1
|
Nair R, Somasundaram V, Kuriakose A, Krishn SR, Raben D, Salazar R, Nair P. Deciphering T-cell exhaustion in the tumor microenvironment: paving the way for innovative solid tumor therapies. Front Immunol 2025; 16:1548234. [PMID: 40236693 PMCID: PMC11996672 DOI: 10.3389/fimmu.2025.1548234] [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: 12/19/2024] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
In solid tumors, the tumor microenvironment (TME) is a complex mix of tumor, immune, stromal cells, fibroblasts, and the extracellular matrix. Cytotoxic T lymphocytes (CTLs) constitute a fraction of immune cells that may infiltrate into the TME. The primary function of these T-cells is to detect and eliminate tumor cells. However, due to the immunosuppressive factors present in the TME primarily mediated by Myeloid-Derived Suppressor Cells (MDSCs), Tumor associated macrophages (TAMs), Cancer Associated Fibroblasts (CAFs) as well as the tumor cells themselves, T-cells fail to differentiate into effector cells or become dysfunctional and are unable to eliminate the tumor. In addition, chronic antigen stimulation within the TME also leads to a phenomenon, first identified in chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, where the T-cells become exhausted and lose their effector functions. Exhausted T-cells (Tex) are characterized by the presence of remarkably conserved inhibitory receptors, transcription and signaling factors and the downregulation of key effector molecules. Tex cells have been identified in various malignancies, including melanoma, colorectal and hepatocellular cancers. Recent studies have indicated novel strategies to reverse T-cell exhaustion. These include checkpoint inhibitor blockade targeting programmed cell death protein 1 (PD-1), T-cell immunoglobulin and mucin-domain containing-3 (Tim-3), cytotoxic T-lymphocyte associated protein 4 (CTLA-4), or combinations of different immune checkpoint therapies (ICTs) or combination of ICTs with cytokine co-stimulation. In this review, we discuss aspects of T-cell dysfunction within the TME with a focus on T-cell exhaustion. We believe that gaining insight into the mechanisms of T-cell exhaustion within the TME of human solid tumors will pave the way for developing therapeutic strategies to target and potentially re-invigorate exhausted T-cells in cancer.
Collapse
Affiliation(s)
- Reshmi Nair
- Syngene International Limited, Bengaluru, India
| | | | | | | | - David Raben
- Bicara Therapeutics, Boston, MA, United States
| | | | - Pradip Nair
- Syngene International Limited, Bengaluru, India
| |
Collapse
|
2
|
Liu T, Yao W, Sun W, Yuan Y, Liu C, Liu X, Wang X, Jiang H. Components, Formulations, Deliveries, and Combinations of Tumor Vaccines. ACS NANO 2024; 18:18801-18833. [PMID: 38979917 DOI: 10.1021/acsnano.4c05065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Tumor vaccines, an important part of immunotherapy, prevent cancer or kill existing tumor cells by activating or restoring the body's own immune system. Currently, various formulations of tumor vaccines have been developed, including cell vaccines, tumor cell membrane vaccines, tumor DNA vaccines, tumor mRNA vaccines, tumor polypeptide vaccines, virus-vectored tumor vaccines, and tumor-in-situ vaccines. There are also multiple delivery systems for tumor vaccines, such as liposomes, cell membrane vesicles, viruses, exosomes, and emulsions. In addition, to decrease the risk of tumor immune escape and immune tolerance that may exist with a single tumor vaccine, combination therapy of tumor vaccines with radiotherapy, chemotherapy, immune checkpoint inhibitors, cytokines, CAR-T therapy, or photoimmunotherapy is an effective strategy. Given the critical role of tumor vaccines in immunotherapy, here, we look back to the history of tumor vaccines, and we discuss the antigens, adjuvants, formulations, delivery systems, mechanisms, combination therapy, and future directions of tumor vaccines.
Collapse
Affiliation(s)
- Tengfei Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyan Yao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyu Sun
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yihan Yuan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Chen Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
3
|
Ash S, Askenasy N. Immunotherapy for neuroblastoma by hematopoietic cell transplantation and post-transplant immunomodulation. Crit Rev Oncol Hematol 2023; 185:103956. [PMID: 36893946 DOI: 10.1016/j.critrevonc.2023.103956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/14/2022] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroblastoma represents a relatively common childhood tumor that imposes therapeutic difficulties. High risk neuroblastoma patients have poor prognosis, display limited response to radiochemotherapy and may be treated by hematopoietic cell transplantation. Allogeneic and haploidentical transplants have the distinct advantage of reinstitution of immune surveillance, reinforced by antigenic barriers. The key factors favorable to ignition of potent anti-tumor reactions are transition to adaptive immunity, recovery from lymphopenia and removal of inhibitory signals that inactivate immune cells at the local and systemic levels. Post-transplant immunomodulation may further foster anti-tumor reactivity, with positive but transient impact of infusions of lymphocytes and natural killer cells both from the donor, the recipient or third party. The most promising approaches include introduction of antigen-presenting cells in early post-transplant stages and neutralization of inhibitory signals. Further studies will likely shed light on the nature and actions of suppressor factors within tumor stroma and at the systemic level.
Collapse
Affiliation(s)
- Shifra Ash
- Department of Pediatric Hematology-Oncology, Rambam Medical Center, Haifa, Israel; Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Nadir Askenasy
- Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
4
|
Papantoniou K, Castaño-Vinyals G, Espinosa A, Turner MC, Martín-Sánchez V, Casabonne D, Aragonés N, Gómez-Acebo I, Ardanaz E, Jimenez-Moleon JJ, Amiano P, Molina-Barceló A, Alguacil J, Fernández-Tardón G, Huerta JM, Hernández-Segura N, Perez-Gomez B, Llorca J, Vidán-Alli J, Olmedo-Requena R, Gil L, Castañon-López C, Pollan M, Kogevinas M, Moreno V. Sleep duration and napping in relation to colorectal and gastric cancer in the MCC-Spain study. Sci Rep 2021; 11:11822. [PMID: 34083698 PMCID: PMC8175745 DOI: 10.1038/s41598-021-91275-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep duration is a novel and potentially modifiable risk factor for cancer. We evaluated the association of self-reported sleep duration and daytime napping with odds of colorectal and gastric cancer. We included 2008 incident colorectal cancer cases, 542 gastric cancer cases and 3622 frequency-matched population controls, recruited in the MCC-Spain case-control study (2008-2013). Sleep information, socio-demographic and lifestyle characteristics were obtained through personal interviews. Multivariable adjusted logistic regression models were used to estimate odds ratios (OR) with 95% confidence intervals (CI) for cancer, across categories of sleep duration (≤ 5, 6, 7, 8, ≥ 9 hours/day), daytime napping frequency (naps/week) and duration (minutes/nap). Compared to 7 hours of sleep, long sleep was associated with increased odds of colorectal (OR≥9 hours: 1.59; 95%CI 1.30-1.94) and gastric cancer (OR≥9 hours: 1.95; 1.37-2.76); short sleep was associated with increased odds of gastric cancer (OR≤5 hours: 1.32; 0.93-1.88). Frequent and long daytime naps increased the odds of colorectal (OR6-7 naps/week, ≥30 min: 1.32; 1.14-1.54) and gastric cancer (OR6-7 naps/week, ≥30 min: 1.56; 1.21-2.02). Effects of short sleep and frequent long naps were stronger among participants with night shift-work history. Sleep and circadian disruption may jointly play a role in the etiology of colorectal and gastric cancer.
Collapse
Affiliation(s)
- Kyriaki Papantoniou
- Department of Epidemiology, Center of Public Health, Medical University of Vienna, Kinderspitalgasse 15, 1090, Vienna, Austria.
- ISGlobal, Barcelona, Spain.
| | - Gemma Castaño-Vinyals
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Ana Espinosa
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Michelle C Turner
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON, Canada
| | - Vicente Martín-Sánchez
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Biomedicine Institute (IBIOMED), University of León, Leon, Spain
| | - Delphine Casabonne
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Unit of Molecular and Genetic Epidemiology in Infections and Cancer (UNIC-Molecular), Cancer Epidemiology Research Programme, IDIBELL, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
| | - Nuria Aragonés
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Epidemiology Section, Public Health Division, Department of Health of Madrid, Madrid, Spain
| | - Inés Gómez-Acebo
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- University of Cantabria, Santander, Spain
- IDIVAL, Santander, Spain
| | - Eva Ardanaz
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Jose-Juan Jimenez-Moleon
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
- Instituto de Investigacion Biosanitaria de Granada (Ibs.GRANADA), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
| | - Pilar Amiano
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Public Health Division of Gipuzkoa, San Sebastian, Spain
- Biodonostia Research Institute, San Sebastian, Spain
| | | | - Juan Alguacil
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Centro de Investigación en Salud y Medio Ambiente (CYSMA), Universidad de Huelva, Campus Universitario de El Carmen, Huelva, Spain
| | - Guillermo Fernández-Tardón
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Health Research Institute of the Principality of Asturias, University of Oviedo, Oviedo, Spain
| | - José María Huerta
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
| | | | - Beatriz Perez-Gomez
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Cancer Epidemiology Unit of the National Center for Epidemiology, Carlos III Institute of Health, Madrid, Spain
- Cancer Epidemiology Research Group, Oncology and Hematology Area, IIS Puerta de Hierro (IDIPHIM), Majadahonda, Madrid, Spain
| | - Javier Llorca
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- University of Cantabria, Santander, Spain
| | - Juana Vidán-Alli
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Rocıo Olmedo-Requena
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
- Instituto de Investigacion Biosanitaria de Granada (Ibs.GRANADA), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
| | - Leire Gil
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Public Health Division of Gipuzkoa, San Sebastian, Spain
- Biodonostia Research Institute, San Sebastian, Spain
| | | | - Marina Pollan
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Cancer Epidemiology Unit of the National Center for Epidemiology, Carlos III Institute of Health, Madrid, Spain
| | - Manolis Kogevinas
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Victor Moreno
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Naseri M, Zöller M, Hadjati J, Ghods R, Ranaei Pirmardan E, Kiani J, Eini L, Bozorgmehr M, Madjd Z. Dendritic cells loaded with exosomes derived from cancer stem cell-enriched spheroids as a potential immunotherapeutic option. J Cell Mol Med 2021; 25:3312-3326. [PMID: 33634564 PMCID: PMC8034455 DOI: 10.1111/jcmm.16401] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/30/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are responsible for therapeutic resistance and recurrence in colorectal cancer. Despite advances in immunotherapy, the inability to specifically eradicate CSCs has led to treatment failure. Hence, identification of appropriate antigen sources is a major challenge in designing dendritic cell (DC)‐based therapeutic strategies against CSCs. Here, in an in vitro model using the HT‐29 colon cancer cell line, we explored the efficacy of DCs loaded with exosomes derived from CSC‐enriched colonospheres (CSCenr‐EXOs) as an antigen source in activating CSC‐specific T‐cell responses. HT‐29 lysate, HT‐29‐EXOs and CSCenr lysate were independently assessed as separate antigen sources. Having confirmed CSCs enrichment in spheroids, CSCenr‐EXOs were purified and characterized, and their impact on DC maturation was investigated. Finally, the impact of the antigen‐pulsed DCs on the proliferation rate and also spheroid destructive capacity of autologous T cells was assessed. CSCenr‐EXOs similar to other antigen groups had no suppressive/negative impacts on phenotypic maturation of DCs as judged by the expression level of costimulatory molecules. Notably, similar to CSCenr lysate, CSCenr‐EXOs significantly increased the IL‐12/IL‐10 ratio in supernatants of mature DCs. CSCenr‐EXO‐loaded DCs effectively promoted T‐cell proliferation. Importantly, T cells stimulated with CSCenr‐EXOs disrupted spheroids' structure. Thus, CSCenr‐EXOs present a novel and promising antigen source that in combination with conventional tumour bulk‐derived antigens should be further explored in pre‐clinical immunotherapeutic settings for the efficacy in hampering recurrence and metastatic spread.
