illumination of the tumor site with a harmless visible light. A light activated photosensitizer can transfer its energy directly to molecular oxygen, leading to production of highly toxic reactive oxygen species (ROS). Antitumor effects of PDT result from the combination of three independent mechanisms involving direct cytotoxicity to tumor cells, destruction of tumor vasculature and induction of the acute local inflammatory response. PDT-mediated inflammatory reaction is accompanied by tumor infiltration of the leukocytes, enhanced production of pro-inflammatory factors and cytokines. Photodynamic therapy is able to effectively stimulate both the innate and the adaptive arm of the immune system. In consequence, this regimen can lead to development of systemic and specific antitumor immune response. However, there are limited studies suggesting that under some specific circumstances, PDT on its own may exert some immunosuppressive effects leading to activation of immunosuppressive cells or cytokines production. In this report we briefly review all immunological aspects of PDT treatment.
Collapse
|
33
|
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
|
34
|
Anzengruber F, Avci P, de Freitas LF, Hamblin MR. T-cell mediated anti-tumor immunity after photodynamic therapy: why does it not always work and how can we improve it? Photochem Photobiol Sci 2015; 14:1492-1509. [PMID: 26062987 PMCID: PMC4547550 DOI: 10.1039/c4pp00455h] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Photodynamic therapy (PDT) uses the combination of non-toxic photosensitizers and harmless light to generate reactive oxygen species that destroy tumors by a combination of direct tumor cell killing, vascular shutdown, and activation of the immune system. It has been shown in some animal models that mice that have been cured of cancer by PDT, may exhibit resistance to rechallenge. The cured mice can also possess tumor specific T-cells that recognize defined tumor antigens, destroy tumor cells in vitro, and can be adoptively transferred to protect naïve mice from cancer. However, these beneficial outcomes are the exception rather than the rule. The reasons for this lack of consistency lie in the ability of many tumors to suppress the host immune system and to actively evade immune attack. The presence of an appropriate tumor rejection antigen in the particular tumor cell line is a requisite for T-cell mediated immunity. Regulatory T-cells (CD25+, Foxp3+) are potent inhibitors of anti-tumor immunity, and their removal by low dose cyclophosphamide can potentiate the PDT-induced immune response. Treatments that stimulate dendritic cells (DC) such as CpG oligonucleotide can overcome tumor-induced DC dysfunction and improve PDT outcome. Epigenetic reversal agents can increase tumor expression of MHC class I and also simultaneously increase expression of tumor antigens. A few clinical reports have shown that anti-tumor immunity can be generated by PDT in patients, and it is hoped that these combination approaches may increase tumor cures in patients.
Collapse
Affiliation(s)
- Florian Anzengruber
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Pinar Avci
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Dermatooncology and Venerology, Semmelweis University School of Medicine, Budapest, 1085, Hungary
| | - Lucas Freitas de Freitas
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Programa de Pos Graduacao Interunidades Bioengenharia – USP – Sao Carlos, Brazil
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- Correspondence to: Michael R Hamblin, PhD, Wellman Center for Photomedicine, Massachusetts General Hospital, 50 Blossom Street, Boston, MA 02114, USA.
| |
Collapse
|
35
|
Harnessing the PD-1 pathway in renal cell carcinoma: current evidence and future directions. BioDrugs 2015; 28:513-26. [PMID: 25445176 DOI: 10.1007/s40259-014-0111-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Programmed cell death-1 (PD-1) is a recognized immune checkpoint. It is frequently upregulated on the T cells that infiltrate tumors, providing an inhibitory signal, which may facilitate immune escape. Blocking antibodies have been developed to interrupt the interaction of PD-1 with its ligands PD-L1/PD-L2, with the goal of increasing the host antitumor immune response. Initial results have been encouraging, with durable responses in both treatment-naive and pretreated patients, along with an acceptable toxicity profile. This tolerability makes PD-1 blockade an excellent potential partner for combination strategies with the approved targeted agents, such as tyrosine kinase inhibitors (TKIs) and anti-vascular endothelial growth factor (anti-VEGF) antibodies, as well as other investigational immune checkpoint inhibitors or agonist antibodies that may costimulate an immune response. PD-L1 expression on tumor cells and tumor-infiltrating immune cells is also being evaluated as a predictive biomarker of response to treatment. This review summarizes the biological basis, preclinical studies, ongoing trials, and future challenges associated with targeting the PD-1 pathway in renal cell carcinoma.
