1
|
Zeng H, Li J, Hou K, Wu Y, Chen H, Ning Z. Melanoma and Nanotechnology-Based Treatment. Front Oncol 2022; 12:858185. [PMID: 35356202 PMCID: PMC8959641 DOI: 10.3389/fonc.2022.858185] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Melanoma is a malignant tumor arising in melanocytes from the basal layer of the epidermis and is the fifth most commonly diagnosed cancer in the United States. Melanoma is aggressive and easily metastasizes, and the survival rate is low. Nanotechnology-based diagnosis and treatment of melanoma have attracted increasing attention. Importantly, nano drug delivery systems have the advantages of increasing drug solubility, enhancing drug stability, prolonging half-life, optimizing bioavailability, targeting tumors, and minimizing side effects; thus, these systems can facilitate tumor cytotoxicity to achieve effective treatment of melanoma. In this review, we discuss current nanosystems used in the diagnosis and treatment of melanoma, including lipid systems, inorganic nanoparticles, polymeric systems, and natural nanosystems. The excellent characteristics of novel and effective drug delivery systems provide a basis for the broad applications of these systems in the diagnosis and treatment of melanoma, particularly metastatic melanoma.
Collapse
Affiliation(s)
- Hong Zeng
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Li
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Hou
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo Chen
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeng Ning
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Liu Q, Das M, Liu Y, Huang L. Targeted drug delivery to melanoma. Adv Drug Deliv Rev 2018; 127:208-221. [PMID: 28939379 DOI: 10.1016/j.addr.2017.09.016] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/29/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022]
Abstract
Melanoma derived from melanocytes is the most aggressive genre of skin cancer. Although the considerable advancement in the study of human cancer biology and drug discovery, most advanced melanoma patients are inevitably unable to be cured. With the emergence of nanotechnology, the use of nano-carriers is widely expected to alter the landscape of melanoma treatment. In this review, we will discuss melanoma biology, current treatment options, mechanisms behind drug resistance, and nano-based solutions for effective anti-cancer therapy, followed by challenges and perspectives in both pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Manisit Das
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
3
|
Zurac S, Neagu M, Constantin C, Cioplea M, Nedelcu R, Bastian A, Popp C, Nichita L, Andrei R, Tebeica T, Tanase C, Chitu V, Caruntu C, Ghita M, Popescu C, Boda D, Mastalier B, Maru N, Daha C, Andreescu B, Marinescu I, Rebosapca A, Staniceanu F, Negroiu G, Ion DA, Nikitovic D, Tzanakakis GN, Spandidos DA, Tsatsakis AM. Variations in the expression of TIMP1, TIMP2 and TIMP3 in cutaneous melanoma with regression and their possible function as prognostic predictors. Oncol Lett 2016; 11:3354-3360. [PMID: 27123116 PMCID: PMC4840923 DOI: 10.3892/ol.2016.4391] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/29/2016] [Indexed: 11/22/2022] Open
Abstract
Regression in melanoma is a frequent biological event of uncertain prognostic value as the lesion exhibits heterogeneous phenotypical features, both at the morphological and immunohistochemical level. In the present study, we examined the expression of tissue inhibitors of metalloproteinases (TIMP1, TIMP2 and TIMP3) in melanoma with regression. We specifically examined the expression levels of these TIMPs in regressed components (RC) and non-regressed components (NRC) of the tumor and compared their expression levels with those in non-regressed melanomas. We found that TIMP1 was overexpressed in the NRC of melanomas with partial regression (PR) compared with the NRC in melanomas with segmental regression (SR) (P=0.011). TIMP2 was overexpressed in the NRC of melanomas with PR compared with the NRC in melanomas with SR (PR/SR, P=0.009); or compared with the NRC in melanomas with simultaneous SR-PR (P=0.002); or compared with melanomas without regression (absence of regression) (P=0.037). Moreover, TIMP3 was overexpressed in the NRC of all melanomas with SR as compared to the RC component (P=0.007). Our findings on the differential expression of TIMP1, TIMP2 and TIMP3 in melanomas with regression support the hypothesis that the morphological differences identified in the melanoma regression spectrum may have a correlation with prognosis. This may explain the controversial findings within the literature concerning the biological and prognostic role of regression in melanoma.
