1
|
Malvicini M, Piccioni F, Bayo J, Fiore E, Atorrasagasti C, Alaniz L, Garcia M, Aquino JB, Gidekel M, Matar P, Mazzolini G. Chemoimmunotherapy for advanced gastrointestinal carcinomas: A successful combination of gene therapy and cyclophosphamide. Oncoimmunology 2021; 1:1626-1628. [PMID: 23264916 PMCID: PMC3525625 DOI: 10.4161/onci.21651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The combination of a single low dose of cyclophosphamide (Cy) with the adenovirus-mediated gene transfer of interleukin-12 (AdIL-12) might represent a successful therapy for experimental gastrointestinal tumors. This approach has been proven to revert immunosuppressive mechanisms elicited by cancer cells and to synergistically promote antitumor immunity. In addition, this therapeutic regimen has been shown to be more efficient in achieving complete tumor regressions in mice than the application of a metronomic schedule of Cy plus AdIL-12.
Collapse
Affiliation(s)
- Mariana Malvicini
- Gene Therapy Laboratory; Liver Unit; School of Medicine; Austral University; Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Afshari F, Soleyman-Jahi S, Keshavarz-Fathi M, Roviello G, Rezaei N. The promising role of monoclonal antibodies for gastric cancer treatment. Immunotherapy 2019; 11:347-364. [PMID: 30678552 DOI: 10.2217/imt-2018-0093] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is the second leading cause of cancer-related death world-wide. Despite improvements in prevention, early detection and various therapeutic options, the prognosis is still poor. GC is often diagnosed at an advanced stage with survivals less than 1 year. Chemotherapy as the mainstay of treatment in advanced stage is not of notable advantages, underlining the need for novel more effective therapeutic options. Based on current knowledge of molecular and cellular mechanisms, a number of novel biologic approaches such as monoclonal antibodies have been recently introduced for cancer treatment that mainly affect the immune system or target signaling pathways playing role in cancer and metastasis development. In this review, various monoclonal antibodies for GC therapy were explained.
Collapse
Affiliation(s)
- Farzaneh Afshari
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Saeed Soleyman-Jahi
- Digestive Diseases Research Cores Center, Division of Gastroenterology, School of Medicine, Washington University, St. Louis, USA
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), St. Louis, USA
- Cancer Research Center, Cancer Institute of Iran, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Giandomenico Roviello
- Medical Oncology Unit, Department of Oncology, San Donato Hospital, Via Nenni 20, Arezzo, Italy
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Arezzo, Italy
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Sheffield, UK
| |
Collapse
|
3
|
Niccolai E, Taddei A, Prisco D, Amedei A. Gastric cancer and the epoch of immunotherapy approaches. World J Gastroenterol 2015; 21:5778-5793. [PMID: 26019442 PMCID: PMC4438012 DOI: 10.3748/wjg.v21.i19.5778] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 02/19/2015] [Accepted: 04/17/2015] [Indexed: 02/06/2023] Open
Abstract
The incidence of gastric cancer (GC) fell dramatically over the last 50 years, but according to IARC-Globocan 2008, it is the third most frequent cause of cancer-related deaths with a case fatality GC ratio higher than other common malignancies. Surgical resection is the primary curative treatment for GC though the overall 5-year survival rate remains poor (approximately 20%-25%). To improve the outcome of resectable gastric cancer, different treatment strategies have been evaluated such as adjuvant or perioperative chemotherapy. In resected gastric cancer, the addition of radiotherapy to chemotherapy does not appear to provide any additional benefit. Moreover, in metastatic patients, chemotherapy is the mainstay of palliative therapy with a median overall survival of 8-10 mo and objective response rates of merely 20%-40%. Therefore, the potential for making key beneficial progress is to investigate the GC molecular biology to realize innovative therapeutic strategies, such as specific immunotherapy. In this review, we provide a panoramic view of the different immune-based strategies used for gastric cancer treatment and the results obtained in the most significant clinical trials. In detail, firstly we describe the therapeutic approaches that utilize the monoclonal antibodies while in the second part we analyze the cell-based immunotherapies.