Collapse
Affiliation(s)
- Marzieh Naseri
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Margot Zöller
- Section Pancreas Research, University Hospital of Surgery, Heidelberg, Germany
| | - Jamshid Hadjati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Ehsan Ranaei Pirmardan
- Department of Radiology, Molecular Biomarkers Nano-imaging Laboratory, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Leila Eini
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Basic Science, Faculty of Veterinary, Science and Research Branch of Islamic, Azad University, Tehran, Iran
| | - Mahmood Bozorgmehr
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
6
|
Iftikhar A, Islam M, Shepherd S, Jones S, Ellis I. Cancer and Stress: Does It Make a Difference to the Patient When These Two Challenges Collide? Cancers (Basel) 2021; 13:cancers13020163. [PMID: 33418900 PMCID: PMC7825104 DOI: 10.3390/cancers13020163] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Head and neck cancers are the sixth most common cancer in the world. The burden of the disease has remained challenging over recent years despite the advances in treatments of other malignancies. The very use of the word malignancy brings about a stress response in almost all adult patients. Being told you have a tumour is not a word anyone wants to hear. We have embarked on a study which will investigate the effect of stress pathways on head and neck cancer patients and which signalling pathways may be involved. In the future, this will allow clinicians to better manage patients with head and neck cancer and reduce the patients’ stress so that this does not add to their tumour burden. Abstract A single head and neck Cancer (HNC) is a globally growing challenge associated with significant morbidity and mortality. The diagnosis itself can affect the patients profoundly let alone the complex and disfiguring treatment. The highly important functions of structures of the head and neck such as mastication, speech, aesthetics, identity and social interactions make a cancer diagnosis in this region even more psychologically traumatic. The emotional distress engendered as a result of functional and social disruption is certain to negatively affect health-related quality of life (HRQoL). The key biological responses to stressful events are moderated through the combined action of two systems, the hypothalamus–pituitary–adrenal axis (HPA) which releases glucocorticoids and the sympathetic nervous system (SNS) which releases catecholamines. In acute stress, these hormones help the body to regain homeostasis; however, in chronic stress their increased levels and activation of their receptors may aid in the progression of cancer. Despite ample evidence on the existence of stress in patients diagnosed with HNC, studies looking at the effect of stress on the progression of disease are scarce, compared to other cancers. This review summarises the challenges associated with HNC that make it stressful and describes how stress signalling aids in the progression of cancer. Growing evidence on the relationship between stress and HNC makes it paramount to focus future research towards a better understanding of stress and its effect on head and neck cancer.
Collapse
|
7
|
Colon-Echevarria CB, Lamboy-Caraballo R, Aquino-Acevedo AN, Armaiz-Pena GN. Neuroendocrine Regulation of Tumor-Associated Immune Cells. Front Oncol 2019; 9:1077. [PMID: 31737559 PMCID: PMC6828842 DOI: 10.3389/fonc.2019.01077] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Mounting preclinical and clinical evidence continues to support a role for the neuroendocrine system in the modulation of tumor biology and progression. Several studies have shown data supporting a link between chronic stress and cancer progression. Dysregulation of the sympathetic nervous system (SNS) and the hypothalamic-pituitary-adrenal (HPA) axis has been implicated in promoting angiogenesis, tumor cell proliferation and survival, alteration of the immune response and exacerbating inflammatory networks in the tumor microenvironment. Here, we review how SNS and HPA dysregulation contributes to disturbances in immune cell populations, modifies cancer biology, and impacts immunotherapy response. We also highlight several interventions aimed at circumventing the adverse effects stress has on cancer patients.
Collapse
Affiliation(s)
- Claudia B Colon-Echevarria
- Division of Pharmacology, Department of Basic Sciences, School of Medicine, Ponce Health Sciences University, Ponce, PR, United States
| | - Rocio Lamboy-Caraballo
- Division of Pharmacology, Department of Basic Sciences, School of Medicine, Ponce Health Sciences University, Ponce, PR, United States
| | - Alexandra N Aquino-Acevedo
- Division of Pharmacology, Department of Basic Sciences, School of Medicine, Ponce Health Sciences University, Ponce, PR, United States
| | - Guillermo N Armaiz-Pena
- Division of Pharmacology, Department of Basic Sciences, School of Medicine, Ponce Health Sciences University, Ponce, PR, United States.,Divisions of Cancer Biology and Women's Health, Ponce Research Institute, Ponce, PR, United States
| |
Collapse
|
8
|
Oncolytic adenovirus coexpressing interleukin-12 and decorin overcomes Treg-mediated immunosuppression inducing potent antitumor effects in a weakly immunogenic tumor model. Oncotarget 2018; 8:4730-4746. [PMID: 28002796 PMCID: PMC5354867 DOI: 10.18632/oncotarget.13972] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/01/2016] [Indexed: 01/02/2023] Open
Abstract
Interleukin (IL)-12 is a potent antitumor cytokine. However, immunosuppressive tumor microenvironments containing transforming growth factor-β (TGF-β) attenuate cytokine-mediated antitumor immune responses. To enhance the efficacy of IL-12-mediated cancer immunotherapy, decorin (DCN) was explored as an adjuvant for overcoming TGF-β-mediated immunosuppression. We designed and generated a novel oncolytic adenovirus (Ad) coexpressing IL-12 and DCN (RdB/IL12/DCN). RdB/IL12/DCN-treated tumors showed significantly greater levels of interferon (IFN)-γ, tumor necrosis factor-α, monocyte chemoattractant protein-1, and IFN-γ-secreting immune cells than tumors treated with cognate control oncolytic Ad expressing a single therapeutic gene (RdB/DCN or RdB/IL12). Moreover, RdB/IL12/DCN attenuated intratumoral TGF-β expression, which positively correlated with reduction of Treg cells in draining lymph nodes and tumor tissues. Furthermore, tumor tissue treated with RdB/IL12/DCN showed increases infiltration of CD8+ T cells and proficient viral spreading within tumor tissues. These results demonstrated that an oncolytic Ad co-expressing IL-12 and DCN induces a potent antitumor immune response via restoration of antitumor immune function in a weakly immunogenic murine 4T1 orthotopic breast cancer model. These findings provide new insights into the therapeutic mechanisms of IL-12 plus DCN, making it a promising cancer immunotherapeutic agent for overcoming tumor-induced immunosuppression.
Collapse
|
9
|
Klein B, Schuppe HC, Bergmann M, Hedger MP, Loveland BE, Loveland KL. An in vitro model demonstrates the potential of neoplastic human germ cells to influence the tumour microenvironment. Andrology 2017; 5:763-770. [PMID: 28544640 DOI: 10.1111/andr.12365] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/10/2017] [Accepted: 03/18/2017] [Indexed: 12/17/2022]
Abstract
Testicular germ cell tumours (TGCT) typically contain high numbers of infiltrating immune cells, yet the functional nature and consequences of interactions between GCNIS (germ cell neoplasia in situ) or seminoma cells and immune cells remain unknown. A co-culture model using the seminoma-derived TCam-2 cell line and peripheral blood mononuclear cells (PBMC, n = 7 healthy donors) was established to investigate how tumour and immune cells each contribute to the cytokine microenvironment associated with TGCT. Three different co-culture approaches were employed: direct contact during culture to simulate in situ cellular interactions occurring within seminomas (n = 9); indirect contact using well inserts to mimic GCNIS, in which a basement membrane separates the neoplastic germ cells and immune cells (n = 3); and PBMC stimulation prior to direct contact during culture to overcome the potential lack of immune cell activation (n = 3). Transcript levels for key cytokines in PBMC and TCam-2 cell fractions were determined using RT-qPCR. TCam-2 cell fractions showed an immediate increase (within 24 h) in several cytokine mRNAs after direct contact with PBMC, whereas immune cell fractions did not. The high levels of interleukin-6 (IL6) mRNA and protein associated with TCam-2 cells implicate this cytokine as important to seminoma physiology. Use of PBMCs from different donors revealed a robust, repeatable pattern of changes in TCam-2 and PBMC cytokine mRNAs, independent of potential inter-donor variation in immune cell responsiveness. This in vitro model recapitulated previous data from clinical TGCT biopsies, revealing similar cytokine expression profiles and indicating its suitability for exploring the in vivo circumstances of TGCT. Despite the limitations of using a cell line to mimic in vivo events, these results indicate how neoplastic germ cells can directly shape the surrounding tumour microenvironment, including by influencing local immune responses. IL6 production by seminoma cells may be a practical target for early diagnosis and/or treatment of TGCT.
Collapse
Affiliation(s)
- B Klein
- Department of Anatomy and Cell Biology, Justus-Liebig University, Giessen, Germany
| | - H-C Schuppe
- Department of Urology, Pediatric Urology and Andrology, Justus-Liebig University, Giessen, Germany
| | - M Bergmann
- Department of Veterinary Anatomy, Histology and Embryology, Justus-Liebig University, Giessen, Germany
| | - M P Hedger
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | | | - K L Loveland
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash Medical Centre, Clayton, VIC, Australia
| |
Collapse
|
10
|
Agarwal R, Chaudhary M, Bohra S, Bajaj S. Evaluation of natural killer cell (CD57) as a prognostic marker in oral squamous cell carcinoma: An immunohistochemistry study. J Oral Maxillofac Pathol 2016; 20:173-7. [PMID: 27601804 PMCID: PMC4989542 DOI: 10.4103/0973-029x.185933] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objectives: Natural killer (NK) cells are important effector lymphocytes. NK cells are considered to represent innate immune system. NK cells target and kill aberrant cells such as virally infected and tumorigenic cells. The purpose of this study was to assess the expression of CD57 in oral squamous cell carcinoma (OSCC) and to correlate the expression of CD57 with 3 years survival in patients with OSCC. Materials and Methods: About 100 histopathologically diagnosed cases of OSCC of various grades were divided into two groups, i.e., Group I (dead patients) and Group II (live patients) from the archives of Department of Oral Pathology and Microbiology. CD57 was detected in these tissues by immunohistochemistry. Result: The results were analyzed using Spearman's correlation coefficient and students unpaired t-test. The mean CD57 labeling index in Group II was significantly higher than that found in Group I (P = 0.000). There was a significant correlation (P = 0.00) in the mean CD57 levels between Groups I and II and prognosis of patient. Conclusion: CD57 could be a good prognostic marker for OSCC patients.
Collapse
Affiliation(s)
- Rashmi Agarwal
- Department of Oral and Maxillofacial Pathology, Dr. B. R. Ambedkar Institute of Dental Sciences and Hospital, Patna, Bihar, India
| | - Minal Chaudhary
- Department of Oral and Maxillofacial Pathology and Microbiology, Sharad Pawar Dental College, Wardha, Maharashtra, India
| | - Shruti Bohra
- Department of Oral and Maxillofacial Pathology and Microbiology, Sharad Pawar Dental College, Wardha, Maharashtra, India
| | - Shree Bajaj
- Department of Oral and Maxillofacial Pathology and Microbiology, Sharad Pawar Dental College, Wardha, Maharashtra, India
| |
Collapse
|
11
|
Purwada A, Tian YF, Huang W, Rohrbach KM, Deol S, August A, Singh A. Self-Assembly Protein Nanogels for Safer Cancer Immunotherapy. Adv Healthc Mater 2016; 5:1413-9. [PMID: 27100566 PMCID: PMC5532174 DOI: 10.1002/adhm.201501062] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/04/2016] [Indexed: 01/15/2023]
Abstract
Soluble antigen-based cancer vaccines have poor retention in tissues along with suboptimal antigen processing by dendritic cells. Multiple booster doses are often needed, leading to dose-limiting systemic toxicity. A versatile, immunomodulatory, self-assembly protein nanogel vaccine is reported that induces robust immune cell response at lower antigen doses than soluble antigens, an important step towards biomaterials-based safer immunotherapy approaches.