Collapse
|
36
|
WANG JING, TAO YUMEI, CHENG XIAOYAN, ZHU TIANFENG, CHEN ZHIFANG, YAO HUI, SU LIANGXIANG. Vascular endothelial growth factor affects dendritic cell activity in hypertensive disorders of pregnancy. Mol Med Rep 2015; 12:3781-3786. [DOI: 10.3892/mmr.2015.3783] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 03/26/2015] [Indexed: 11/06/2022] Open
|
37
|
Papaspyridonos M, Matei I, Huang Y, do Rosario Andre M, Brazier-Mitouart H, Waite JC, Chan AS, Kalter J, Ramos I, Wu Q, Williams C, Wolchok JD, Chapman PB, Peinado H, Anandasabapathy N, Ocean AJ, Kaplan RN, Greenfield JP, Bromberg J, Skokos D, Lyden D. Id1 suppresses anti-tumour immune responses and promotes tumour progression by impairing myeloid cell maturation. Nat Commun 2015; 6:6840. [PMID: 25924227 PMCID: PMC4423225 DOI: 10.1038/ncomms7840] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/04/2015] [Indexed: 12/15/2022] Open
Abstract
A central mechanism of tumour progression and metastasis involves the generation of an immunosuppressive ‘macroenvironment' mediated in part through tumour-secreted factors. Here we demonstrate that upregulation of the Inhibitor of Differentiation 1 (Id1), in response to tumour-derived factors, such as TGFβ, is responsible for the switch from dendritic cell (DC) differentiation to myeloid-derived suppressor cell expansion during tumour progression. Genetic inactivation of Id1 largely corrects the myeloid imbalance, whereas Id1 overexpression in the absence of tumour-derived factors re-creates it. Id1 overexpression leads to systemic immunosuppression by downregulation of key molecules involved in DC differentiation and suppression of CD8 T-cell proliferation, thus promoting primary tumour growth and metastatic progression. Furthermore, advanced melanoma patients have increased plasma TGFβ levels and express higher levels of ID1 in myeloid peripheral blood cells. This study reveals a critical role for Id1 in suppressing the anti-tumour immune response during tumour progression and metastasis. Tumour progression is promoted by the generation of an immunosuppressive macroenvironment. Here, the authors demonstrate that the Inhibitor of Differentiation 1 promotes the switch from dendritic cell differentiation towards myeloid-derived suppressor cell expansion during tumour progression.
Collapse
Affiliation(s)
- Marianna Papaspyridonos
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | - Yujie Huang
- 1] Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA [2] Department of Neurosurgery, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Maria do Rosario Andre
- 1] Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA [2] Department of Genetics, Oncology and Human Toxicology, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisbon, Portugal
| | - Helene Brazier-Mitouart
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | | | - April S Chan
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | - Julie Kalter
- Regeneron Pharmaceuticals, Tarrytown, New York 10591, USA
| | - Ilyssa Ramos
- Regeneron Pharmaceuticals, Tarrytown, New York 10591, USA
| | - Qi Wu
- Regeneron Pharmaceuticals, Tarrytown, New York 10591, USA
| | - Caitlin Williams
- Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA
| | - Jedd D Wolchok
- 1] Melanoma and Immunotherapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA [2] Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA
| | - Paul B Chapman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA
| | - Hector Peinado
- 1] Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA [2] Tumor Metastasis Laboratory, Fundación Centro Nacional de Investigaciones Oncológicas, Calle Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Niroshana Anandasabapathy
- Brigham and Women's Hospital, Department of Dermatology, Harvard Medical School, 221 Longwood Avenue EBRC, Room 513, Boston, Massachusetts 02118, USA
| | - Allyson J Ocean
- Department of Medicine, Weill Cornell Medical College and Medical Oncology/Solid Tumor Program, 1305 York Avenue, New York City, New York 10021, USA
| | - Rosandra N Kaplan
- Center for Cancer Research, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10-Hatfield CRC, Room 1-3940, Bethesda, Maryland 20892, USA
| | - Jeffrey P Greenfield
- Department of Neurosurgery, Weill Cornell Medical College, 1300 York Avenue, New York City, New York 10065, USA
| | - Jacqueline Bromberg
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA
| | | | - David Lyden
- 1] Children's Cancer and Blood Foundation Laboratories and Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medical College, 413 East 69th Street, New York City, New York 10021, USA [2] Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York City, New York 10065, USA
| |
Collapse
|
38
|
Immune effects of bevacizumab: killing two birds with one stone. CANCER MICROENVIRONMENT 2014; 8:15-21. [PMID: 25326055 DOI: 10.1007/s12307-014-0160-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 10/14/2014] [Indexed: 12/31/2022]
Abstract
Angiogenesis or new vessel formation is essential for tumour growth and progression. Therefore, targeting angiogenesis has been an attractive strategy in the treatment ofcancer. Bevacizumab is a recombinant humanized monoclonal IgG1 antibody thattargets vascular endothelial growth factor-A (VEGF-A) - a key molecular player inangiogenesis. Bevacizumumab has shown clinical efficacy in phase III clinical trials inseveral advanced solid malignancies. The clinical efficacy of bevacizumumab isprimarily due to its antiangiogenic effects; however, there are direct antitumor effectsand immunomodulatory effects. Enhancing the immune system to restore itsantitumour activity has been utilized successfully in clinical setting. In this article we willdiscuss the possible immunomodulatory effects of the most clinically usedantiangiogenic agent; bevacizumumab.