Collapse
|
4
|
Wang J, Reiss KA, Khatri R, Jaffee E, Laheru D. Immune Therapy in GI Malignancies: A Review. J Clin Oncol 2015; 33:1745-53. [PMID: 25918295 DOI: 10.1200/jco.2015.60.7879] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The balance between tumor-promoting and tumor-suppressing immune responses and the difference between them ultimately determine whether a cancer escapes immune recognition mechanisms. Defining the complex relationships between the tumor itself, the tumor environment, and the immune system has been critical in facilitating the development of successful immunotherapies. This review explores the role of oncogenes in inducing cancer-associated inflammation, the local and systemic factors that lead to immune suppression, and immunotherapy approaches to overcome immune privilege.
Collapse
Affiliation(s)
- Judy Wang
- All authors: Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Sidney Kimmel Cancer Center at The Johns Hopkins University, Baltimore, MD
| | - Kim A Reiss
- All authors: Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Sidney Kimmel Cancer Center at The Johns Hopkins University, Baltimore, MD
| | - Rina Khatri
- All authors: Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Sidney Kimmel Cancer Center at The Johns Hopkins University, Baltimore, MD
| | - Elizabeth Jaffee
- All authors: Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Sidney Kimmel Cancer Center at The Johns Hopkins University, Baltimore, MD
| | - Dan Laheru
- All authors: Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Sidney Kimmel Cancer Center at The Johns Hopkins University, Baltimore, MD
| |
Collapse
|
5
|
Amedei A, Benagiano M, della Bella C, Niccolai E, D'Elios MM. Novel immunotherapeutic strategies of gastric cancer treatment. J Biomed Biotechnol 2011; 2011:437348. [PMID: 22253528 PMCID: PMC3255571 DOI: 10.1155/2011/437348] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 09/26/2011] [Indexed: 12/23/2022] Open
Abstract
Gastric cancer (GC) is the fourth most common cancer and the second most frequent cause of cancer-related deaths, accounting for 10.4% of cancer deaths worldwide. Despite the improvements, estimated cure rates for patients with advanced stages remain poor, and in the metastatic setting, chemotherapy is the mainstay of palliative therapy and results in objective response rates (ORRs) of only 20-40% and median overall survivals (OS) of 8-10 months. Therefore, many investigators believe that the potential for making significant progress lies in understanding and exploiting the molecular biology of these tumors to investigate new therapeutic strategies to combat GC, such as specific immunotherapy. In this paper, we analyze the different approaches used for immune-based (especially dendritic and T cells) therapies to gastric cancer treatment and discuss the results obtained in preclinical models as in clinical trials.
Collapse
Affiliation(s)
- Amedeo Amedei
- Department of Internal Medicine, University of Florence, Viale Morgagni 85, 50134 Florence, Italy.
| | | | | | | | | |
Collapse
|
6
|
Harris F, Dennison SR, Singh J, Phoenix DA. On the selectivity and efficacy of defense peptides with respect to cancer cells. Med Res Rev 2011; 33:190-234. [PMID: 21922503 DOI: 10.1002/med.20252] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Here, we review potential determinants of the anticancer efficacy of innate immune peptides (ACPs) for cancer cells. These determinants include membrane-based factors, such as receptors, phosphatidylserine, sialic acid residues, and sulfated glycans, and peptide-based factors, such as residue composition, sequence length, net charge, hydrophobic arc size, hydrophobicity, and amphiphilicity. Each of these factors may contribute to the anticancer action of ACPs, but no single factor(s) makes an overriding contribution to their overall selectivity and toxicity. Differences between the anticancer actions of ACPs seem to relate to different levels of interplay between these peptide and membrane-based factors.