Collapse
Affiliation(s)
- Alberto Purwada
- Meinig School of Biomedical Engineering, College of Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Ye F. Tian
- Sibley School of Mechanical and Aerospace Engineering, College of Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Weishan Huang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Kathleen M. Rohrbach
- Department of Material Science and Engineering, College of Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Simrita Deol
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Ankur Singh
- Sibley School of Mechanical and Aerospace Engineering, College of Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
12
|
Ramzan M, Sturm N, Decaens T, Bioulac-Sage P, Bancel B, Merle P, Tran Van Nhieu J, Slama R, Letoublon C, Zarski JP, Jouvin-Marche E, Marche PN, Leroy V. Liver-infiltrating CD8(+) lymphocytes as prognostic factor for tumour recurrence in hepatitis C virus-related hepatocellular carcinoma. Liver Int 2016. [PMID: 26215124 DOI: 10.1111/liv.12927] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Chronic liver inflammation and immune/inflammatory response promote hepatocellular carcinoma. The aim of this study was to characterize the immune status of HCV-related cirrhosis in patients with hepatocellular carcinoma (HCV-HCC) as compared to HCV patients without hepatocellular carcinoma. METHOD Immune markers (CD3, CD4, CD8, CD20, CD56, TCRγδ, FoxP3) and gene expression profiles (CD8α, CD8β, FoxP3, IL-6, IFN-γ, perforin, RANTES) were analysed in a test cohort by immunohistochemistry and quantitative RT-PCR analysis on serial non-tumorous and tumorous tissues. RESULTS Immune micro-environment was more inflammatory in HCV-HCC than HCV cirrhotic livers. The number of CD3(+) , CD4(+) , CD8(+) and CD20(+) liver-infiltrating lymphocytes was significantly higher, whereas the number of CD56(+) cells was significantly lower in HCV-HCC compared to HCV cirrhotic parenchyma. These differences were restricted to fibrous septa for CD4(+) and CD20(+) cells and to nodular parenchyma for CD8(+) cells. Gene expressions of CD8α, FoxP3 and RANTES were also significantly higher in HCV-HCC than in HCV cirrhosis. Interestingly, in a large cohort of 63 HCV-HCC patients. The number of CD8(+) cells ≥100/field was associated with significant higher tumour recurrence (P = 0.003) and lower overall survival (P = 0.05) at 5 years. CONCLUSION High densities of liver-infiltrating lymphocytes in HCV-HCC cirrhotic parenchyma prevail inflammatory conditions and could contribute to tumorigenesis and tumour recurrence. These results could contribute towards better clinical evaluation of patients susceptible for HCC recurrence after curative surgery.
Collapse
Affiliation(s)
- Muhammad Ramzan
- INSERM, Unité 823, Grenoble, France.,Université Grenoble-Alpes, Faculté de Médecine, UMR-S823, Grenoble, France
| | - Nathalie Sturm
- INSERM, Unité 823, Grenoble, France.,Université Grenoble-Alpes, Faculté de Médecine, UMR-S823, Grenoble, France.,Département d'Anatomie et de Cytologie Pathologiques, CHU de Grenoble, Grenoble, France
| | - Thomas Decaens
- INSERM, Unité 823, Grenoble, France.,Université Grenoble-Alpes, Faculté de Médecine, UMR-S823, Grenoble, France.,Clinique d' Hépatogastroentérologie, pôle DigiDune, CHU de Grenoble, Grenoble, France
| | - Paulette Bioulac-Sage
- Service d'Anatomie et de Cytologie Pathologiques, Hôpital Pellegrin, Bordeaux, France
| | - Brigitte Bancel
- Service de Pathologie, Hôpital de la Croix Rousse, Lyon, France.,Université Claude Bernard Lyon 1, INSERM UMR-S1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Hospices Civils de Lyon, Lyon, France
| | - Philippe Merle
- Université Claude Bernard Lyon 1, INSERM UMR-S1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Hospices Civils de Lyon, Lyon, France.,Service d'Hépatologie, Hôpital de la Croix-Rousse, Lyon, France
| | - Jeanne Tran Van Nhieu
- Département de Pathologie, APHP, Groupe Hospitalier Henri Mondor, Val de Marne, Université Paris Est-Créteil, Créteil, France
| | - Rémy Slama
- INSERM, Unité 823, Grenoble, France.,Université Grenoble-Alpes, Faculté de Médecine, UMR-S823, Grenoble, France
| | - Christian Letoublon
- INSERM, Unité 823, Grenoble, France.,Université Grenoble-Alpes, Faculté de Médecine, UMR-S823, Grenoble, France.,Clinique de Chirurgie Digestive, pôle DIGIDUNE, CHU de Grenoble, Grenoble, France
| | - Jean-Pierre Zarski
- INSERM, Unité 823, Grenoble, France.,Université Grenoble-Alpes, Faculté de Médecine, UMR-S823, Grenoble, France.,Clinique d' Hépatogastroentérologie, pôle DigiDune, CHU de Grenoble, Grenoble, France
| | - Evelyne Jouvin-Marche
- INSERM, Unité 823, Grenoble, France.,Université Grenoble-Alpes, Faculté de Médecine, UMR-S823, Grenoble, France
| | - Patrice N Marche
- INSERM, Unité 823, Grenoble, France.,Université Grenoble-Alpes, Faculté de Médecine, UMR-S823, Grenoble, France.,Clinique d' Hépatogastroentérologie, pôle DigiDune, CHU de Grenoble, Grenoble, France
| | - Vincent Leroy
- INSERM, Unité 823, Grenoble, France.,Université Grenoble-Alpes, Faculté de Médecine, UMR-S823, Grenoble, France.,Clinique d' Hépatogastroentérologie, pôle DigiDune, CHU de Grenoble, Grenoble, France
| |
Collapse
|
13
|
McCormick AA, Palmer KE. Genetically engineered Tobacco mosaic virus as nanoparticle vaccines. Expert Rev Vaccines 2014; 7:33-41. [DOI: 10.1586/14760584.7.1.33] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
14
|
Ridolfi R, Ridolfi L, Petrini M, Fiammenghi L, Riccobon A. Dendritic cell vaccination and immunostimulation in advanced melanoma. Expert Rev Vaccines 2014; 2:825-33. [PMID: 14711365 DOI: 10.1586/14760584.2.6.825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The most recent advances in immunology bear witness to the fact that tumors, in particular melanoma, escape recognition by the host's immune system and can locally inactivate its effectors, T-cells and antigen presenting cells. There is, however, preclinical evidence that the immune system, opportunely stimulated, is capable of recognizing and killing tumor cells. It has been verified that the activation of autologous dendritic cells, derived from peripheral blood and pulsed with tumor antigens, results in the specific stimulation of T-cells against the tumor. Preliminary data from dendritic cell vaccination trials, mainly of advanced melanoma, show unequivocal evidence of immunization and of the first clinical responses. Many questions remain to be answered before more effective and widespread use of this type of vaccination is possible, especially in the early stages of the disease.
Collapse
Affiliation(s)
- Ruggero Ridolfi
- Dept. of Medical Oncology, Pierantoni Hospital, Via Forlanini 34, 47100 Forlì, Italy.
| | | | | | | | | |
Collapse
|
15
|
Frick LR, Rapanelli M, Arcos MLB, Cremaschi GA, Genaro AM. Oral administration of fluoxetine alters the proliferation/apoptosis balance of lymphoma cells and up-regulates T cell immunity in tumor-bearing mice. Eur J Pharmacol 2011; 659:265-72. [PMID: 21497159 DOI: 10.1016/j.ejphar.2011.03.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 03/02/2011] [Accepted: 03/22/2011] [Indexed: 11/19/2022]
Abstract
Antidepressants have a controversial role with regard to their influence on cancer and immunity. Recently, we showed that fluoxetine administration induces an enhancement of the T-cell mediated immunity in naïve mice, resulting in the inhibition of tumor growth. Here we studied the effects of fluoxetine on lymphoma proliferation/apoptosis and immunity in tumor bearing-mice. We found an increase of apoptotic cells (active Caspase-3(+)) and a decrease of proliferative cells (PCNA(+)) in tumors growing in fluoxetine-treated animals. In addition, differential gene expressions of cell cycle and death markers were observed. Cyclins D3, E and B were reduced in tumors from animals treated with fluoxetine, whereas the tumor suppressor p53 and the cell cycle inhibitors p15/INK4B, p16/INK4A and p27/Kip1 were increased. Besides, the expression of the antiapoptotic factor Bcl-2 and the proapoptotic factor Bad were lower and higher respectively in these animals. These changes were accompanied by increased IFN-γ and TNF-α levels as well as augmented circulating CD8(+) T lymphocytes in tumor-bearing mice treated with the antidepressant. Therefore, we propose that the up-regulation of T-cell mediated antitumor immunity may be contributing to the alterations of tumor cell proliferation and apoptosis thus resulting in the inhibition of tumor progression.
Collapse
Affiliation(s)
- Luciana Romina Frick
- Centro de Estudios Farmacológicos y Botánicos, Consejo Nacional de Investigaciones Científicas y Técnicas, 1° Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155 Piso 15, Buenos Aires (1121), Argentina.
| | | | | | | | | |
Collapse
|
16
|
Wulf-Goldenberg A, Eckert K, Fichtner I. Intrahepatically transplanted human cord blood cells reduce SW480 tumor growth in the presence of bispecific EpCAM/CD3 antibody. Cytotherapy 2011; 13:108-13. [DOI: 10.3109/14653249.2010.515577] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
17
|
Cabrera R, Ararat M, Eksioglu EA, Cao M, Xu Y, Wasserfall C, Atkinson MA, Liu C, Nelson DR. Influence of serum and soluble CD25 (sCD25) on regulatory and effector T-cell function in hepatocellular carcinoma. Scand J Immunol 2010; 72:293-301. [PMID: 20883314 DOI: 10.1111/j.1365-3083.2010.02427.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our previous studies showed that high levels of soluble CD25 (sCD25) in the serum of patients with hepatocellular carcinoma (HCC) correlated with blunted effector T-cells (Teff) responses, tumour burden and poor survival. Understanding the interactions between Teff, CD4+CD25+ regulatory T cells (Treg) and soluble factors can identify novel therapeutic targets. In this study, we characterize the mechanisms by which HCC serum and sCD25 mediate suppression of Teff and evaluate the effect of sCD25 on the suppression assays with normal healthy control cells (NHC) at a 1:1 Treg to Teff cell ratio to determine whether sCD25 has any impact on Treg suppression. HCC serum and sCD25 suppressed Teff proliferation and downregulated CD25 expression on HCC Teff in a dose-dependent fashion with sCD25 doses above 3000 pg/ml. Treg from HCC and cirrhosis patients suppressed proliferation of target CD4+CD25- Teff in serum-free medium (SFM). HCC Treg showed a higher degree of suppression than cirrhosis-derived Treg. In contrast, Treg from NHC did not suppress target Teff in SFM. However, isolated Treg from all three study subjects (HCC, cirrhosis and NHC) suppressed CD4+CD25- Teff in serum conditions or in the presence of sCD25 in the range 6000-12,000 pg/ml. In conclusion, downregulation of CD25 cell surface expression on Teff is part of the overall suppressive mechanism of sCD25 and HCC serum on Teff responses. The observed sCD25 and HCC serum-mediated suppression is further influenced via novel immune-inhibitory interaction between CD4+CD25+ Treg and sCD25.
