51
|
Abstract
One of the hallmarks of advanced malignancies is continuous cell growth and this almost universally correlates with the reactivation of telomerase. Although there is still much we do not understand about the regulation of telomerase, it remains a very attractive and novel target for cancer therapeutics. Several clinical trials have been initiated, and in this review we highlight some of the most promising approaches and conclude by speculating on the role of telomerase in cancer stem cells.
Collapse
Affiliation(s)
- J W Shay
- Department of Cell Biology, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75390-9030, USA.
| | | |
Collapse
|
52
|
Domchek SM, Recio A, Mick R, Clark CE, Carpenter EL, Fox KR, DeMichele A, Schuchter LM, Leibowitz MS, Wexler MH, Vance BA, Beatty GL, Veloso E, Feldman MD, Vonderheide RH. Telomerase-specific T-cell immunity in breast cancer: effect of vaccination on tumor immunosurveillance. Cancer Res 2007; 67:10546-55. [PMID: 17974999 DOI: 10.1158/0008-5472.can-07-2765] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The human telomerase reverse transcriptase (hTERT) is nearly universally overexpressed in human cancer, contributes critically to oncogenesis, and is recognized by cytotoxic T cells that lyse tumors. CD8+ T cells specific for hTERT naturally occur in certain populations of cancer patients in remission, but it remains poorly understood whether such T cells could contribute to tumor immunosurveillance. To address this issue, we induced hTERT-specific T cells in vivo via peptide vaccination in 19 patients with metastatic breast cancer who otherwise had no measurable T-cell responses to hTERT at baseline. Tumor-infiltrating lymphocytes (TIL) were evident after, but not before vaccination, with 4% to 13% of postvaccine CD8+ TIL specific for the immunizing hTERT peptide. Induction of TIL manifested clinically with tumor site pain and pruritus and pathologically with alterations in the tumor microenvironment, featuring histiocytic accumulation and widespread tumor necrosis. hTERT-specific CD8+ T cells were also evident after vaccination in the peripheral blood of patients and exhibited effector functions in vitro including proliferation, IFN-gamma production, and tumor lysis. An exploratory landmark analysis revealed that median overall survival was significantly longer in those patients who achieved an immune response to hTERT peptide compared with patients who did not. Immune response to a control cytomegalovirus peptide in the vaccine did not correlate with survival. These results suggest that hTERT-specific T cells could contribute to the immunosurveillance of breast cancer and suggest novel opportunities for both therapeutic and prophylactic vaccine strategies for cancer.
Collapse
Affiliation(s)
- Susan M Domchek
- Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Neal ZC, Sondel PM, Bates MK, Gillies SD, Herweijer H. Flt3-L gene therapy enhances immunocytokine-mediated antitumor effects and induces long-term memory. Cancer Immunol Immunother 2007; 56:1765-74. [PMID: 17426968 PMCID: PMC11030117 DOI: 10.1007/s00262-007-0320-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 03/19/2007] [Indexed: 12/12/2022]
Abstract
Therapeutic treatment with hu14.18-IL-2 immunocytokine (IC) or Flt3-L (FL) protein is initially effective at resolving established intradermal NXS2 neuroblastoma tumors in mice. However, many treated animals develop recurrent disease. We previously found that tumors recurring following natural killer (NK) mediated IC treatment show augmented MHC class I expression, while the tumors that recurred following T cell dependent Flt3-L treatment exhibited decreased MHC class I expression. We hypothesized that this divergent MHC modulation on recurrent tumors was due to therapy-specific immunoediting. We further postulated that combining IC and Flt3-L treatments might decrease the likelihood of recurrent disease by preventing MHC modulation as a mechanism for immune escape. We now report that combinatorial treatment of FL plus hu14.18-IL-2 IC provides greater antitumor benefit than treatment with either alone, suppressing development of recurrent disease. We administered FL by gene therapy using a clinically relevant approach: hydrodynamic limb vein (HLV) delivery of DNA for transgene expression by myofibers. Delivery of FL DNA by HLV injection in mice resulted in systemic expression of >10 ng/ml of FL in blood at day 3, and promoted up to a fourfold and tenfold increase in splenic NK and dendritic cells (DCs), respectively. Furthermore, the combination of FL gene therapy plus suboptimal IC treatment induced a greater expansion in the absolute number of splenic NK and DCs than achieved by individual component treatments. Mice that received combined FL gene therapy plus IC exhibited complete and durable resolution of established NXS2 tumors, and demonstrated protection from subsequent rechallenge with NXS2 tumor.