Collapse
|
39
|
Sun LX, Lin ZB, Duan XS, Qi HH, Yang N, Li M, Xing EH, Sun Y, Yu M, Li WD, Lu J. Suppression of the Production of Transforming Growth Factor β1, Interleukin-10, and Vascular Endothelial Growth Factor in the B16F10 Cells byGanoderma lucidumPolysaccharides. J Interferon Cytokine Res 2014; 34:667-75. [PMID: 24673200 DOI: 10.1089/jir.2012.0101] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Li-Xin Sun
- The Affiliated Hospital of Chengde Medical College, Chengde, Hebei Province, China
| | - Zhi-Bin Lin
- Department of Pharmacology, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xin-Suo Duan
- The Affiliated Hospital of Chengde Medical College, Chengde, Hebei Province, China
| | - Hai-Hua Qi
- The Affiliated Hospital of Chengde Medical College, Chengde, Hebei Province, China
| | - Ning Yang
- The Affiliated Hospital of Chengde Medical College, Chengde, Hebei Province, China
| | - Min Li
- Department of Pharmacology, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China
| | - En-Hong Xing
- The Affiliated Hospital of Chengde Medical College, Chengde, Hebei Province, China
| | - Yu Sun
- The Affiliated Hospital of Chengde Medical College, Chengde, Hebei Province, China
| | - Min Yu
- The Affiliated Hospital of Chengde Medical College, Chengde, Hebei Province, China
| | - Wei-Dong Li
- Department of Pharmacology, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jie Lu
- The Affiliated Hospital of Chengde Medical College, Chengde, Hebei Province, China
| |
Collapse
|
40
|
Shi Y, Yu P, Zeng D, Qian F, Lei X, Zhao Y, Tang B, Hao Y, Luo H, Chen J, Tan Y. Suppression of vascular endothelial growth factor abrogates the immunosuppressive capability of murine gastric cancer cells and elicits antitumor immunity. FEBS J 2014; 281:3882-93. [PMID: 25041128 DOI: 10.1111/febs.12923] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/29/2014] [Accepted: 07/08/2014] [Indexed: 01/21/2023]
Abstract
The mechanisms underlying immune evasion by gastric cancer have not been well described due to a lack of gastric tumor models in immunocompetent mice. In the current study, we found that supernatants from MFC cells, a murine gastric cancer line, inhibited the lipopolysaccharide (LPS) induced maturation and cross-presentation of bone-marrow-derived dendritic cells (BMDCs). Moreover, MFC tumor-derived factors markedly altered the cytokine profiles of BMDCs, leading to a trend of increased levels of interleukin 4 (IL4), IL6, IL23 and transforming growth factor β, as well as decreased levels of tumor necrosis factor α. qPCR and ELISA revealed that MFC cells expressed a high level of vascular endothelial growth factor (VEGF). Downregulating VEGF expression abrogated the inhibitory effect of MFC-derived factors on the maturation and cross-presentation of BMDCs. In addition, VEGF knockdown greatly impaired the tumorigenicity of MFC cells in immunocompetent mice. Compared with parental MFC tumors, VEGF-low MFC tumors grew much more slowly and the survival of tumor-inoculated mice was significantly improved. More importantly, mice rejecting inoculated VEGF-low MFC tumor cells gained resistance to re-challenged parental tumors, which was attributed to an antitumor immunity response against parental MFC tumors. These results reveal an immunosuppressive role for VEGF in murine gastric cancer.