Collapse
Affiliation(s)
- Frederick Harris
- School of Forensic and Investigative Sciences, University of Central Lancashire, Preston, Lancashire, United Kingdom
| | | | | | | |
Collapse
|
7
|
Malvicini M, Ingolotti M, Piccioni F, Garcia M, Bayo J, Atorrasagasti C, Alaniz L, Aquino JB, Espinoza JA, Gidekel M, Scharovsky OG, Matar P, Mazzolini G. Reversal of gastrointestinal carcinoma-induced immunosuppression and induction of antitumoural immunity by a combination of cyclophosphamide and gene transfer of IL-12. Mol Oncol 2011; 5:242-55. [PMID: 21515097 DOI: 10.1016/j.molonc.2011.03.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 03/29/2011] [Accepted: 03/30/2011] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy-based strategies for gastrointestinal carcinomas (GIC) have been exploited so far, but these approaches have to face strong mechanisms of immune escape induced by tumours. We previously demonstrated that sub-therapeutic doses of an adenovirus expressing IL-12 genes (AdIL-12) mediated a potent antitumour effect against subcutaneous (s.c.) colorectal carcinomas (CRC) in mice pre-treated with low doses of cyclophosphamide (Cy). In our study we used this combination to assess its impact on the immunosuppressive microenvironment. In s.c. CRC model we demonstrated that non-responder mice failed to decrease Tregs in tumour, spleen and peripheral blood. Reconstitution of Tregs into tumour-bearing mice treated with combined therapy abolished the antitumoural effect. In addition, Cy + AdIL-12 modified Tregs functionality by inhibiting the in vitro secretion of IL-10 and TGF-β and their ability to inhibit dendritic cells activation. Combined treatment decreased the number of myeloid-derived suppressor cells (MDSCs) in comparison to non-treated mice and, interestingly, administration of Tregs restored splenic MDSCs population. Furthermore, combined therapy potently generated specific cytotoxic IFN-γ-secreting CD4+ T cells able to eradicate established CRC tumours after adoptive transfer. Finally, we evaluated the combination on disseminated CRC and pancreatic carcinoma (PC). Cy + AdIL-12 were able to eradicate liver metastatic CRC (47%) and PC tumour nodules (40%) and to prolong animal survival. The results of this study support the hypothesis that Cy + AdIL-12 might be a valid immunotherapeutic strategy for advanced GIC.
Collapse
Affiliation(s)
- Mariana Malvicini
- Gene Therapy Laboratory, Liver Unit, School of Medicine, Austral University, Derqui-Pilar, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Grimm M, Gasser M, Bueter M, Strehl J, Wang J, Nichiporuk E, Meyer D, Germer CT, Waaga-Gasser AM, Thalheimer A. Evaluation of immunological escape mechanisms in a mouse model of colorectal liver metastases. BMC Cancer 2010; 10:82. [PMID: 20205946 PMCID: PMC2838824 DOI: 10.1186/1471-2407-10-82] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 03/07/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The local and systemic activation and regulation of the immune system by malignant cells during carcinogenesis is highly complex with involvement of the innate and acquired immune system. Despite the fact that malignant cells do have antigenic properties their immunogenic effects are minor suggesting tumor induced mechanisms to circumvent cancer immunosurveillance. The aim of this study is the analysis of tumor immune escape mechanisms in a colorectal liver metastases mouse model at different points in time during tumor growth. METHODS CT26.WT murine colon carcinoma cells were injected intraportally in Balb/c mice after median laparotomy using a standardized injection technique. Metastatic tumor growth in the liver was examined by standard histological procedures at defined points in time during metastatic growth. Liver tissue with metastases was additionally analyzed for cytokines, T cell markers and Fas/Fas-L expression using immunohistochemistry, immunofluorescence and RT-PCR. Comparisons were performed by analysis of variance or paired and unpaired t test when appropriate. RESULTS Intraportal injection of colon carcinoma cells resulted in a gradual and time dependent metastatic growth. T cells of regulatory phenotype (CD4+CD25+Foxp3+) which might play a role in protumoral immune response were found to infiltrate peritumoral tissue increasingly during carcinogenesis. Expression of cytokines IL-10, TGF-beta and TNF-alpha were increased during tumor growth whereas IFN-gamma showed a decrease of the expression from day 10 on following an initial increase. Moreover, liver metastases of murine colon carcinoma show an up-regulation of FAS-L on tumor cell surface with a decreased expression of FAS from day 10 on. CD8+ T cells express FAS and show an increased rate of apoptosis at perimetastatic location. CONCLUSIONS This study describes cellular and macromolecular changes contributing to immunological escape mechanisms during metastatic growth in a colorectal liver metastases mouse model simulating the situation in human cancer.
Collapse
Affiliation(s)
- Martin Grimm
- Department of General Surgery, Molecular Oncology and Immunology, University of Wuerzburg Hospital, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Grávalos C, Grande E, Gasent JM. The potential role of sunitinib in gastrointestinal cancers other than GIST. Crit Rev Oncol Hematol 2010; 76:36-43. [PMID: 20133148 DOI: 10.1016/j.critrevonc.2010.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 12/16/2009] [Accepted: 01/06/2010] [Indexed: 01/09/2023] Open
Abstract
Gastrointestinal tumors are the most frequent and lethal malignancies worldwide. The deeper knowledge in molecular biology mechanisms involved in carcinogenesis has allowed the design of new targeted drugs mainly directed against the epidermal growth factor receptor (EGFR), the vascular endothelial growth factor (VEGF) and its receptors (VEGFRs). Sunitinib is an oral multitargeted inhibitor of the VEGF, platelet-derived growth factor (PDGF), and c-KIT, among others, tyrosine kinase receptors. Therefore, sunitinib acts in a dual mode as antiangiogenic agent and as antitumoral drug. The aim of this review is to gather the preclinical rationale behind the clinical use of sunitinib in gastrointestinal malignancies other than gastrointestinal stromal tumors (GIST) and to summarize the clinical data from phase I to III trials currently available.