Collapse
Affiliation(s)
- Zane C. Neal
- Mirus Bio Corporation, 505 S. Rosa Road, Madison, WI 53719 USA
- Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI USA
| | - Paul M. Sondel
- Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI USA
- Departments of Pediatrics and Human Oncology, University of Wisconsin-Madison, Madison, WI USA
| | - Mary Kay Bates
- Mirus Bio Corporation, 505 S. Rosa Road, Madison, WI 53719 USA
| | | | - Hans Herweijer
- Mirus Bio Corporation, 505 S. Rosa Road, Madison, WI 53719 USA
| |
Collapse
|
54
|
Curigliano G, Rescigno M, Goldhirsch A. Immunology and breast cancer: therapeutic cancer vaccines. Breast 2007; 16 Suppl 2:S20-6. [PMID: 17706425 DOI: 10.1016/j.breast.2007.07.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Cancer immunosurveillance is a process that results from activity of recognition and destruction of cancer cells by innate and adaptive immune effector cells and molecules. Cancer cells can avoid immunosurveillance through the immunoselection, that is the development of poorly immunogenic tumor-cell variants, and through subversion of the immune system (also known as immunosubversion). Identification of tumor antigens (Ags) that can be recognized by immune effector cells has opened the perspective of developing therapeutic vaccines in the field of breast cancer. Breast cancer vaccines can induce immunogenic response against tumors weakly immunogenic; usually have a good tolerance and safety profile and can induce a long-term immune memory, critical to prevent efficiently tumor recurrence. Several studies evaluating breast cancer vaccines have been performed in patients with extended metastatic breast cancer, usually refractory to other standard treatments so that clinical efficacy was difficult to achieve. Significant immune responses against tumor Ags induced upon vaccinations were described to several tumor Ag vaccines. A better understanding of the relation between innate and adaptive immune responses, of the immune escape mechanisms employed by tumor cells and acknowledgment of the importance of both cell-mediated and humoral adaptive immunity for the control of tumor growth are necessary for leading to a more comprehensive immunotherapeutic approach in breast cancer.
Collapse
Affiliation(s)
- Giuseppe Curigliano
- Department of Medicine, Division of Medical Oncology, European Institute of Oncology, Via Ripamonti 435, 20141 Milano, Italy.
| | | | | |
Collapse
|
55
|
Prospects and challenges of building a cancer vaccine targeting telomerase. Biochimie 2007; 90:173-80. [PMID: 17716803 DOI: 10.1016/j.biochi.2007.07.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Accepted: 07/10/2007] [Indexed: 12/30/2022]
Abstract
Despite their origin from self-tissue, tumor cells can be immunogenic and trigger immune responses that can profoundly influence tumor growth and development. Clinically, it may be possible to amplify or induce anti-tumor immune responses to achieve tumor rejection in patients. Increasing data over the last 8 years suggest that the human telomerase reverse transcriptase hTERT is immunogenic both in vitro and in vivo and may be a suitable target for novel cancer immunotherapy. Peptides derived from hTERT are naturally processed by tumors and presented on MHC molecules and trigger effector functions of specific T lymphocytes. Vaccination of cancer patients against hTERT epitopes induces anti-tumor T cells without clinical toxicity. If second-generation vaccines and other strategies are able to generate optimal cellular immunity against hTERT without toxicity in humans, the possibility of broad-spectrum immunotherapy or even immunoprevention therapy of cancer may be possible.
Collapse
|
56
|
Abstract
One of the hallmarks of cancer is limitless proliferative capacity, which is tightly associated with the ability to maintain telomeres. Over the last decade, the telomere biology of pediatric cancers has begun to be elucidated. Most pediatric leukemias and embryonal solid tumors activate the enzyme telomerase, a specialized reverse transcriptase that adds nucleotide repeats to telomeres. In general, high levels of tumor telomerase expression are associated with unfavorable outcome, although results vary according to tumor type. Some pediatric tumors, including osteosarcoma and glioblastoma multiforme, lack telomerase activity and maintain telomeres via a recombination-based mechanism called ALT (alternative lengthening of telomeres). Telomerase is a highly attractive therapeutic target for pediatric cancer because the enzyme plays a key role in conferring cellular immortality, is present in most tumors, and is relatively specific for cancer cells. Telomerase inhibitors have been evaluated in preclinical models of adult cancers, but few studies have been conducted on pediatric cancers. Further research is required to define how telomere biology can be used to clinical advantage in malignancies of childhood.