Collapse
Affiliation(s)
- Yan Shi
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Stockmann C, Schadendorf D, Klose R, Helfrich I. The impact of the immune system on tumor: angiogenesis and vascular remodeling. Front Oncol 2014; 4:69. [PMID: 24782982 PMCID: PMC3986554 DOI: 10.3389/fonc.2014.00069] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/20/2014] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, as well as inflammation with massive infiltration of leukocytes are hallmarks of various tumor entities. Various epidemiological, clinical, and experimental studies have not only demonstrated a link between chronic inflammation and cancer onset but also shown that immune cells from the bone marrow such as tumor-infiltrating macrophages significantly influence tumor progression. Tumor angiogenesis is critical for tumor development as tumors have to establish a blood supply in order to progress. Although tumor cells were first believed to fuel tumor angiogenesis, numerous studies have shown that the tumor microenvironment and infiltrating immune cell subsets are important for regulating the process of tumor angiogenesis. These infiltrates involve the adaptive immune system including several types of lymphocytes as well as cells of the innate immunity such as macrophages, neutrophils, eosinophils, mast cells, dendritic cells, and natural killer cells. Besides their known immune function, these cells are now recognized for their crucial role in regulating the formation and the remodeling of blood vessels in the tumor. In this review, we will discuss for each cell type the mechanisms that regulate the vascular phenotype and its impact on tumor growth and metastasis.
Collapse
Affiliation(s)
- Christian Stockmann
- UMR 970, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale (INSERM) , Paris , France
| | - Dirk Schadendorf
- Skin Cancer Unit, Dermatology Department, Medical Faculty, University Duisburg-Essen , Essen , Germany
| | - Ralph Klose
- UMR 970, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale (INSERM) , Paris , France
| | - Iris Helfrich
- Skin Cancer Unit, Dermatology Department, Medical Faculty, University Duisburg-Essen , Essen , Germany
| |
Collapse
|
42
|
Zeng J, Yuan D, Liu H, Song Y. [Vascular normalization and cancer immunotherapy]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 17:273-6. [PMID: 24667268 PMCID: PMC6019376 DOI: 10.3779/j.issn.1009-3419.2014.03.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
免疫治疗是一种颇有前景的抗肿瘤策略。然而,肿瘤中的免疫抑制微环境阻碍了免疫治疗的发展。异常肿瘤血管造成的缺氧,使免疫细胞趋向免疫抑制。并且异常血管通过分泌生长因子及细胞因子,改变免疫细胞的增殖、分化及功能,最终形成免疫抑制的微环境。因此,有效的利用血管生成及肿瘤免疫之间的相互作用,适当的抑制血管形成,促进肿瘤血管正常化,可以改变肿瘤的免疫抑制微环境,成为改善免疫治疗的新策略。现就血管正常化与肿瘤免疫的关系,及二者的联合治疗进行综述。。
Collapse
Affiliation(s)
- Junli Zeng
- Graduate School, Nanfang Medical University, 510515 Guangzhou, China;Department of Respiratory Medicine, Nanjing General Hospital of Nanjing Command, Nanjing 210002, China
| | - Dongmei Yuan
- Department of Respiratory Medicine, Nanjing General Hospital of Nanjing Command, Nanjing 210002, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Nanjing General Hospital of Nanjing Command, Nanjing 210002, China
| | - Yong Song
- Department of Respiratory Medicine, Nanjing General Hospital of Nanjing Command, Nanjing 210002, China
| |
Collapse
|
43
|
Marti LC, Pavon L, Severino P, Sibov T, Guilhen D, Moreira-Filho CA. Vascular endothelial growth factor-A enhances indoleamine 2,3-dioxygenase expression by dendritic cells and subsequently impacts lymphocyte proliferation. Mem Inst Oswaldo Cruz 2013; 109:70-9. [PMID: 24141959 PMCID: PMC4005532 DOI: 10.1590/0074-0276130252] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 06/21/2013] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are antigen (Ag)-presenting cells that activate and stimulate
effective immune responses by T cells, but can also act as negative regulators of
these responses and thus play important roles in immune regulation. Pro-angiogenic
vascular endothelial growth factor (VEGF) has been shown to cause defective DC
differentiation and maturation. Previous studies have demonstrated that the addition
of VEGF to DC cultures renders these cells weak stimulators of Ag-specific T cells
due to the inhibitory effects mediated by VEGF receptor 1 (VEGFR1) and/or VEGFR2
signalling. As the enzyme indoleamine 2,3-dioxygenase (IDO) is recognised as an
important negative regulator of immune responses, this study aimed to investigate
whether VEGF affects the expression of IDO by DCs and whether VEGF-matured DCs
acquire a suppressor phenotype. Our results are the first to demonstrate that VEGF
increases the expression and activity of IDO in DCs, which has a suppressive effect
on Ag-specific and mitogen-stimulated lymphocyte proliferation. These mechanisms have
broad implications for the study of immunological responses and tolerance under
conditions as diverse as cancer, graft rejection and autoimmunity.