Collapse
Affiliation(s)
- Sabina Zurac
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania; Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Monica Neagu
- Department of Immunology, 'Victor Babes' National Institute of Pathology, Bucharest 050096, Romania
| | - Carolina Constantin
- Department of Immunology, 'Victor Babes' National Institute of Pathology, Bucharest 050096, Romania
| | - Mirela Cioplea
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania; Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Roxana Nedelcu
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Alexandra Bastian
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania; Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Cristiana Popp
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania
| | - Luciana Nichita
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania; Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Razvan Andrei
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania
| | - Tiberiu Tebeica
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Cristiana Tanase
- Department of Immunology, 'Victor Babes' National Institute of Pathology, Bucharest 050096, Romania
| | - Virginia Chitu
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania; Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Constantin Caruntu
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Mihaela Ghita
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Catalin Popescu
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania; Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Daniel Boda
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Bogdan Mastalier
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania; Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Nicoleta Maru
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Claudiu Daha
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Bogdan Andreescu
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania
| | - Ioan Marinescu
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania
| | - Adrian Rebosapca
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania
| | - Florica Staniceanu
- Department of Pathology, Colentina University Hospital, Bucharest 020125, Romania; Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Gabriela Negroiu
- Institute of Biochemistry of Romanian Academy, Bucharest 060031, Romania
| | - Daniela A Ion
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Dragana Nikitovic
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, University of Crete, Heraklion 71003, Greece
| | - George N Tzanakakis
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, University of Crete, Heraklion 71003, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion 71409, Greece
| | - Aristidis M Tsatsakis
- Department of Forensic Sciences and Toxicology, Medical School, University of Crete, Heraklion 71003, Greece
| |
Collapse
|
4
|
Abstract
Vaccination started around the 10th century AD as a means of preventing smallpox. By the end of the 19th century such therapeutic vaccines were well established with both active and passive preparations being used in clinical practice. Active immunization involved administering an immunogen that might be live/ attenuated, killed/ inactivated, toxoid or subunit in origin. Passive immunization involved giving pre-formed antibodies, usually to very recently exposed individuals. At about the same time such approaches were also tried to treat a variety of cancers - proof of principle for the protective role of the immune response against malignancy was established by the observation that tumors transplanted into syngeneic hosts were rejected by the host innate and adaptive responses. The impact of these therapeutic vaccination has taken a considerable time to become established - in part because target antigens against which an adaptive response can be directed do not appear to be uniquely expressed on malignant transformed cells; and also because tumor cells are able to manipulate their environment to downregulate the host immune response. Therapeutic cancer vaccines are also divided into active and passive types - the latter being subdivided into specific and non-specific vaccines. Active immunization utilizes an immunogen to generate a host response designed to eliminate the malignant cells, whereas in passive immunization preformed antibodies or cells are administered to directly eliminate the transformed cells - examples of each are considered in this review.
Collapse
Affiliation(s)
- David Baxter
- a Honorary Lecturer; Manchester University Medical School; Manchester, UK
| |
Collapse
|
5
|
Seetharamu N, Ott PA, Pavlick AC. Novel therapeutics for melanoma. Expert Rev Anticancer Ther 2014; 9:839-49. [DOI: 10.1586/era.09.40] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
6
|
Srivastava PK, Duan F. Harnessing the antigenic fingerprint of each individual cancer for immunotherapy of human cancer: genomics shows a new way and its challenges. Cancer Immunol Immunother 2013; 62:967-74. [PMID: 23604106 PMCID: PMC3634982 DOI: 10.1007/s00262-013-1422-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 03/26/2013] [Indexed: 11/24/2022]
Abstract
The idea that individual tumors are antigenically unique has been around since the very dawn of our recognition of adaptive immune response to tumors. That idea has inspired a small number of attempts at individualized immunotherapy of human cancers. Such previous attempts for solid tumors have been hobbled by an inability to define the individually unique antigenic repertoire of tumors because of technological difficulties. The new availability of rapid and cheap high throughput DNA sequencing promises to overcome that hurdle. Using this new ability, coupled with bio-informatic tools, it is now possible to define the immunogenic repertoire of any tumor to a high degree of granularity within a practical time frame and an acceptable cost. The development of these ideas, and a small number of such studies that underscore this promise, is discussed. This new way--of characterizing the tumor immunome through characterization of the tumor genome--has distinct challenges, including selection of the appropriate peptides, choosing methods of immunizations that can incorporate tens of epitopes, and addressing issues of antigenic heterogeneity of tumors. However, tools for meeting these challenges exist and are emergent.