Collapse
Affiliation(s)
- R Cabrera
- Section of Hepatobiliary Diseases, Department of Medicine, University of Florida, Gainesville, FL 32610-0214, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Türkseven MR, Oygür T. Evaluation of natural killer cell defense in oral squamous cell carcinoma. Oral Oncol 2010; 46:e34-7. [DOI: 10.1016/j.oraloncology.2010.02.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 02/19/2010] [Accepted: 02/19/2010] [Indexed: 11/15/2022]
|
19
|
Garrido C, Algarra I, Maleno I, Stefanski J, Collado A, Garrido F, Garcia-Lora AM. Alterations of HLA class I expression in human melanoma xenografts in immunodeficient mice occur frequently and are associated with higher tumorigenicity. Cancer Immunol Immunother 2010; 59:13-26. [PMID: 19430787 PMCID: PMC11031073 DOI: 10.1007/s00262-009-0716-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Accepted: 04/16/2009] [Indexed: 11/25/2022]
Abstract
Animal models are widely used to study the biological behavior of human tumors in vivo. Murine immunodeficient models are used to test novel human anti-tumor therapies, and humanized mice are employed to study immunotherapeutic protocols. We find that human melanoma cell lines lose HLA class I surface expression after growth in immunodeficient mice and that this phenomenon occurs frequently and is reproducible. This HLA loss is due to a coordinated down-regulation of APM and HLA heavy chain expression at the transcriptional level. It is produced by epigenetic modifications and can be reversed by treatment with histone deacetylase inhibitors or IFN-gamma. These HLA alterations only appear during in vivo growth and not during successive in vitro passages. Interestingly, these new tumor variants with HLA class I loss show higher tumorigenicity per se and may represent a more advanced state of the original tumor. Lack of MHC class I expression on tumor cells represents a frequent escape mechanism from the immune response. Our results indicate that tumor variants with alterations in MHC can also appear in vivo after the immunoescape phase in the absence of anti-tumor immune response. Our findings suggest that any studied parameter, i.e., HLA expression, of malignant cells in xenograft models, has to be evaluated before and after growth in immunodeficient mice, in order to design more appropriate immunotherapy and chemotherapy treatments against tumor cells growing in vivo.
Collapse
Affiliation(s)
- Cristina Garrido
- Servicio de Análisis Clínicos e Inmunologia, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
- Departamento de Bioquimica y Biologia Molecular III e Inmunologia, Universidad de Granada, Granada, Spain
| | - Ignacio Algarra
- Departamento Ciencias de La Salud, Universidad de Jaén, 23014 Jaén, Spain
| | - Isabel Maleno
- Servicio de Análisis Clínicos e Inmunologia, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
| | - Julia Stefanski
- Servicio de Análisis Clínicos e Inmunologia, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
| | - Antonia Collado
- Unidad de Investigación, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain
| | - Federico Garrido
- Servicio de Análisis Clínicos e Inmunologia, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
- Departamento de Bioquimica y Biologia Molecular III e Inmunologia, Universidad de Granada, Granada, Spain
| | - Angel M. Garcia-Lora
- Servicio de Análisis Clínicos e Inmunologia, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
| |
Collapse
|
20
|
Rauch D, Gross S, Harding J, Bokhari S, Niewiesk S, Lairmore M, Piwnica-Worms D, Ratner L. T-cell activation promotes tumorigenesis in inflammation-associated cancer. Retrovirology 2009; 6:116. [PMID: 20017942 PMCID: PMC2806367 DOI: 10.1186/1742-4690-6-116] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 12/17/2009] [Indexed: 02/03/2023] Open
Abstract
Chronic inflammation has long been associated with a wide range of malignancies, is now widely accepted as a risk factor for development of cancer, and has been implicated as a promoter of a variety of cancers including hematopoietic malignancies. We have described a mouse model uniquely suited to examine the link between inflammation and lymphoma in which the Tax oncogene, expressed in activated T and NK cells, perpetuates chronic inflammation that begins as microscopic intraepithelial lesions and develops into inflammatory nodules, subcutaneous tumors, and large granular lymphocytic leukemia. The use of bioluminescent imaging in these mice has expanded our ability to interrogate aspects of inflammation and tumorigenesis non-invasively. Here we demonstrate that bioluminescence induction in these mice correlated with inflammation resulting from wounding, T cell activation, and exposure to chemical agents. In experiments in which long-term effects of inflammation on disease outcome were monitored, the development of lymphoma was promoted by an inflammatory stimulus. Finally we demonstrated that activation of T-cells in T-cell receptor (TCR) transgenic TAX-LUC animals dramatically exacerbated the development of subcutaneous TCR- CD16+ LGL tumors. The role of activated T-cells and acquired immunity in inflammation-associated cancers is broadly applicable to hematopoietic malignancies, and we propose these mice will be of use in dissecting mechanisms by which activated T-cells promote lymphomagenesis in vivo.
Collapse
Affiliation(s)
- Dan Rauch
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Honma I, Torigoe T, Hirohashi Y, Kitamura H, Sato E, Masumori N, Tamura Y, Tsukamoto T, Sato N. Aberrant expression and potency as a cancer immunotherapy target of alpha-methylacyl-coenzyme A racemase in prostate cancer. J Transl Med 2009; 7:103. [PMID: 20003233 PMCID: PMC2797764 DOI: 10.1186/1479-5876-7-103] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 12/09/2009] [Indexed: 12/28/2022] Open
Abstract
Alpha-methylacyl-CoA racemase (AMACR) is an enzyme playing an important role in the beta-oxidation of branched-chain fatty acids and fatty acid derivatives. High expression levels of AMACR have been described in various cancers, including prostate cancer, colorectal cancer and kidney cancer. Because of its cancer-specific and frequent expression, AMACR could be an attractive target for cytotoxic T-lymphocyte (CTL)-based immunotherapy for cancer. In the present study, we examined the induction of AMACR-specific CTLs from prostate cancer patients' peripheral blood mononuclear cells (PBMCs) and determined HLA-A24-restricted CTL epitopes. RT-PCR and immunohistochemical analysis revealed that AMACR was strongly expressed in prostate cancer cell lines and tissues as compared with benign or normal prostate tissues. Four AMACR-derived peptides carrying the HLA-A24-binding motif were synthesized from the amino acid sequence of this protein and analyzed to determine their binding affinities to HLA-A24. By stimulating patient's PBMCs with the peptides, specific CTLs were successfully induced in 6 of 11 patients. The peptide-specific CTLs exerted significant cytotoxic activity against AMACR-expressing prostate cancer cells in the context of HLA-A24. Our study demonstrates that AMACR could become a target antigen for prostate cancer immunotherapy, and that the AMACR-derived peptides might be good peptide vaccine candidates for HLA-A24-positive AMACR-expressing cancer patients.
Collapse
Affiliation(s)
- Ichiya Honma
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ash S, Gigi V, Askenasy N, Fabian I, Stein J, Yaniv I. Graft versus neuroblastoma reaction is efficiently elicited by allogeneic bone marrow transplantation through cytolytic activity in the absence of GVHD. Cancer Immunol Immunother 2009; 58:2073-84. [PMID: 19437016 PMCID: PMC11030755 DOI: 10.1007/s00262-009-0715-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 04/16/2009] [Indexed: 12/01/2022]
Abstract
Continuous efforts are dedicated to develop immunotherapeutic approaches to neuroblastoma (NB), a tumor that relapses at high rates following high-dose conventional cytotoxic therapy and autologous bone marrow cell (BMC) reconstitution. This study presents a series of transplant experiments aiming to evaluate the efficacy of allogeneic BMC transplantation. Neuro-2a cells were found to express low levels of class I major histocompatibility complex (MHC) antigens. While radiation and syngeneic bone marrow transplantation (BMT) reduced tumor growth (P < 0.001), allogeneic BMT further impaired subcutaneous development of Neuro-2a cells (P < 0.001). Allogeneic donor-derived T cells displayed direct cytotoxic activity against Neuro-2a in vitro, a mechanism of immune-mediated suppression of tumor growth. The proliferation of lymphocytes from congenic mice bearing subcutaneous tumors was inhibited by tumor lysate, suggesting that a soluble factor suppresses cytotoxic activity of syngeneic lymphocytes. However, the growth of Neuro-2a cells was impaired when implanted into chimeric mice at various times after syngeneic and allogeneic BMT. F1 (donor-host) splenocytes were infused attempting to foster immune reconstitution, however they engrafted transiently and had no effect on tumor growth. Taken together, these data indicate: (1) Neuro-2a cells express MHC antigens and immunogenic tumor associated antigens. (2) Allogeneic BMT is a significantly better platform to develop graft versus tumor (GVT) immunotherapy to NB as compared to syngeneic (autologous) immuno-hematopoietic reconstitution. (3) An effective GVT reaction in tumor bearing mice is primed by MHC disparity and targets tumor associated antigens.
Collapse
Affiliation(s)
- Shifra Ash
- Department of Pediatric Hematology-Oncology, Schneider Children’s Medical Center of Israel, 14 Kaplan Street, 49202 Petach Tikva, Israel
- Frankel Laboratory, Schneider Children’s Medical Center of Israel, 14 Kaplan Street, 49202 Petach Tikva, Israel
| | - Vered Gigi
- Frankel Laboratory, Schneider Children’s Medical Center of Israel, 14 Kaplan Street, 49202 Petach Tikva, Israel
- Department of Cell Biology, Sackler School of Medicine, Tel Aviv University, 69788 Ramat Aviv, Israel
| | - Nadir Askenasy
- Frankel Laboratory, Schneider Children’s Medical Center of Israel, 14 Kaplan Street, 49202 Petach Tikva, Israel
| | - Ina Fabian
- Department of Cell Biology, Sackler School of Medicine, Tel Aviv University, 69788 Ramat Aviv, Israel
| | - Jerry Stein
- Department of Pediatric Hematology-Oncology, Schneider Children’s Medical Center of Israel, 14 Kaplan Street, 49202 Petach Tikva, Israel
| | - Isaac Yaniv
- Department of Pediatric Hematology-Oncology, Schneider Children’s Medical Center of Israel, 14 Kaplan Street, 49202 Petach Tikva, Israel
| |
Collapse
|
23
|
Onuma K, Sato Y, Ogawara S, Shirasawa N, Kobayashi M, Yoshitake J, Yoshimura T, Iigo M, Fujii J, Okada F. Nano-scaled particles of titanium dioxide convert benign mouse fibrosarcoma cells into aggressive tumor cells. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:2171-83. [PMID: 19815711 DOI: 10.2353/ajpath.2009.080900] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Nanoparticles are prevalent in both commercial and medicinal products; however, the contribution of nanomaterials to carcinogenesis remains unclear. We therefore examined the effects of nano-sized titanium dioxide (TiO(2)) on poorly tumorigenic and nonmetastatic QR-32 fibrosarcoma cells. We found that mice that were cotransplanted subcutaneously with QR-32 cells and nano-sized TiO(2), either uncoated (TiO(2)-1, hydrophilic) or coated with stearic acid (TiO(2)-2, hydrophobic), did not form tumors. However, QR-32 cells became tumorigenic after injection into sites previously implanted with TiO(2)-1, but not TiO(2)-2, and these developing tumors acquired metastatic phenotypes. No differences were observed either histologically or in inflammatory cytokine mRNA expression between TiO(2)-1 and TiO(2)-2 treatments. However, TiO(2)-2, but not TiO(2)-1, generated high levels of reactive oxygen species (ROS) in cell-free conditions. Although both TiO(2)-1 and TiO(2)-2 resulted in intracellular ROS formation, TiO(2)-2 elicited a stronger response, resulting in cytotoxicity to the QR-32 cells. Moreover, TiO(2)-2, but not TiO(2)-1, led to the development of nuclear interstices and multinucleate cells. Cells that survived the TiO(2) toxicity acquired a tumorigenic phenotype. TiO(2)-induced ROS formation and its related cell injury were inhibited by the addition of antioxidant N-acetyl-l-cysteine. These results indicate that nano-sized TiO(2) has the potential to convert benign tumor cells into malignant ones through the generation of ROS in the target cells.