Collapse
Affiliation(s)
| | - Lorena Pavon
- Centro de Pesquisa Experimental, Instituto Israelita de Ensino e Pesquisa Albert Einstein
| | - Patricia Severino
- Centro de Pesquisa Experimental, Instituto Israelita de Ensino e Pesquisa Albert Einstein
| | - Tatiana Sibov
- Centro de Pesquisa Experimental, Instituto do Cérebro, Hospital Israelita Albert Einstein, São PauloSP, Brasil
| | - Daiane Guilhen
- Centro de Pesquisa Experimental, Instituto Israelita de Ensino e Pesquisa Albert Einstein
| | | |
Collapse
|
44
|
Virtej A, Løes SS, Berggreen E, Bletsa A. Localization and signaling patterns of vascular endothelial growth factors and receptors in human periapical lesions. J Endod 2013; 39:605-11. [PMID: 23611377 DOI: 10.1016/j.joen.2012.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 11/28/2012] [Accepted: 12/10/2012] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are key players in vasculogenesis and are also involved in pathologic conditions with bone destruction. Vasculogenesis is critical for disease progression, and bone resorption is a hallmark of apical periodontitis. However, the localization of VEGFs and VEGFRs and their gene signaling pathways in human apical periodontitis have not been thoroughly investigated. The aim of this study was to localize VEGFs and VEGFRs and analyze their gene expression as well as signaling pathways in human periapical lesions. METHODS Tissue was collected after endodontic surgery from patients diagnosed with chronic apical periodontitis. Periodontal ligament samples from extracted healthy wisdom teeth was also collected and used as control tissue. In lesion cryosections, VEGFs/VEGFRs were identified by immunohistochemistry/double immunofluorescence by using specific antibodies. A human VEGF signaling polymerase chain reaction array system was used for gene expression analysis comparing lesions with periodontal ligament samples. RESULTS The histologic evaluation revealed heterogeneous morphology of the periapical lesions with various degrees of inflammatory infiltrates. In the lesions, all investigated factors and receptors were identified in blood vessels and various immune cells. No lymphatic vessels were detected. Gene expression analysis revealed up-regulation of VEGF-A and VEGFR-3, although not significant. Phosphatidylinositol-3-kinases, protein kinase C, mitogen-activated protein kinases, and phospholipases, all known to be involved in VEGF-mediated angiogenic activity, were significantly up-regulated. CONCLUSIONS The cellular and vascular expressions of VEGFs and VEGFRs in chronic apical periodontitis, along with significant alterations of genes mediating VEGF-induced angiogenic responses, suggest ongoing vascular remodeling in established chronic periapical lesions.