Collapse
Affiliation(s)
- Cristina Grávalos
- Medical Oncology Department, 12 de Octubre Universitary Hospital, Avda. de Andalucía s/n Km 5.400, 28041 Madrid, Spain.
| | | | | |
Collapse
|
10
|
Amedei A, Niccolai E, Della Bella C, Cianchi F, Trallori G, Benagiano M, Bencini L, Bernini M, Farsi M, Moretti R, Del Prete G, D'Elios MM. Characterization of tumor antigen peptide-specific T cells isolated from the neoplastic tissue of patients with gastric adenocarcinoma. Cancer Immunol Immunother 2009; 58:1819-1830. [PMID: 19319530 PMCID: PMC11030125 DOI: 10.1007/s00262-009-0693-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 03/04/2009] [Indexed: 12/17/2022]
Abstract
Gastric cancer is a significant cause of morbidity and mortality worldwide. Surgical resection remains the primary curative treatment for gastric adenocarcinoma, but the poor (15-35%) survival rate at 5 years has prompted many studies for new therapeutic strategies, such as specific immunotherapy. The aim of this study was to analyze the functional properties of the T cell response to different antigen peptides related to gastric cancer in patients with gastric adenocarcinoma. To this purpose, we have cloned and characterized tumor-infiltrating T cells (TILs) isolated from the neoplastic gastric tissue samples. A T cell response specific to different peptides of gastric cancer antigens tested was documented in 17 out of 20 patients, selected for their HLA-A02 and/or -A24 alleles. Most of the cancer peptide-specific TILs expressed a Th1/Tc1 profile and cytotoxic activity against target cells. The effector functions of cancer peptide-specific T cells obtained from the peripheral blood of the same patients were also studied. The majority of peripheral blood peptide-specific T cells also expressed the Th1/Tc1 functional profile. In conclusion, in most of the patients with gastric adenocarcinoma, a specific type-1 T cell response to gastric cancer antigens was detectable and would have the potential of hamper tumor cell growth. However, in order to get tumor cell killing in vivo, the activity and the number of cancer peptide-specific Th1/Tc1 cells probably need to be enhanced by vaccination with the appropriate cancer antigenic peptides or by injection of the autologus tumor peptide-specific T cells expanded in vitro.
Collapse
Affiliation(s)
- Amedeo Amedei
- Department of Internal Medicine, University of Florence, Viale Morgagni 85, 50134 Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Shablak A, Hawkins RE, Rothwell DG, Elkord E. T cell-based immunotherapy of metastatic renal cell carcinoma: modest success and future perspective. Clin Cancer Res 2009; 15:6503-10. [PMID: 19843660 DOI: 10.1158/1078-0432.ccr-09-1605] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Metastatic renal cell carcinoma (MRCC) remains a challenging malignancy to treat. Cancer immunotherapies have been extensively explored in melanoma and RCC as they poorly respond to conventional cytotoxic agents but show responses to a variety of immunologic agents. The recent considerable success of T cell-based immunotherapy in melanoma warrants further efforts to apply this treatment to other cancers including MRCC. Although RCC is an immunosensitive cancer, similar attempts in MRCC have shown a very limited success. In this review, we summarize the clinical data on T cell-based immunotherapies for MRCC showing the modest success that has been achieved to date. More importantly, we discuss potential strategies for improving its efficacy for the treatment of MRCC in light of the important achievements for treating metastatic melanoma. In particular, the growing evidence of success by combining expanded tumor-infiltrating lymphocytes with lymphodepletion merits investigation in MRCC. Identifying new RCC-associated antigens, optimized methods, and conditions for detection, isolation, and/or modification and expansion of tumor-specific T cells are all important strategies to be pursued for improving T cell-based immunotherapy of MRCC.