Collapse
Affiliation(s)
- Kunishige Onuma
- Department of Biochemistry and Molecular Biology, Yamagata University, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Canonical small interfering RNA (siRNA) duplexes are potent activators of the mammalian innate immune system. The induction of innate immunity by siRNA is dependent on siRNA structure and sequence, method of delivery, and cell type. Synthetic siRNA in delivery vehicles that facilitate cellular uptake can induce high levels of inflammatory cytokines and interferons after systemic administration in mammals and in primary human blood cell cultures. This activation is predominantly mediated by immune cells, normally via a Toll-like receptor (TLR) pathway. The siRNA sequence dependency of these pathways varies with the type and location of the TLR involved. Alternatively nonimmune cell activation may also occur, typically resulting from siRNA interaction with cytoplasmic RNA sensors such as RIG1. As immune activation by siRNA-based drugs represents an undesirable side effect due to the considerable toxicities associated with excessive cytokine release in humans, understanding and abrogating this activity will be a critical component in the development of safe and effective therapeutics. This review describes the intracellular mechanisms of innate immune activation by siRNA, the design of appropriate sequences and chemical modification approaches, and suitable experimental methods for studying their effects, with a view toward reducing siRNA-mediated off-target effects.
Collapse
|
25
|
Bühler P, Molnar E, Dopfer EP, Wolf P, Gierschner D, Wetterauer U, Schamel WW, Elsässer-Beile U. Target-dependent T-cell Activation by Coligation With a PSMA×CD3 Diabody Induces Lysis of Prostate Cancer Cells. J Immunother 2009; 32:565-73. [DOI: 10.1097/cji.0b013e3181a697eb] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
26
|
Frick LR, Arcos MLB, Rapanelli M, Zappia MP, Brocco M, Mongini C, Genaro AM, Cremaschi GA. Chronic restraint stress impairs T-cell immunity and promotes tumor progression in mice. Stress 2009; 12:134-43. [PMID: 18609297 DOI: 10.1080/10253890802137437] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Long-term exposure to stressful situations can affect the immune system. The T-cell response is an important component of anti-tumoral immunity. Hence, impairment of the immune function induced by a chronic stressor has been postulated to alter the immunosurveillance of tumors, thus leading to a worse neoplastic prognosis. Here, we show that chronic restraint stress affects T-cell mediated immunity in mice. This was evidenced by a decrease of mitogen-induced T-cell proliferation, a reduction in CD4(+)T lymphocyte number and a decrease of tumor necrosis factor-alpha (TNF-alpha) and Interferon-gamma (IFN-gamma) production in stressed mice. Additionally, mice subjected to chronic restraint stress displayed an enhancement of tumor growth in a syngeneic lymphoma model, i.e. an increase of tumor proliferation and a reduction of animal survival. Finally, stressed mice had a reduced specific cytotoxic response against these tumor cells. These results suggest that chronic exposure to stress promotes cancer establishment and subsequent progression, probably by depressing T-cell mediated immunity. The T-cell immunity impairment as well as the tumor progression enhancement emphasize the importance of the therapeutic management of stress to improve the prognosis of cancer patients.
Collapse
Affiliation(s)
- L R Frick
- Centro de Estudios Farmacológicos y Botánicos, Consejo Nacional de Investigaciones Cientificas y Técnicas, 1 Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The stimulation of a tumour-specific T-cell response has several theoretical advantages over other forms of cancer treatment. First, T cells can home in to antigen-expressing tumour deposits no matter where they are located in the body-even in deep tissue beds. Additionally, T cells can continue to proliferate in response to immunogenic proteins expressed in cancer until all the tumour cells are eradicated. Finally, immunological memory can be generated, allowing for eradication of antigen-bearing tumours if they reoccur. We will highlight two direct methods of stimulating tumour-specific T-cell immunity: active immunisation with cancer vaccines and infusion of competent T cells via adoptive T-cell treatment. Preclinical and clinical studies have shown that modulation of the tumour microenvironment to support the immune response is as important as stimulation of the most appropriate effector T cells. The future of T-cell immunity stimulation to treat cancer will need combination approaches focused on both the tumour and the T cell.
Collapse
Affiliation(s)
- Mary L Disis
- Center for Translational Medicine in Women's Health, Tumor Vaccine Group, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
28
|
Nonaka M, Ma BY, Kaswasaki T. Colorectal carcinoma-associated carbohydrate recognition and tumor-immunological regulation by C-type lectin DC-SIGN. TRENDS GLYCOSCI GLYC 2009. [DOI: 10.4052/tigg.21.228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
29
|
Judge A, MacLachlan I. Overcoming the innate immune response to small interfering RNA. Hum Gene Ther 2008; 19:111-24. [PMID: 18230025 DOI: 10.1089/hum.2007.179] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many types of nucleic acid, including canonical small interfering RNA (siRNA) duplexes, are potent activators of the mammalian innate immune system. Synthetic siRNA duplexes can induce high levels of inflammatory cytokines and type I interferons, in particular interferon-alpha, after systemic administration in mammals and in primary human blood cell cultures. These responses are greatly potentiated by the use of delivery vehicles that facilitate cellular uptake of the siRNA. Although the immunomodulatory effects of nucleic acids may be harnessed therapeutically, for example, in oncology and allergy applications, in many cases immune activation represents a significant undesirable side effect due to the toxicities associated with excessive cytokine release and associated inflammatory syndromes. The potential for siRNA-based drugs to be rendered immunogenic is also a cause for concern because the establishment of an antibody response may severely compromise both safety and efficacy. Clearly, there are significant implications both for the development of siRNA-based drugs and in the interpretation of gene-silencing effects elicited by siRNA. This review provides the background information required to anticipate, manage, and abrogate the immunological effects of siRNA and will assist the reader in the successful in vivo application of siRNA-based drugs.
Collapse
Affiliation(s)
- Adam Judge
- Protiva Biotherapeutics, Burnaby, British Columbia, Canada V5G 4Y1
| | | |
Collapse
|
30
|
Ridolfi L, Petrini M, Fiammenghi L, Riccobon A, Ridolfi R. Human embryo immune escape mechanisms rediscovered by the tumor. Immunobiology 2008; 214:61-76. [PMID: 19159828 DOI: 10.1016/j.imbio.2008.03.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 02/29/2008] [Accepted: 03/10/2008] [Indexed: 01/05/2023]
Abstract
Towards the end of the 1990s, the two opposing theories on immunosurveillance and immunostimulation were extensively studied by researchers in an attempt to understand the complex mechanisms that regulate the relation between tumors and the host's immune system. Both theories probably have elements that would help us to comprehend how the host can induce anti-tumor clinical responses through stimulation of the immune system and which could also give us a deeper insight into the mechanisms of tumor immunosuppression. The model that most resembles the behavior of tumor cells in terms of growth, infiltration and suppression of the immune system of the environment in which they live is undoubtedly that of the embryonic cell. The fetus behaves like an allogenic transplant within the mother's body, using every means it has to escape from and defend itself against the mother's immune system. The majority of these mechanisms are the same as those found in tumor cells: antigenic loss, lack of expression of classic HLA-I molecules, production of immunosuppressive cytokines, induction of lack of expression of co-stimulatory molecules in antigen presenting cells, and induction of apoptosis in infiltrating lymphocytes, with activation of a type Th2 regulatory lymphocyte response. A careful and comparative study of key mechanisms capable of triggering tolerance or cytotoxicity in both embryonic and tumor cells could prove immensely valuable in designing new strategies for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Laura Ridolfi
- Immunotherapy and Somatic Cell Therapy Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Via Maroncelli 40, Meldola 47014, Italy.
| | | | | | | | | |
Collapse
|
31
|
Muraro M, Mereuta OM, Saglio F, Carraro F, Berger M, Madon E, Fagioli F. Interactions between osteosarcoma cell lines and dendritic cells immune function: An in vitro study. Cell Immunol 2008; 253:71-80. [PMID: 18565501 DOI: 10.1016/j.cellimm.2008.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2008] [Revised: 04/24/2008] [Accepted: 05/06/2008] [Indexed: 11/24/2022]
Abstract
Dendritic cells (DCs) might be partly responsible for the defective immune response in tumor bearing hosts, but no study in osteosarcoma patients is still available. Therefore, we investigated in vitro whether human osteosarcoma cell lines have an inhibitor effect on different types of DCs: CD14+DCs, DC1 and DC2. DCs derived from healthy donors were cultured with osteosarcoma cell lines and appropriate cytokine cocktails and analysed for the expression of co-stimulatory molecules (CD40, CD80, CD83, CD86, HLA-DR). Each interaction resulted in a lower phenotypic expression of the DCs maturation markers, especially on DC2. Moreover, the addition of various cytokines and compounds (rhIL-12, CD40L, Indometacin) induced the DC1 and DC2 subsets towards the Th1 pattern as shown by ELISA. Osteosarcoma highly interferes with an in vitro DCs immune function as antigen presenting cells. The understanding of tumor biology underlines the need for a specific osteosarcoma immunotherapy able to reverse this immune-surveillance inhibition.
Collapse
Affiliation(s)
- Michela Muraro
- Stem Cell Transplantation and Cellular Therapy Unit, Pediatric Onco-Hematology Department, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | | | | | | | | | | | | |
Collapse
|
32
|
Sega EI, Lu Y, Ringor M, Leamon CP, Low PS. Low-dose radiation potentiates the therapeutic efficacy of folate receptor-targeted hapten therapy. Int J Radiat Oncol Biol Phys 2008; 71:559-66. [PMID: 18411004 DOI: 10.1016/j.ijrobp.2008.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 02/04/2008] [Accepted: 02/04/2008] [Indexed: 11/27/2022]
Abstract
PURPOSE Human cancers frequently overexpress a high-affinity cell-surface receptor for the vitamin folic acid. Highly immunogenic haptens can be targeted to folate receptor-expressing cell surfaces by administration of folate-hapten conjugates, rendering the decorated tumor cell surfaces more recognizable by the immune system. Treatment of antihapten-immunized mice with folate-hapten constructs results in elimination of moderately sized tumors by the immune system. However, when subcutaneous tumors exceed 300 mm(3) before initiation of therapy, antitumor activity is significantly decreased. In an effort to enhance the efficacy of folate-targeted hapten immunotherapy (FTHI) against large tumors, we explored the combination of targeted hapten immunotherapy with low-dose radiotherapy. METHODS AND MATERIALS Mice bearing 300-mm(3) subcutaneous tumors were treated concurrently with FTHI (500 nmol/kg of folate conjugated to fluorescein isothiocyanate, 20,000 U/dose of interleukin 2, and 25,000 U/dose of interferon alpha) and low-dose radiotherapy (3 Gy/dose focused directly on the desired tumor mass). The efficacy of therapy was evaluated by measuring tumor volume. RESULTS Tumor growth analyses show that radiotherapy synergizes with FTHI in antihapten-immunized mice, thereby allowing for cures of animals bearing tumors greater than 300 mm(3). More importantly, nonirradiated distal tumor masses in animals containing locally irradiated tumors also showed improved response to hapten immunotherapy, suggesting that not all tumor lesions must be identified and irradiated to benefit from the combination therapy. CONCLUSIONS These results suggest that simultaneous treatment with FTHI and radiation therapy can enhance systemic antitumor activity in tumor-bearing mice.