Collapse
Affiliation(s)
- Pramod K Srivastava
- Department of Immunology, Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT 06030, USA.
| | | |
Collapse
|
7
|
Klein O, Schmidt C, Knights A, Davis ID, Chen W, Cebon J. Melanoma vaccines: developments over the past 10 years. Expert Rev Vaccines 2011; 10:853-73. [PMID: 21692705 DOI: 10.1586/erv.11.74] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Decades of preclinical evaluation and clinical trials into melanoma vaccines have yielded spectacular progress in our understanding of melanoma antigens and the immune mechanisms of tumor rejection. Key insights and the results of their clinical evaluation are reviewed in this article. Unfortunately, durable clinical benefit following vaccination remains uncommon. Two recent clinical advances that will impact on melanoma vaccine development are trials with inhibitors of CTLA-4 and oncogenic BRAF. Long-term therapeutic control of melanoma will require integration of specific active immunotherapy with these emerging successful therapies from the disparate fields of immune regulation and signal transduction.
Collapse
Affiliation(s)
- Oliver Klein
- Ludwig Institute for Cancer Research, Austin Branch, Austin Hospital, Studley Road, Heidelberg, Victoria, 3084, Australia
| | | | | | | | | | | |
Collapse
|
8
|
Kluger HM, Hoyt K, Bacchiocchi A, Mayer T, Kirsch J, Kluger Y, Sznol M, Ariyan S, Molinaro A, Halaban R. Plasma markers for identifying patients with metastatic melanoma. Clin Cancer Res 2011; 17:2417-25. [PMID: 21487066 DOI: 10.1158/1078-0432.ccr-10-2402] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE With the rising incidence of melanoma, more patients are undergoing surveillance for disease recurrence. Our purpose was to study levels of proteins that might be secreted in the blood of patients with metastatic melanoma that can be used for monitoring these individuals. METHODS Genome-wide gene expression data were used to identify abundantly expressed genes in melanoma cells that encode for proteins likely to be present in the blood of cancer patients, based on high expression levels in tumors. ELISA assays were employed to measure proteins in plasma of 216 individuals; 108 metastatic melanoma patients and 108 age- and gender-matched patients with resected stage I/II disease split into equal-sized training and test cohorts. RESULTS Levels of seven markers, CEACAM (carcinoembryonic antigen-related cell adhesion molecule), ICAM-1 (intercellular adhesion molecule 1), osteopontin, MIA (melanoma inhibitory activity), GDF-15 (growth differentiation factor 15), TIMP-1 (tissue inhibitor of metalloproteinase 1), and S100B, were higher in patients with unresected stage IV disease than in patients with resected stage I/II disease. About 81% of the stage I/II patients in the training set had no marker elevation, whereas 69% of the stage IV patients had elevation of at least one marker (P < 0.0001). Receiver operating characteristic curves for the markers in combination in these two patient populations had an area under curve (AUC) of 0.79 in the training set and 0.8 in the test set. A CART (Classification and Regression Trees) model developed in the training set further improved the AUC in the test set to 0.898. CONCLUSIONS Plasma markers, particularly when assessed in combination, can be used to monitor patients for disease recurrence and can compliment currently used lactate dehydrogenase and imaging studies; prospective validation is warranted.