Collapse
Affiliation(s)
- Alaaeldin Shablak
- Department of Medical Oncology, School of Cancer, Enabling Sciences and Technology, The University of Manchester, Manchester Academic Health Science Centre, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | | | | |
Collapse
|
12
|
Elkord E, Dangoor A, Burt DJ, Southgate TD, Daayana S, Harrop R, Drijfhout JW, Sherlock D, Hawkins RE, Stern PL. Immune evasion mechanisms in colorectal cancer liver metastasis patients vaccinated with TroVax (MVA-5T4). Cancer Immunol Immunother 2009; 58:1657-67. [PMID: 19221742 PMCID: PMC11029831 DOI: 10.1007/s00262-009-0674-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 01/27/2009] [Indexed: 01/01/2023]
Abstract
We have recently reported the results of a phase II trial in which two TroVax [modified vaccinia ankara (MVA) encoding the tumour antigen 5T4] vaccinations were given to patients both pre- and post-surgical resection of liver metastases secondary to colorectal cancer (CRC). 5T4-specific cellular responses were assessed at the entry and 2 weeks after each vaccination by proliferation of fresh lymphocytes and ELISA for antibody responses; 18 from the 19 CRC patients mounted a 5T4-specific cellular and/or humoral response. Here, we present a comparison of individual and between patient responses over the course of the treatments using cryopreserved peripheral blood mononuclear cells (PBMC) samples from the baseline until after the fourth vaccination at 14 weeks. Assays used were proliferation assay with 5T4-Fc fusion protein, overlapping 32mer 5T4 peptides, MVA-LacZ and MVA-5T4 infected autologous monocytes. Responses to 5T4 protein or one or more peptide pools were pre-existing in 12/20 patients and subsequently 10 and 12 patients showed boosted and/or de novo responses, respectively. Cumulatively, 13/20 patients showed proliferative responses by week 14. We also assessed the levels of systemic T regulatory cells, plasma cytokine levels, phenotype of tumour-infiltrating lymphocytes including T regulatory cells and tumour HLA class I loss of expression. More than half of the patients showed phenotypes consistent with relative immune suppression and/or escape highlighting the complexity of positive and negative factors challenging any simple correlation with clinical outcome.
Collapse
Affiliation(s)
- Eyad Elkord
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
- Department of Medical Oncology, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
- Present Address: Cellular Immunology Section, Laboratory of Immunology, National Institutes of Health, Bethesda, MD 20892 USA
| | - Adam Dangoor
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
- Department of Medical Oncology, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Deborah J. Burt
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Thomas D. Southgate
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Sai Daayana
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Richard Harrop
- Oxford BioMedica, Medawar Centre, Oxford Science Park, Oxford, OX4 4GA UK
| | - Jan W. Drijfhout
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, Netherlands
| | - David Sherlock
- Department of Surgery, North Manchester Healthcare NHS Trust, Manchester, UK
| | - Robert E. Hawkins
- Department of Medical Oncology, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| | - Peter L. Stern
- CR UK Immunology Group, Paterson Institute for Cancer Research, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester, M20 4BX UK
| |
Collapse
|
13
|
Matar P, Alaniz L, Rozados V, Aquino JB, Malvicini M, Atorrasagasti C, Gidekel M, Silva M, Scharovsky OG, Mazzolini G. Immunotherapy for liver tumors: present status and future prospects. J Biomed Sci 2009; 16:30. [PMID: 19272130 PMCID: PMC2662798 DOI: 10.1186/1423-0127-16-30] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Accepted: 03/06/2009] [Indexed: 12/22/2022] Open
Abstract
Increasing evidence suggests that immune responses are involved in the control of cancer and that the immune system can be manipulated in different ways to recognize and attack tumors. Progress in immune-based strategies has opened new therapeutic avenues using a number of techniques destined to eliminate malignant cells. In the present review, we overview current knowledge on the importance, successes and difficulties of immunotherapy in liver tumors, including preclinical data available in animal models and information from clinical trials carried out during the lasts years. This review shows that new options for the treatment of advanced liver tumors are urgently needed and that there is a ground for future advances in the field.
Collapse
Affiliation(s)
- Pablo Matar
- Institute of Experimental Genetics, School of Medical Sciences, National University of Rosario, Santa Fe 3100, (2000) Rosario, Argentina.
| | | | | | | | | | | | | | | | | | | |
Collapse
|