Collapse
Affiliation(s)
- Emanuela I Sega
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | |
Collapse
|
33
|
Nunes EA, Bonatto SJ, de Oliveira HHP, Rivera NLM, Maiorka A, Krabbe EL, Tanhoffer RA, Fernandes LC. The effect of dietary supplementation with 9-cis:12-trans and 10-trans:12-cis conjugated linoleic acid (CLA) for nine months on serum cholesterol, lymphocyte proliferation and polymorphonuclear cells function in Beagle dogs. Res Vet Sci 2008; 84:62-7. [PMID: 17521690 DOI: 10.1016/j.rvsc.2007.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Revised: 02/01/2007] [Accepted: 03/25/2007] [Indexed: 11/20/2022]
Abstract
Conjugated linoleic acids (CLA), 9-cis:11-trans and 10-trans:12-cis, have been shown to be able to modify some immune cells parameters and plasma lipids in a variety of experiment models. Since lymphocytes and polymorphonuclear cells (PMNC) have a large spectrum functions in the immune response, the knowledge in this field has to be expanded. Beagle dogs were fed a control diet or a CLA supplemented diet for nine months. Blood was collected for biochemical analysis and lymphocyte and PMNC isolation. PMNC were assayed for lysosome content, phagocytic activity and superoxide anion production. A lymphocyte proliferation capacity assay was done. The CLA fed dogs had a 34% reduction in total cholesterol (P < 0.05), 28% in LDL (P < 0.05) and 28% non-HDL-cholesterol (P < 0.05). Neither of the PMNC parameters evaluated demonstrated significant alteration. Lymphocytes from CLA group increased by 45% their mitotic capacity (P < 0.05). Our study demonstrates that CLA can successfully modify the lipid profile of dogs (monogastrics) when fed at reasonable levels, but did not significantly alter inflammatory function as would generally predicted. Further, we had some indication that CLA modulated T cell responsiveness.
Collapse
Affiliation(s)
- E A Nunes
- Cell Metabolism Laboratory, Physiology Department, Biological Science Building, Universidade Federal do Paraná, 81530-990, Curitiba-PR, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Pinzon-Charry A, Maxwell T, López JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol 2008; 83:451-61. [PMID: 16174093 DOI: 10.1111/j.1440-1711.2005.01371.x] [Citation(s) in RCA: 231] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Several reports have demonstrated that tumours are not intrinsically resistant to the immune response. However, neoplasias commonly fail to initiate and maintain adequate immunity. A number of factors have been implicated in causing the failure, including aberrant antigen processing by tumour cells, anergy or deletion of T cells, and recruitment of inhibitory/regulatory cell types. It has been suggested that dysfunction of dendritic cells (DC) induced by the tumour is one of the critical mechanisms to escape immune surveillance. As a minor subset of leucocytes, DC are the key APC for initiating immune responses. DC are poised at the boundaries of the periphery and the inner tissues, sampling antigens of diverse origin. Following their encounter with antigen or danger signals, DC migrate to lymph nodes, where they activate effector cells essential for tumour clearance. Although the DC system is highly heterogeneous, the differentiation and function of DC populations is largely regulated by exogenous factors. Malignancies appear to exploit this by producing a plethora of immunosuppressive factors capable of affecting DC, thus exerting systemic effects on immune function. This review examines recent findings on the effects of tumour-derived factors inducing DC dysfunction and in particular examines the findings on alteration of DC differentiation, maturation and longevity as a potent mechanism for immune suppression in cancer.
Collapse
Affiliation(s)
- Alberto Pinzon-Charry
- Dendritic Cell and Cancer Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | | | | |
Collapse
|
35
|
Krawczyk P, Wojas K, Milanowski J, Roliński J. The influence of different culture microenvironments on the generation of dendritic cells from non-small-cell lung cancer patients. Arch Immunol Ther Exp (Warsz) 2007; 55:405-15. [PMID: 18060367 PMCID: PMC2766451 DOI: 10.1007/s00005-007-0046-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Accepted: 10/25/2007] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Monocyte-derived dendritic cells (DCs) are currently under extensive evaluation as cell vaccines for cancer treatment. Many protocols regarding DCs generation in vitro with different protein components, especially autologous proteins, have been described. On the other hand, active tumor-derived factors in patients' serum could impair monocytes, which might result in their abrogated differentiation into DCs in vitro. MATERIALS AND METHODS Autologous DCs from non-small-cell lung cancer (NSCLC)-bearing patients were generated in different culture microenvironments. Peripheral blood mononuclear cells (PBMCs) were cultured in the presence of interleukin-4 and granulocyte-monocyte-stimulating factor with supplementation of 10% autologous serum, 10% allogenic serum, or 2% human albumin. The course of apoptosis, phagocytic ability, and the immunophenotype of the generated DCs were analyzed using flow cytometric methods. RESULTS After 48 h of culture, we found a lower percentage of CD1a+/CD14+ and a higher percentage of CD1a+/CD14(-) cells in the culture supplemented with human albumin than in the cultures supplemented with serums. The lowest CD14 antigen expression was found in the human albumin-supplemented 48-h cultures. After 48 h in the cultures carried out with human albumin we found significantly higher percentages of AV+/PI+ cells and AV(-)/PI+ cells than in cultures supplemented with autologous or allogenic serum. We also noted that the expression of FITC-dextran after 4 and 24 h of incubation was significantly higher in the cultures supplemented with both serums than in the HA-SC. The percentage of semi-mature DCs and of CD83 expression was lowest in the culture supplemented with 2% human albumin. CONCLUSIONS The kind of culture supplementation had a great impact on the apoptosis of cultured PBMCs. It could also influence the yield of monocyte-derived DCs. It was also confirmed that autologous and allogenic serums provide suitable microenvironments for the generation of autologous DCs from NSCLC patients. The choice of culture supplementation for DC generation is still unsolved and further studies should be undertaken.
Collapse
Affiliation(s)
- Paweł Krawczyk
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950, Lublin, Poland.
| | | | | | | |
Collapse
|
36
|
Champine PJ, Michaelson J, Weimer BC, Welch DR, DeWald DB. Microarray analysis reveals potential mechanisms of BRMS1-mediated metastasis suppression. Clin Exp Metastasis 2007; 24:551-65. [PMID: 17896182 PMCID: PMC2214901 DOI: 10.1007/s10585-007-9092-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 07/27/2007] [Indexed: 12/14/2022]
Abstract
We used Affymetrix microarrays to compare gene expression profiles of the metastatic parental breast cancer cell line MDA-MB-435 (435) and the non-metastatic daughter cell line created by the stable expression of the BReast cancer Metastasis Suppressor 1 (BRMS1) gene in 435 cells, MDA-MB-435-BRMS1 (435/BRMS1). Analysis of microarray data provided insight into some of the potential mechanisms by which BRMS1 inhibits tumor formation at secondary sites. Furthermore, due to the importance of the microenvironment, we also examined gene expression under different growth conditions (i.e., plus or minus serum). Expression of 565 genes was significantly (adjusted P-value <0.05) altered regardless of in vitro growth conditions. BRMS1 expression significantly increased multiple major histocompatability complex (MHC) genes and significantly decreased expression of several genes associated with protein localization and secretion. The pattern of gene expression associated with BRMS1 expression suggests that metastasis suppression may be mediated by enhanced immune recognition, altered transport, and/or secretion of metastasis-associated proteins.
Collapse
Affiliation(s)
- Patricia J. Champine
- Center for Integrated BioSystems, Utah State University, Logan, Utah 84322-4700, USA
| | - Jacob Michaelson
- Center for Integrated BioSystems, Utah State University, Logan, Utah 84322-4700, USA
| | - Bart C. Weimer
- Center for Integrated BioSystems, Utah State University, Logan, Utah 84322-4700, USA
| | - Danny R. Welch
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
- National Foundation for Cancer Research, Center for Metastasis Research, Logan, Utah 84322-5305, USA
| | - Daryll B. DeWald
- National Foundation for Cancer Research, Center for Metastasis Research, Logan, Utah 84322-5305, USA
- Department of Biology, Utah State University, Logan, Utah 84322-5305, USA
- Correspondence to: D.B. DeWald, Department of Biology and Center for Integrated BioSystems, Utah State University, Logan, Utah 84322-5305 (435)-797-1909
| |
Collapse
|
37
|
Manning EA, Ullman JGM, Leatherman JM, Asquith JM, Hansen TR, Armstrong TD, Hicklin DJ, Jaffee EM, Emens LA. A vascular endothelial growth factor receptor-2 inhibitor enhances antitumor immunity through an immune-based mechanism. Clin Cancer Res 2007; 13:3951-9. [PMID: 17606729 DOI: 10.1158/1078-0432.ccr-07-0374] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Given the complex tumor microenvironment, targeting multiple cellular components may be the most effective cancer treatment strategy. Therefore, we tested whether antiangiogenic and immune-based therapy might synergize by characterizing the activity of DC101, an antiangiogenic monoclonal antibody specific for vascular endothelial growth factor receptor-2 (VEGF-R2), alone and with HER-2/neu (neu)-targeted vaccination. EXPERIMENTAL DESIGN Neu-expressing breast tumors were measured in treated nontolerant FVB mice and immune-tolerant neu transgenic (neu-N) mice. Neu-specific and tumor cell-specific immune responses were assessed by intracellular cytokine staining, ELISPOT, and CTL assays. RESULTS DC101 decreased angiogenesis and increased tumor cell apoptosis. Although DC101 increased serum levels of the immunosuppressive cytokine VEGF, no evidence of systemic immune inhibition was detected. Moreover, DC101 did not impede the influx of tumor-infiltrating lymphocytes. In FVB mice, DC101 inhibited tumor growth in part through a T cell-dependent mechanism, resulting in both increased tumor-specific CD8(+) T cells and tumor regression. Combining DC101 with neu-specific vaccination accelerated tumor regression, augmenting the lytic activity of CD8(+) cytotoxic T cells. In tolerant neu-N mice, DC101 only delayed tumor growth without inducing frank tumor regression or antigen-specific T-cell activation. Notably, mitigating immune tolerance by inhibiting regulatory T cell activity with cyclophosphamide revealed DC101-mediated augmentation of antitumor responses in vaccinated neu-N mice. CONCLUSIONS This is the first report of DC101-induced antitumor immune responses. It establishes the induction of tumor-specific T-cell responses as one consequence of VEGF-R2 targeting with DC101. These data support the development of multitargeted cancer therapy combining immune-based and antiangiogenic agents for clinical translation.