Collapse
Affiliation(s)
- Harriet M Kluger
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Abstract
After decades of research on solid tumor immunology, immunotherapy has shown effectiveness in patients with metastatic solid cancers. Immune modulators such as IL-2 and anti-CTLA-4 can mediate tumor regression in patients with metastatic melanoma and renal cancer, two tumor types that appear exceptional in their ability to spontaneously harbor endogenous antitumor immune cells. The responses can be long lasting, but the number of patients who benefit from these molecules remains limited. Combinations of these agents with cytotoxic and biologic agents are being investigated as a means to increase response rates and in an attempt to broaden application to other cancer types. Rare responses to cancer vaccines suggest that a better understanding of the underlying biology and mechanism of actions may lead to wider application in the future. The most effective form of immunotherapy thus far, capable of eradicating large tumor burdens in melanoma patients, is the ACT of TIL given to patients after lymphodepletion. As an alternative, lymphocytes engineered to recognize tumor-associated antigens can be safely infused to patients. With this approach, tumor regression is now being reported for cancers other than melanoma, but success remains constrained by the identification of antigens expressed with high specificity by cancer cells and not by normal tissues.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/immunology
- Cancer Vaccines/therapeutic use
- Cytotoxicity, Immunologic/immunology
- Genetic Engineering
- Humans
- Immunotherapy
- Ipilimumab
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/transplantation
- Melanoma/drug therapy
- Melanoma/secondary
- Neoplasm Metastasis/therapy
- Receptors, Antigen, T-Cell/drug effects
- Receptors, Antigen, T-Cell/immunology
- Skin Neoplasms/drug therapy
- Skin Neoplasms/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Simon Turcotte
- Surgery Branch, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | | |
Collapse
|
10
|
Itoh K, Yamada A, Mine T, Noguchi M. Recent advances in cancer vaccines: an overview. Jpn J Clin Oncol 2008; 39:73-80. [PMID: 19015149 DOI: 10.1093/jjco/hyn132] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The field of cancer vaccines is currently in an active state of clinical investigations. Human papilloma virus vaccine has been approved as a prophylactic cancer vaccine, while Oncophage (heat shock protein-peptide complex) was recently approved in Russia for a certain stage of kidney cancer, although to date none has been approved in Japan or the USA. We reviewed recent clinical trials of several different types of cancer vaccines, mainly by using PubMed from 2005 to 2008. There have been slow but substantial advances in peptide vaccines and dendritic cell-based vaccines with regard to both clinical responses and immunological markers. A personalized approach to boost immune responses, addition of chemotherapy to overcome robust cancers and changing of endpoints from tumor reduction to overall survival seem to be the three key elements for the development of therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Kyogo Itoh
- Department of Immunology and Immunotherapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka 830-0011, Japan.
| | | | | | | |
Collapse
|
11
|
Abstract
Immunotherapy for melanoma has undergone significant change since the first attempts to treat patients with high dose IL-2. Herein, strategies to boost patient antitumor immunity through vaccination, treatment with agents that augment host immunity, and adoptive cell transfer will be discussed. The first two strategies have yielded only limited clinical success, but adoptive cell transfer therapy, particularly following a lymphodepleting, preconditioning regimen has resulted in objective response rates approaching 50%. For a number of reasons, lymphodepletion appears to be critical for maintenance of circulating antitumor T cells following adoptive cell transfer. Balancing antitumor efficacy, autoimmunity, and reconstitution of a functioning immune system remain challenging and potentially life-threatening issues.
Collapse
Affiliation(s)
- Lei Fang
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Anke S Lonsdorf
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sam T Hwang
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
12
|
Siva A, Xin H, Qin F, Oltean D, Bowdish KS, Kretz-Rommel A. Immune modulation by melanoma and ovarian tumor cells through expression of the immunosuppressive molecule CD200. Cancer Immunol Immunother 2008; 57:987-96. [PMID: 18060403 PMCID: PMC11030565 DOI: 10.1007/s00262-007-0429-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Accepted: 11/13/2007] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND OBJECTIVE Immune escape by tumors can occur by multiple mechanisms, each a significant barrier to immunotherapy. We previously demonstrated that upregulation of the immunosuppressive molecule CD200 on chronic lymphocytic leukemia cells inhibits Th1 cytokine production required for an effective cytotoxic T cell response. CD200 expression on human tumor cells in animal models prevents human lymphocytes from rejecting the tumor; treatment with an antagonistic anti-CD200 antibody restored lymphocyte-mediated tumor growth inhibition. The current study evaluated CD200 expression on solid cancers, and its effect on immune response in vitro. METHODS AND RESULTS CD200 protein was expressed on the surface of 5/8 ovarian cancer, 2/4 melanoma, 2/2 neuroblastoma and 2/3 renal carcinoma cell lines tested, but CD200 was absent on prostate, lung, breast, astrocytoma, or glioblastoma cell lines. Evaluation of patient samples by immunohistochemistry showed strong, membrane-associated CD200 staining on malignant cells of melanoma (4/4), ovarian cancer (3/3) and clear cell renal cell carcinoma (ccRCC) (2/3), but also on normal ovary and kidney. CD200 expression on melanoma metastases was determined by RT-QPCR, and was found to be significantly higher in jejunum metastases (2/2) and lung metastases (2/6) than in normal samples. Addition of CD200-expressing, but not CD200-negative solid tumor cell lines to mixed lymphocyte reactions downregulated the production of Th1 cytokines. Inclusion of antagonistic anti-CD200 antibody restored Th1 cytokine responses. CONCLUSION These data suggest that melanoma, ccRCC and ovarian tumor cells can express CD200, thereby potentially suppressing anti-tumor immune responses. CD200 blockade with an antagonistic antibody may permit an effective anti-tumor immune response in these solid tumor types.