Collapse
Affiliation(s)
- Anca Virtej
- Institute of Biomedicine, Faculty of Medicine-Dentistry, University of Bergen, Bergen, Norway
| | | | | | | |
Collapse
|
45
|
Heuvers ME, Aerts JG, Cornelissen R, Groen H, Hoogsteden HC, Hegmans JP. Patient-tailored modulation of the immune system may revolutionize future lung cancer treatment. BMC Cancer 2012; 12:580. [PMID: 23217146 PMCID: PMC3533940 DOI: 10.1186/1471-2407-12-580] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 11/15/2012] [Indexed: 12/16/2022] Open
Abstract
Cancer research has devoted most of its energy over the past decades on unraveling the control mechanisms within tumor cells that govern its behavior. From this we know that the onset of cancer is the result of cumulative genetic mutations and epigenetic alterations in tumor cells leading to an unregulated cell cycle, unlimited replicative potential and the possibility for tissue invasion and metastasis. Until recently it was often thought that tumors are more or less undetected or tolerated by the patient’s immune system causing the neoplastic cells to divide and spread without resistance. However, it is without any doubt that the tumor environment contains a wide variety of recruited host immune cells. These tumor infiltrating immune cells influence anti-tumor responses in opposing ways and emerges as a critical regulator of tumor growth. Here we provide a summary of the relevant immunological cell types and their complex and dynamic roles within an established tumor microenvironment. For this, we focus on both the systemic compartment as well as the local presence within the tumor microenvironment of late-stage non-small cell lung cancer (NSCLC), admitting that this multifaceted cellular composition will be different from earlier stages of the disease, between NSCLC patients. Understanding the paradoxical role that the immune system plays in cancer and increasing options for their modulation may alter the odds in favor of a more effective anti-tumor immune response. We predict that the future standard of care of lung cancer will involve patient-tailor-made combination therapies that associate (traditional) chemotherapeutic drugs and biologicals with immune modulating agents and in this way complement the therapeutic armamentarium for this disease.
Collapse
Affiliation(s)
- Marlies E Heuvers
- Department of Pulmonary Medicine, Erasmus Medical Center, Postbox 2040, 3000 CA, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
46
|
Riihijärvi S, Nurmi H, Holte H, Björkholm M, Fluge Ø, Pedersen LM, Rydström K, Jerkeman M, Eriksson M, Leppä S. High serum vascular endothelial growth factor level is an adverse prognostic factor for high-risk diffuse large B-cell lymphoma patients treated with dose-dense chemoimmunotherapy. Eur J Haematol 2012; 89:395-402. [DOI: 10.1111/ejh.12005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2012] [Indexed: 12/16/2022]
Affiliation(s)
| | - Heidi Nurmi
- Department of Oncology; Helsinki University Central Hospital; Helsinki; Finland
| | - Harald Holte
- Department of Oncology; Oslo University Hospital; Oslo; Norway
| | - Magnus Björkholm
- Department of Medicine; Karolinska University Hospital; Stockholm; Sweden
| | - Øystein Fluge
- Department of Oncology and Medical Physics; Haukeland University Hospital; Bergen; Norway
| | | | - Karin Rydström
- Department of Oncology; Lund University Hospital; Lund; Sweden
| | - Mats Jerkeman
- Department of Oncology; Lund University Hospital; Lund; Sweden
| | - Mikael Eriksson
- Department of Oncology; Lund University Hospital; Lund; Sweden
| | | |
Collapse
|
47
|
Bruneau S, Woda CB, Daly KP, Boneschansker L, Jain NG, Kochupurakkal N, Contreras AG, Seto T, Briscoe DM. Key Features of the Intragraft Microenvironment that Determine Long-Term Survival Following Transplantation. Front Immunol 2012; 3:54. [PMID: 22566935 PMCID: PMC3342046 DOI: 10.3389/fimmu.2012.00054] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/02/2012] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss how changes in the intragraft microenvironment serve to promote or sustain the development of chronic allograft rejection. We propose two key elements within the microenvironment that contribute to the rejection process. The first is endothelial cell proliferation and angiogenesis that serve to create abnormal microvascular blood flow patterns as well as local tissue hypoxia, and precedes endothelial-to-mesenchymal transition. The second is the overexpression of local cytokines and growth factors that serve to sustain inflammation and, in turn, function to promote a leukocyte-induced angiogenesis reaction. Central to both events is overexpression of vascular endothelial growth factor (VEGF), which is both pro-inflammatory and pro-angiogenic, and thus drives progression of the chronic rejection microenvironment. In our discussion, we focus on how inflammation results in angiogenesis and how leukocyte-induced angiogenesis is pathological. We also discuss how VEGF is a master control factor that fosters the development of the chronic rejection microenvironment. Overall, this review provides insight into the intragraft microenvironment as an important paradigm for future direction in the field.