Collapse
Affiliation(s)
- Elizabeth A Manning
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Yang ZM, Li EM, Lai BC, Wang YL, Si LS. Anti-CD3 scFv-B7.1 fusion protein expressed on the surface of HeLa cells provokes potent T-lymphocyte activation and cytotoxicityThis paper is one of a selection of papers in this Special Issue, entitled International Symposium on Recent Advances in Molecular, Clinical, and Social Medicine, and has undergone the Journal's usual peer-review process. Biochem Cell Biol 2007; 85:196-202. [PMID: 17534400 DOI: 10.1139/o07-013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The targeting of tumor cells by cytotoxic T lymphocytes is a promising strategy for biotherapy, but T cells require 2 signals via the T-cell receptor – CD3 complex and CD28 molecules for activation. To bridge the gap between cytotoxic T lymphocytes and tumor cells, our objective in this study was to describe the construction and the cell surface-anchored expression of a fusion protein, anti-CD3 scFv-B7.1, derived from inserting a fusion gene encoding anti-CD3 scFv and the extra-cellular domain of B7.1 fused by the splicing by overlap extension method into a mammalian expression vector, pDisplay. Transfection of the recombinant vector by electroporation into HeLa cells resulted in the production of protein migrating at approximately 57 kDa under reducing conditions. The expressed fusion protein could bind to T lymphocytes and induce strong T-cell activation. Meanwhile, a potent cytotoxicity was induced in the mixed culture of T-cell-modified tumor cells in a 96 h methyl-thiazolyl-diphenyl tetrazolium bromide assay. Our results indicate that this bifunctional protein, through activating T lymphocytes to lyse homologous human carcinomas, may be of potential value for T-cell-based immunotherapeutical treatment protocols in vivo.
Collapse
Affiliation(s)
- Zhang-Min Yang
- Department of Biochemistry and Molecular Biology, Medical College, Shantou University, Shantou 515041, PR China.
| | | | | | | | | |
Collapse
|
39
|
Kim R, Emi M, Tanabe K, Arihiro K. Potential functional role of plasmacytoid dendritic cells in cancer immunity. Immunology 2007; 121:149-57. [PMID: 17371541 PMCID: PMC2265944 DOI: 10.1111/j.1365-2567.2007.02579.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs), as well as myeloid dendritic cells (mDCs), have a dual role not only in initiating immune responses but also in inducing tolerance to exogenous and endogenous antigens. Tumour antigens originate from endogenous self-antigens, which are poorly immunogenic and also subject to change during tumour progression. In general, tumour antigens derived from apoptotic cells are captured by immature mDCs, antigen presentation by which is most likely to result in immune tolerance. In contrast, tumour antigens may be taken up by pDCs through Toll-like receptor 9 (TLR9) via receptor-mediated endocytosis. TLR9-dependent activation of pDCs results in the secretion of pro-inflammatory cytokines such as interleukin (IL)-12 and type I interferons (IFNs) through a MyD88-dependent pathway. Type I IFNs also activate mDCs for T-cell priming. Although pDCs recruited to the tumour site are implicated in facilitating tumour growth via immune suppression, they can be released from the tumour as a result of cell death caused by primary systemic chemotherapy, and can then be activated through TLR9. Thus, synergistically with mDCs, pDCs may also play a crucial role in mediating cancer immunity. In this review, the potential functional duality and plasticity of pDCs mediated by TLR9 ligation in cancer immunity will be discussed.
Collapse
Affiliation(s)
- Ryungsa Kim
- International Radiation Information Center, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| | | | | | | |
Collapse
|
40
|
Altin JG, Parish CR. Liposomal vaccines--targeting the delivery of antigen. Methods 2007; 40:39-52. [PMID: 16997712 DOI: 10.1016/j.ymeth.2006.05.027] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 05/05/2006] [Indexed: 01/05/2023] Open
Abstract
Vaccines that can prime the adaptive immune system for a quick and effective response against a pathogen or tumor cells, require the generation of antigen (Ag)-specific memory T and B cells. The unique ability of dendritic cells (DCs) to activate naïve T cells, implies a key role for DCs in this process. The generation of tumor-specific CD8(+) cytotoxic T cells (CTLs) is dependent on both T cell stimulation with Ag (peptide-MHC-complexes) and costimulation. Interestingly, tumor cells that lack expression of T cell costimulatory molecules become highly immunogenic when transfected to express such molecules on their surface. Adoptive immunotherapy with Ag-pulsed DCs also is a strategy showing promise as a treatment for cancer. The use of such cell-based vaccines, however, is cumbersome and expensive to use clinically, and/or may carry risks due to genetic manipulations. Liposomes are particulate vesicular lipid structures that can incorporate Ag, immunomodulatory factors and targeting molecules, and hence can serve as potent vaccines. Similarly, Ag-containing plasma membrane vesicles (PMV) derived from tumor cells can be modified to incorporate a T cell costimulatory molecule to provide both TCR stimulation, and costimulation. PMVs also can be modified to contain IFN-gamma and molecules for targeting DCs, permitting delivery of both Ag and a DC maturation signal for initiating an effective immune response. Our results show that use of such agents as vaccines can induce potent anti-tumor immune responses and immunotherapeutic effects in tumor models, and provide a strategy for the development of effective vaccines and immunotherapies for cancer and infectious diseases.
Collapse
Affiliation(s)
- Joseph G Altin
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia.
| | | |
Collapse
|
41
|
Hamdy S, Elamanchili P, Alshamsan A, Molavi O, Satou T, Samuel J. Enhanced antigen-specific primary CD4+ and CD8+ responses by codelivery of ovalbumin and toll-like receptor ligand monophosphoryl lipid A in poly(D,L-lactic-co-glycolic acid) nanoparticles. J Biomed Mater Res A 2007; 81:652-62. [PMID: 17187395 DOI: 10.1002/jbm.a.31019] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The purpose of this research was to investigate the use of biodegradable poly(D,L-lactic-co-glycolic acid) nanoparticles (PLGA-NP) as a vaccine delivery system to codeliver antigen, ovalbumin (OVA) along with monophosphoryl lipid A (MPLA) as adjuvant for induction of potent CD4(+) and CD8(+) T cell responses. The primary CD4(+) T responses to OVA/MPLA NP were investigated using OVA-specific T cells from DO11.10 transgenic mice. Following adoptive transfer of these cells, mice were immunized s.c. by NP formulations. For assessing the CD8(+) responses, bone marrow derived dendritic cells (DCs) were pulsed with different OVA formulations, then, cocultured with CD8(+) T cells from OT-1 mice. T cell proliferation/activation and IFN-gamma secretion profile have been examined. Particulate delivery of OVA and MPLA to the DCs lead to markedly increase in in vitro CD8(+) T cell T cell proliferative responses (stimulation index >3000) and >13-folds increase in in vivo clonal expanded CD4(+) T cells. The expanded T cells were capable of cytokine secretion and expressed an activation and memory surface phenotype (CD62L(lo), CD11a(hi), and CD44(hi)). Codelivery of antigen and MPLA in PLGA-NP offers an effective method for induction of potent antigen specific CD4(+) and CD8(+) T cell responses.
Collapse
Affiliation(s)
- Samar Hamdy
- Faculty of Pharmacy and Pharmaceutical Sciences, 3133 Dentistry/Pharmacy Centre, University of Alberta, Edmonton, Alberta, Canada, T6G 2N8
| | | | | | | | | | | |
Collapse
|
42
|
Zhang J, Wang Q, Zhao D, Cao X. Induction of potent anti-tumor immunity by direct injection of Ad-LIGHT at the site of tumor inoculation. Cytotherapy 2007; 9:386-96. [PMID: 17573614 DOI: 10.1080/14653240701326749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND The aim of this study was to observe the therapeutic effects of adenovirus-mediated LIGHT gene transfer in murine B16 melanoma in vivo. METHODS C57BL/6 mice were inoculated subcutaneously with B16 cells to establish the murine melanoma model. The tumor-bearing mice were injected at the site of tumor inoculation with recombinant adenoviral vectors expressing the murine LIGHT gene. The tumor growth and survival period of tumor-bearing mice were observed. The splenic NK and CTL activity were measured in vitro by lactate dehydrogenase (LDH) release assay. The amounts of cytokines were determined with ELISA kits. RESULTS The LIGHT gene could be efficiently transduced into tumor tissue after injection of Ad-LIGHT. Treatment with Ad-LIGHT significantly inhibited the tumor growth and prolonged the survival period of the tumor-bearing mice. The splenic NK and CTL activity of the mice was also enhanced after LIGHT gene transfer. The production of IL-2 and IFN-gamma from lymphocytes derived from mice treated with Ad-LIGHT was increased significantly compared with control groups. DISCUSSION Our results indicate that local expression of the LIGHT gene can induce potent anti-tumor immunity and may be a promising treatment strategy for melanoma.
Collapse
Affiliation(s)
- J Zhang
- Department of Medical Microbiology and Immunology, Medical School, Shaoxing University, Shaoxing, P. R. China.
| | | | | | | |
Collapse
|
43
|
Al-Waili NS, Saloom KY, Al-Waili T, Al-Waili A, Al-Waili H. Modulation of prostaglandin activity, part 1: prostaglandin inhibition in the management of nonrheumatologic diseases: immunologic and hematologic aspects. Adv Ther 2007; 24:189-222. [PMID: 17526477 DOI: 10.1007/bf02850008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prostaglandins (PGs) are active biologic substances that are involved in a wide range of physiologic processes; when their production is out of balance, they are factors in the pathogenesis of illness. Modulation of PGs by inhibition or stimulation is promising for the management of various conditions. PG inhibitors are widely used to relieve pain and inflammation in patients with rheumatologic disease. Interest in the use of PG inhibitors to prevent cancer and cardiovascular events is growing. More than 27 y ago, investigators found that PG depresses antibody production in vivo; reduces serum iron, hemoglobin, and leukoid series in bone marrow during acute and chronic blood loss; reduces albumin during antigenic stimulation; suppresses hypercalcemia after bleeding; and reduces fasting blood sugar and hyperglycemia after ether anesthesia and bleeding. Chronic conditions that produce large quantities of PGs are associated with immunosuppression and secondary anemia. Investigators in the present study hypothesized (1) that the overproduction of PGs is responsible for immunosuppression and secondary anemia in conditions associated with increased PG synthesis, such as pathologic inflammation, malignancy, trauma, and injury, and (2) that PG inhibitors reverse immunosuppression and secondary anemia, thereby enhancing the immune response. This is supported by many reports that show the immunosuppressive effects of PGs and their role in the immunosuppression associated with pathologic inflammation, burns, trauma, and tumors. Inhibition of PGs can be achieved through the use of synthetic medicines and natural products. This article reviews the effects of PGs and inhibition of increased synthesis of PGs on the lymphoid system, hematologic indices, and bone marrow elements in trauma, injury, burns, and tumors. The Medline database (1966-2006) was used in this study. Investigators in the present study and others have provided evidence that shows the involvement of PGs in immunosuppression and secondary anemia, as well as the efficacy of inhibited overproduction of PGs in many pathologic conditions other than rheumatologic disease.