Collapse
Affiliation(s)
- A. Siva
- Alexion Antibody Technologies, Inc., 3985 Sorrento Valley Blvd, Ste A, San Diego, CA 92121 USA
| | - H. Xin
- Alexion Antibody Technologies, Inc., 3985 Sorrento Valley Blvd, Ste A, San Diego, CA 92121 USA
| | - F. Qin
- Alexion Antibody Technologies, Inc., 3985 Sorrento Valley Blvd, Ste A, San Diego, CA 92121 USA
| | - D. Oltean
- Alexion Antibody Technologies, Inc., 3985 Sorrento Valley Blvd, Ste A, San Diego, CA 92121 USA
| | - K. S. Bowdish
- Alexion Antibody Technologies, Inc., 3985 Sorrento Valley Blvd, Ste A, San Diego, CA 92121 USA
| | - A. Kretz-Rommel
- Alexion Antibody Technologies, Inc., 3985 Sorrento Valley Blvd, Ste A, San Diego, CA 92121 USA
| |
Collapse
|
13
|
Adjuvant treatment for high risk melanoma. Where are we now? Oncol Rev 2008. [DOI: 10.1007/s12156-008-0056-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
14
|
Skak K, Kragh M, Hausman D, Smyth MJ, Sivakumar PV. Interleukin 21: combination strategies for cancer therapy. Nat Rev Drug Discov 2008; 7:231-40. [DOI: 10.1038/nrd2482] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
Romero P. Current State of Vaccine Therapies in Non–Small-Cell Lung Cancer. Clin Lung Cancer 2008; 9 Suppl 1:S28-36. [DOI: 10.3816/clc.2008.s.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
16
|
Teng MWL, Westwood JA, Darcy PK, Sharkey J, Tsuji M, Franck RW, Porcelli SA, Besra GS, Takeda K, Yagita H, Kershaw MH, Smyth MJ. Combined natural killer T-cell based immunotherapy eradicates established tumors in mice. Cancer Res 2007; 67:7495-504. [PMID: 17671220 DOI: 10.1158/0008-5472.can-07-0941] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A rational monoclonal antibody (mAb)-based antitumor therapy approach has previously been shown to eradicate various established experimental and carcinogen-induced tumors in a majority of mice. This therapy comprised an agonistic mAb reactive with tumor necrosis factor-related apoptosis-inducing ligand receptor (DR5), expressed by tumor cells, an agonistic anti-CD40 mAb to mature dendritic cells, and an agonistic anti-4-1BB mAb to costimulate CD8(+) T cells. Because agonists of CD40 have been toxic in patients, we were interested in substituting anti-CD40 mAb with other dendritic cell-maturing agents, such as glycolipid ligands recognized by invariant natural killer T (iNKT) cells. Here, we show that CD1d-restricted glycolipid ligands for iNKT cells effectively substitute for anti-CD40 mAb and reject established experimental mouse breast and renal tumors when used in combination with anti-DR5 and anti-4-1BB mAbs (termed "NKTMab" therapy). NKTMab therapy-induced tumor rejection was dependent on CD4(+) and CD8(+) T cells, NKT cells, and the cytokine IFN-gamma. NKTMab therapy containing either alpha-galactosylceramide (alpha-GC) or alpha-C-galactosylceramide (alpha-c-GC) at high concentrations induced similar rates of tumor rejection in mice; however, toxicity was observed at the highest doses of alpha-GC (>250 ng/injection), limiting the use of this glycolipid. By contrast, even very low doses of alpha-c-GC (25 ng/injection) retained considerable antitumor activity when used in combination with anti-DR5/anti-4-1BB, and thus, alpha-c-GC showed a considerably greater therapeutic index. In summary, sequential tumor cell apoptosis and amplification of dendritic cell function by NKT cell agonists represents an exciting and novel approach for cancer treatment.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Apoptosis
- CD8-Positive T-Lymphocytes
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/therapy
- Cell Line, Tumor
- Combined Modality Therapy
- Galactosylceramides/immunology
- Immunotherapy
- Kidney Neoplasms/immunology
- Kidney Neoplasms/pathology
- Kidney Neoplasms/therapy