Collapse
Affiliation(s)
- Sarah Bruneau
- The Division of Nephrology, Transplantation Research Center, Children's Hospital Boston Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev 2011; 30:83-95. [PMID: 21249423 DOI: 10.1007/s10555-011-9281-4] [Citation(s) in RCA: 245] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immune system regulates angiogenesis in cancer with both pro- and antiangiogenic activities. The induction of angiogenesis is mediated by tumor-associated macrophages and myeloid-derived suppressor cells (MDSC) which produce proinflammatory cytokines, endothelial growth factors (VEGF, bFGF…), and protease (MMP9) implicated in neoangiogenesis. Some cytokines (IL-6, IL-17…) activated Stat3 which also led to the production of VEGF and bFGF. In contrast, other cytokines (IFN, IL-12, IL-21, and IL-27) display an antiangiogenic activity. Recently, it has been shown that some antiangiogenic molecules alleviates immunosuppression associated with cancer by decreasing immunosuppressive cells (MDSC, regulatory T cells), immunosuppressive cytokines (IL-10, TGFβ), and inhibitory molecules on T cells (PD-1). Some of these broad effects may result from the ability of some antiangiogenic molecules, especially cytokines to inhibit the Stat3 transcription factor. The association often observed between angiogenesis and immunosuppression may be related to hypoxia which induces both neoangiogenesis via activation of HIF-1 and VEGF and favors the intratumor recruitment and differentiation of regulatory T cells and MDSC. Preliminary studies suggest that modulation of immune markers (intratumoral MDSC and IL-8, peripheral regulatory T cells…) may predict clinical response to antiangiogenic therapy. In preclinical models, a synergy has been observed between antiangiogenic molecules and immunotherapy which may be explained by an improvement of immune status in tumor-bearing mice after antiangiogenic therapy. In preclinical models, antiangiogenic molecules promoted intratumor trafficking of effector cells, enhance endogenous anti-tumor response, and synergyzed with immunotherapy protocols to cure established murine tumors. All these results warrant the development of clinical trials combining antiangiogenic drugs and immunotherapy.
Collapse
|
49
|
Jewett A, Tseng HC. Tumor induced inactivation of natural killer cell cytotoxic function; implication in growth, expansion and differentiation of cancer stem cells. J Cancer 2011; 2:443-57. [PMID: 21850212 PMCID: PMC3157021 DOI: 10.7150/jca.2.443] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Accepted: 08/05/2011] [Indexed: 12/20/2022] Open
Abstract
Accumulated evidence indicates that cytotoxic function of immune effectors is largely suppressed in the tumor microenvironment by a number of distinct effectors and their secreted factors. The aims of this review are to provide a rationale and a potential mechanism for immunosuppression in cancer and to demonstrate the significance of such immunosuppression in cellular differentiation and progression of cancer. To that end, we have recently shown that NK cells mediate significant cytotoxicity against primary oral squamous carcinoma stem cells (OSCSCs) as compared to their more differentiated oral squamous carcinoma cells (OSCCs). In addition, human embryonic stem cells (hESCs), Mesenchymal Stem Cells (hMSCs), dental pulp stem cells (hDPSCs) and induced pluripotent stem cells (hiPSCs) were all significantly more susceptible to NK cell mediated cytotoxicity than their differentiated counterparts or parental cells from which they were derived. We have also reported that inhibition of differentiation or reversion of cells to a less-differentiated phenotype by blocking NFκB or targeted knock down of COX2 in primary monocytes in vivo significantly augmented NK cell function. Total population of monocytes and those depleted of CD16(+) subsets were able to substantially prevent NK cell mediated lysis of OSCSCs, MSCs and DPSCs. Taken together, our results suggest that stem cells are significant targets of the NK cell cytotoxicity. The concept of split anergy in NK cells and its contribution to tissue repair and regeneration and in tumor resistance and progression will be discussed in this review.
Collapse
Affiliation(s)
- Anahid Jewett
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, and Division of Oral Biology and Medicine, UCLA School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | | |
Collapse
|
50
|
Mroz P, Hamblin MR. The immunosuppressive side of PDT. Photochem Photobiol Sci 2011; 10:751-8. [PMID: 21437314 PMCID: PMC3441049 DOI: 10.1039/c0pp00345j] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2010] [Accepted: 02/25/2011] [Indexed: 12/29/2022]
Abstract
Photodynamic therapy (PDT) is a promising novel therapeutic procedure for the management of a variety of solid tumors and many non-malignant diseases. PDT has been described as having a significant effect on the immune system, which may be either immunostimulatory or, in some circumstances, immunosuppressive. The immunosuppressive effects of PDT have nearly all been concerned with the suppression of the contact hypersensitivity reaction in mice. Here, we review the immunosuppressive aspects of PDT treatment and discuss some additional mechanisms that may be involved.
Collapse
Affiliation(s)
- Pawel Mroz
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA.
| | | |
Collapse
|