Collapse
Affiliation(s)
- Noori S Al-Waili
- Al-Waili's Charitable Foundation for Science and Trading, New York City, NY, USA.
| | | | | | | | | |
Collapse
|
44
|
Alves PMS, Faure O, Graff-Dubois S, Cornet S, Bolonakis I, Gross DA, Miconnet I, Chouaib S, Fizazi K, Soria JC, Lemonnier FA, Kosmatopoulos K. STEAP, a prostate tumor antigen, is a target of human CD8+ T cells. Cancer Immunol Immunother 2006; 55:1515-23. [PMID: 16622681 PMCID: PMC11030764 DOI: 10.1007/s00262-006-0165-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2005] [Accepted: 03/18/2006] [Indexed: 12/12/2022]
Abstract
STEAP is a recently identified protein shown to be particularly overexpressed in prostate cancer and also present in numerous human cancer cell lines from prostate, pancreas, colon, breast, testicular, cervical, bladder and ovarian carcinoma, acute lymphocytic leukemia and Ewing sarcoma. This expression profile renders STEAP an appealing candidate for broad cancer immunotherapy. In order to investigate if STEAP is a tumor antigen that can be targeted by specific CD8(+) T cells, we identified two high affinity HLA-A*0201 restricted peptides (STEAP(86-94) and STEAP(262-270)). These peptides were immunogenic in vivo in HLA-A*0201 transgenic HHD mice. Peptide specific murine CD8 T cells recognized COS-7 cells co-transfected with HHD (HLA-A*0201) and STEAP cDNA constructs and also HLA-A*0201(+) STEAP(+) human tumor cells. Furthermore, STEAP(86-94) and STEAP(262-270) stimulated specific CD8(+) T cells from HLA-A*0201(+) healthy donors, and these peptide specific CD8(+) T cells recognized STEAP positive human tumor cells in an HLA-A*0201-restricted manner. Importantly, STEAP(86-94)-specific T cells were detected and reactive in the peripheral blood mononuclear cells in NSCLC and prostate cancer patients ex vivo. These results show that STEAP can be a target of anti-tumor CD8(+) T cells and that STEAP peptides can be used for a broad-spectrum-tumor immunotherapy.
Collapse
Affiliation(s)
- Pedro M S Alves
- INSERM 487, Institut Gustave Roussy, 94805, Villejuif, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Provenzano M, Panelli MC, Mocellin S, Bracci L, Sais G, Stroncek DF, Spagnoli GC, Marincola FM. MHC–peptide specificity and T-cell epitope mapping: where immunotherapy starts. Trends Mol Med 2006; 12:465-72. [PMID: 16962375 DOI: 10.1016/j.molmed.2006.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Revised: 08/04/2006] [Accepted: 08/24/2006] [Indexed: 11/21/2022]
Abstract
The evaluation and characterization of epitope-specific human leukocyte antigen (HLA)-restricted memory T-cell reactivity is an important step for the development of preventive vaccines and peptide-based immunotherapies for viral and tumor diseases. The past decade has witnessed the use of HLA-restricted peptides as tools to activate strong immune responses of naïve or memory T cells specifically. This has fuelled an active search for methodological approaches focusing on HLA and peptide associations. Here, we outline new perspective on the emerging opportunity of evaluating HLA and peptide restriction by using novel approaches, such as quantitative real-time PCR, that can identify epitope specificities that are potentially useful in clinical settings.
Collapse
Affiliation(s)
- Maurizio Provenzano
- Institute for Surgical Research and Hospital Management, University of Basel, 4031 Basel, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Liu A, Hu P, Khawli LA, Epstein AL. B7.1/NHS76: a new costimulator fusion protein for the immunotherapy of solid tumors. J Immunother 2006; 29:425-35. [PMID: 16799338 DOI: 10.1097/01.cji.0000208260.80791.3d] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Tumor evasion from immune surveillance is due to the anergic status of tumor-infiltrating lymphocytes, especially T cells. Inappropriate or absent expression of costimulatory molecules such as B7.1 and B7.2 lead to anergy and apoptosis of tumor-infiltrating T cells. To reverse this situation, a tumor-targeted fusion protein, human B7.1/NHS76, was generated by molecular engineering, which retains both the costimulatory activity of B7.1 and the tumor-targeting ability of NHS76 antibody. NHS76 is a human tumor necrosis therapy monoclonal antibody derived from phage display, and is capable of binding intracellular antigens, which are accessible and abundant in necrotic regions of tumors. As human B7.1 can interact functionally with murine B7.1 counter-receptors, the immunotherapeutic potential of this fusion protein was tested in 3 mouse tumor models (Colon 26, RENCA, and MAD109), and animal studies showed a 35% to 55% reduction in tumor volume. To modulate the immune inhibitory microenvironment in tumors, naturally occurring CD4+ CD25+ Treg cells were depleted by cytotoxic CD4 or CD25 antibodies. Combination therapy with anti-Treg and B7.1/NHS76 produced complete regression of established tumors and was associated with increased effector T-cell infiltration in tumors. Rechallenge experiments performed 3 months after mice attained complete remission by combination therapy showed that immunologic memory was established by these treatments. These studies indicate that the targeting of B7.1 to necrotic areas of tumors, where both the release of tumor antigens and infiltrating lymphocytes are prevalent, may be a new approach for the immunotherapy of solid tumors. Our results also suggest that the manifestation of immune-inhibitory factors such as the presence of Treg cells at the tumor site and associated draining lymph nodes may be a major cause for immune system failure to eradicate solid tumors.
Collapse
Affiliation(s)
- Aihong Liu
- Department of Pathology, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | | | | | | |
Collapse
|
47
|
Unitt E, Marshall A, Gelson W, Rushbrook SM, Davies S, Vowler SL, Morris LS, Coleman N, Alexander GJM. Tumour lymphocytic infiltrate and recurrence of hepatocellular carcinoma following liver transplantation. J Hepatol 2006; 45:246-53. [PMID: 16580084 DOI: 10.1016/j.jhep.2005.12.027] [Citation(s) in RCA: 191] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2005] [Revised: 12/22/2005] [Accepted: 12/29/2005] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIMS Liver transplantation is an effective treatment for highly selected patients with hepatocellular carcinoma (HCC), but tumour recurrence remains an important cause of mortality. There are few data on the relation between the recurrence of HCC and lymphocytic infiltration following liver transplantation. METHODS The tumour CD4+, CD8+, CD25+ and Foxp3+ lymphocyte infiltrate was assessed by immunohistochemistry in explant tissue of 69 patients who underwent liver transplantation for HCC between 1985 and 2001. The data were analysed according to HCC recurrence and factors known to be associated with outcome. RESULTS Tumour size (Hazard ratio (95% CI: 1.19 (1.02, 1.39), P = 0.03)), vascular invasion (P = 0.02), lymphocyte infiltration (P = 0.02) and CD4:CD8 ratio (P = 0.001) were identified as significant univariate predictors of tumour recurrence. On multivariate analysis CD4:8 ratio (P = 0.001), vascular invasion (P = 0.01), tumour size (P = 0.06) and reduced lymphocyte infiltration (P = 0.03) were significant independent predictors of recurrence. The presence of Foxp3+ T-lymphocytes was not predictive of recurrence, but was associated with vascular invasion (FE = 9.02, P = 0.04). CONCLUSIONS The data support the hypothesis that immune responses are important in HCC and that the phenotype of infiltrating lymphocytes is informative regarding prognosis.
Collapse
Affiliation(s)
- Esther Unitt
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's NHS Trust, Hills Road, Cambridge CB2 2QQ, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Liu C, Leung MYK, Koon JCM, Zhu LF, Hui YZ, Yu B, Fung KP. Macrophage activation by polysaccharide biological response modifier isolated from Aloe vera L. var. chinensis (Haw.) Berg. Int Immunopharmacol 2006; 6:1634-41. [PMID: 16979117 DOI: 10.1016/j.intimp.2006.04.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Revised: 04/25/2006] [Accepted: 04/25/2006] [Indexed: 11/26/2022]
Abstract
A mannose-rich polysaccharide biological response modifier (BRM), derived from Aloe vera L. var. chinensis (Haw.) Berg., was demonstrated to be a potent murine B- and T-cell stimulator in our previous study. We here report the stimulatory activity of PAC-I on murine peritoneal macrophage. The polysaccharide when injected into mice enhanced the migration of macrophages to the peritoneal cavity. Peritoneal macrophage when treated by PAC-I in vitro had increased expression of MHC-II and FcgammaR, and enhanced endocytosis, phagocytosis, nitric oxide production, TNF-alpha secretion and tumor cell cytotoxicity. The administration of PAC-I into allogeneic ICR mice stimulated systemic TNF-alpha production in a dose-dependent manner and prolonged the survival of tumor-bearing mice. PAC-I is thus a potent stimulator of murine macrophage and the in vitro observed tumoricidal properties of activated macrophage might account for the in vivo antitumor properties of PAC-I. Our research findings may have therapeutic implications in tumor immunotherapy.
Collapse
Affiliation(s)
- C Liu
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
49
|
Pinheiro SP, Schernhammer ES, Tworoger SS, Michels KB. A Prospective Study on Habitual Duration of Sleep and Incidence of Breast Cancer in a Large Cohort of Women. Cancer Res 2006; 66:5521-5. [PMID: 16707482 DOI: 10.1158/0008-5472.can-05-4652] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mounting evidence suggests habitual sleep duration is associated with various health outcomes; both short and long sleep duration have been implicated in increased risk of cardiovascular disease, diabetes, and all-cause mortality. However, data on the relation between sleep duration and cancer risk are sparse and inconclusive. A link between low levels of melatonin, a hormone closely related to sleep, and increased risk of breast cancer has recently been suggested but it is unclear whether duration of sleep may affect breast cancer risk. We explored the association between habitual sleep duration reported in 1986 and subsequent risk of breast cancer in the Nurses' Health Study using Cox proportional hazards models. During 16 years of follow-up, 4,223 incident cases of breast cancer occurred among 77,418 women in this cohort. Compared with women sleeping 7 hours, covariate-adjusted hazard ratios and 95% confidence intervals for those sleeping < or =5, 6, 8, and > or =9 hours were 0.93 (0.79-1.09), 0.98 (0.91-1.06), 1.05 (0.97-1.13), and 0.95 (0.82-1.11), respectively. A moderate trend in risk increase towards longer sleep duration was observed when analyses were restricted to participants who reported same sleep duration in 1986 and 2000 (P(trend) = 0.05). In this prospective study, we found no convincing evidence for an association between sleep duration and the incidence of breast cancer.
Collapse
Affiliation(s)
- Simone P Pinheiro
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA.
| | | | | | | |
Collapse
|
50
|
Scouten WT, Francis GL. Thyroid cancer and the immune system: a model for effective immune surveillance. Expert Rev Endocrinol Metab 2006; 1:353-366. [PMID: 30764074 DOI: 10.1586/17446651.1.3.353] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Differentiated thyroid cancers, including papillary and follicular variants, are a useful model with which to examine interactions between cancer and the immune system. Differentiated thyroid cancers are detected in only 20,000 individuals annually in the USA, but thyroid microcarcinomas (< 1 cm in diameter) are far more common. This suggests that the immune system might restrain the growth of these microcarcinomas. On the clinical level, patients with lymphocytes that infiltrate into papillary thyroid cancer have improved survival, supporting the notion that immune system activation might improve this. Together, these observations suggest that the growth and distant spread of thyroid carcinoma are suppressed by mechanisms of immune surveillance, possibly involving lymphocytes, macrophages and their secreted products. In this review, we examine the general hypothesis of immune surveillance and the data pertaining to the roles of lymphocytes, dendritic cells and cytokines in the immune response against thyroid cancers.
Collapse
Affiliation(s)
- William T Scouten
- a Division of Pediatric Endocrinology, Portsmouth Naval Medical Center, 620 John Paul Jones Circle, Portsmouth, VA 23708, USA.
| | - Gary L Francis
- b Division of Pediatric Endocrinology, Virginia Commonwealth University, Medical College of Virginia, PO Box 980140, Richmond, VA 23298, USA.
| |
Collapse
|