- Killer Cells, Natural/immunology
- Liver/immunology
- Lymphocyte Activation
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred BALB C
- Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology
- Survival Rate
- TNF-Related Apoptosis-Inducing Ligand
- Transaminases/blood
Collapse
Affiliation(s)
- Michele W L Teng
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zhang S, Wang Q, Miao B. Review: Dendritic Cell-Based Vaccine in the Treatment of Patients with Advanced Melanoma. Cancer Biother Radiopharm 2007; 22:501-7. [PMID: 17803444 DOI: 10.1089/cbr.2007.354] [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] [Indexed: 11/13/2022] Open
Abstract
Traditionally, immunology is mainly about the study of the immune response against foreign antigens, such as bacteria and viruses. Accordingly, tumor cells expressing alien or altered antigens make the attractive targets against which cancer immunology is initiated. However, recent comprehensive studies demonstrated that most prevalent antigens recognized by our immune system in cancer are those shared, nonmutated self-antigens expressed also by normal tissue cells. Thus, how to break the self-tolerance and avoid the concomitant autoimmunity remain the two challenges in cancer immunology. Dendritic cells (DCs) are the most effective antigen-presenting cells. They are capable of capturing, processing, and presenting antigens to T- and B-cells. This feature targets dendritic cells as the ideal candidates for breaking self-tolerance in cancer immunology. Clinical trials have demonstrated that dendritic cells are effective messengers. They circulate around the body and stimulate cytotoxic T-lymphocytes to clear tumor cells. Vaccination with DCs led to a clinical response in patients with melanoma, specifically those without significant autoimmunity. In this paper, we will examine the strategies and efficacies of DC-based vaccinations in the treatment of patients with melanoma.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Medical Oncology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China.
| | | | | |
Collapse
|
18
|
Terando AM, Faries MB, Morton DL. Vaccine therapy for melanoma: current status and future directions. Vaccine 2007; 25 Suppl 2:B4-16. [PMID: 17646038 DOI: 10.1016/j.vaccine.2007.06.033] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Revised: 06/11/2007] [Accepted: 06/13/2007] [Indexed: 12/21/2022]
Abstract
A vaccine is typically defined as any preparation used as a preventive inoculation to confer immunity against a specific disease. Vaccines for infectious diseases are highly effective, acting by inducing antigen-specific immunity that prevents subsequent infection. Unfortunately, the success of vaccines in infectious diseases has not been mirrored in oncology. This failure is the result of several challenges facing cancer vaccines, including the conceptual shift from disease prevention to disease treatment, tumor-induced immunosuppression and other mechanisms of immune escape, the similarity between tumor antigens and self antigens to which the patient is tolerant, unfavorable effector-to-target ratios in patients with established tumors, and financial and regulatory issues. Despite this, cancer remains a promising target for vaccine therapy. Melanoma in particular is known for its inherent immunogenicity on the basis of many anecdotal reports of spontaneous immune-based tumor regression, and thus has been the focus of immunotherapeutic approaches. Rare but significant vaccine-induced clinical regression of melanoma has spurred intensive investigations to augment vaccine efficacy. This review explores the many vaccine strategies that have been clinically tested for the treatment of melanoma and considers future approaches of cancer immunotherapy.
Collapse
Affiliation(s)
- Alicia M Terando
- John Wayne Cancer Institute at Saint John's Health Center, 2200 Santa Monica Boulevard, Santa Monica, CA 90404, USA
| | | | | |
Collapse
|
19
|
Abstract
Cytoreductive surgery represents a therapeutic attempt to improve patient outcomes by reducing overall tumor burden to render postsurgical therapy effective or at least increase its effectiveness. The intent of cytoreduction differs from palliative or curative-intent surgery for oligometastatic melanoma. Both palliative surgery and attempted curative resection have important roles to play in the management of patients with melanoma that has spread beyond the regional nodes or recurred "in transit" between the primary and the regional nodal basin. To date, however, no evidence shows that cytoreductive surgery offers any meaningful benefit to patients with metastatic melanoma, and, outside of a clinical trial, there is no role for cytoreductive surgery in melanoma. To date, adjuvant vaccine therapy after complete resection of metastatic melanoma has not proved to be efficacious in clinical trials, so there is little reason to believe that the use of currently available immunotherapy strategies will be enhanced after incomplete tumor resections.
Collapse
Affiliation(s)
- James M McLoughlin
- Division of Surgical Oncology, Department of Surgey, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | |
Collapse
|
20
|
Abstract
There are a large number of tumor antigens, which may either be specific to the tumor or inappropriately expressed or processed (tumor-associated antigen, TAA). Over the last few years, hundreds of new TAAs have been identified. Some of these represent good targets for both passive (antibody based) and active (vaccine based) therapies. Antibody treatments targeted on tumor-specific antigens, such as Herceptin and Cetuximab, have been effective in clinical trials and are now licensed. In addition, TAAs act as good surrogate markers for use in both the diagnosis and assessment of treatment in cancer patients.
Collapse
Affiliation(s)
- Angus Dalgleish
- St. George's University of London, Cranmer, London SW17 0RE, United Kingdom
| | | |
Collapse
|
21
|
Abstract
With increasing knowledge of tumor-associated antigens and T cell epitopes, and the mechanisms of induction and regulation of T-cellular immune responses, therapeutic vaccination is increasingly being explored as a treatment option for cancer. Several clinical cancer vaccination trials, the majority of them with melanoma patients, have demonstrated efficient induction of tumor-specific cellular immune responses in patients. However, these immune responses, in most cases, do not translate into clinical responses. The clinical response rates in these trials are relatively low. The most likely causes for the lack of correlation of immunological and clinical responsiveness are loss of antigenicity and immune suppression. Nonetheless, many patients in the vaccination trials have experienced extended survival compared to clinical experience. Therapeutic vaccination thus appears suited for maintenance therapy where cure is not possible and is an interesting option for adjuvant therapy after surgical tumor resection. While the clinical efficacy of vaccination is expected to be better for early-stage cancer, advancement of the treatment of advanced-stage disease will require combination with other therapeutic principles.
Collapse
Affiliation(s)
- Peter Walden
- Department of Dermatology, Venerology and Allergy, Clinical Research Group Tumor Immunology, Berlin, Germany
| |
Collapse
|
22
|
Jack A, Boyes C, Aydin N, Alam K, Wallack M. The treatment of melanoma with an emphasis on immunotherapeutic strategies. Surg Oncol 2006; 15:13-24. [PMID: 16815006 DOI: 10.1016/j.suronc.2006.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Melanoma continues to be one of the most difficult to treat of all solid tumors. Many new advances have been made in the surgical management of melanoma, including new guidelines for margins of excision, as well as sentinel node biopsy for the diagnosis of lymph node micrometastases. The search continues for an effective adjuvant melanoma treatment that can prevent local and distant recurrences. Melanoma is one of the most immunogenic of all tumors, and several clinical trials testing the immunotherapy of melanoma have been conducted, including trials in interferon, interleukin-2, and melanoma vaccines. Here we discuss many of the recent clinical trials in the surgical management of melanoma, in addition to the advances that have been made in the field of immunotherapy. A new second-generation melanoma vaccine, DC-MelVac (patent # 11221/5), has recently been granted FDA approval for Phase I clinical trials and will be introduced in this review.
Collapse
Affiliation(s)
- Angela Jack
- Surgery Research Laboratory, Department of Surgery, Saint Vincent's Catholic Medical Centers/New York Medical College, 153 West 11th Street, Cronin Building, Room 667, New York, NY 10011, USA
| | | | | | | | | |
Collapse
|