Collapse
Affiliation(s)
- Uri Tabori
- Department of Hematology/Oncology, Hospital for Sick Children, Toronto, Canada
| | | |
Collapse
|
57
|
Waters JM, Richardson GD, Jahoda CAB. Hair follicle stem cells. Semin Cell Dev Biol 2007; 18:245-54. [PMID: 17481931 DOI: 10.1016/j.semcdb.2007.02.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2007] [Accepted: 02/05/2007] [Indexed: 12/17/2022]
Abstract
The increasing use of the hair follicle as a stem cell paradigm is due in part to the complex interplay between epithelial, dermal and other cell types, each with interesting differentiation potential and prospective therapeutic applications. This review focuses on research into the environmental niche, gene expression profiles and plasticity of hair follicle stem cell populations, where many recent advances have come about through novel technological and experimental approaches. We discuss major developmental pathways involved in the establishment and control of the epithelial stem cell niche, and evidence of plasticity between stem and transit amplifying cell populations.
Collapse
Affiliation(s)
- James M Waters
- Department of Biological Sciences, University of Durham, Durham, UK
| | | | | |
Collapse
|
58
|
Yamano T, Kaneda Y, Hiramatsu SH, Huang S, Tran AN, Giuliano AE, Hoon DSB. Immunity against breast cancer by TERT DNA vaccine primed with chemokine CCL21. Cancer Gene Ther 2007; 14:451-9. [PMID: 17318199 DOI: 10.1038/sj.cgt.7701035] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human telomerase reverse transcriptase (TERT) has been considered a potential tumor-associated antigen for active-specific immunotherapy. However, effective specific tumor antigen-specific immunity has been difficult to induce consistently by various TERT vaccine formulations. New adjuvant strategies have been employed, such as utilizing chemokines to attract T cells and antigen-presenting cells. Chemokine adjuvant strategies may enhance tumor antigen-specific immunity induced by vaccines. Therefore, we utilized chemokine ligand 21 (CCL21) as an adjuvant with a xenogeneic TERT DNA vaccine to induce tumor antigen-specific immunity against TERT-expressing breast cancer. The TERT DNA vaccine consisted of a plasmid containing the COOH terminal end of the TERT (cTERT) gene, encapsulated in multilayered liposomes with hemagglutinating virus of Japan coating. We demonstrated that CCL21 treatment before cTERT DNA vaccine, given intramuscularly, induced significantly higher anti-TERT specific cell-mediated immunity compared to cTERT DNA vaccine alone. Effective tumor antigen-specific immunity was shown both in prophylactic and therapeutic regimens against TS/A murine breast cancer. The study demonstrated that CCL21 administration before cTERT DNA vaccination significantly augmented tumor antigen-specific immunity against breast cancer.
Collapse
Affiliation(s)
- T Yamano
- Department of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | | | | | | | | | | | | |
Collapse
|
59
|
Ding ZY, Wu Y, Luo Y, Su JM, Li Q, Zhang XW, Liu JY, He QM, Yang L, Tian L, Zhao X, Deng HX, Wen YJ, Li J, Kang B, Wei YQ. Mannan-modified adenovirus as a vaccine to induce antitumor immunity. Gene Ther 2007; 14:657-63. [PMID: 17287861 DOI: 10.1038/sj.gt.3302893] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tumor vaccine is a useful strategy for cancer therapy. However, priming of the immune system requires the relevant antigen to be presented by antigen-presenting cells (APCs). Here, we employed telomerase reverse transcriptase as a model antigen to explore the feasibility of using mannan-modified adenovirus as a tumor vaccine. We found that tumor immunogene therapy with the vaccine was effective at protective antitumor immunity in mice. The antigen-specific cytotoxic T lymphocytes were found in in vitro cytotoxicity assay. The elevation of the killing activity could be abrogated by anti-CD8 or anti-major histocompatibility complex-I antibodies. Adoptive transfer of purified CD8+ cells, and CD4+ cells to a less extent, was effective at antitumor activity. In vivo antitumor activity could be abrogated by depleting CD4+ T lymphocytes. A possible explanation for the antitumor effects may be the antigen was transferred to APCs in the presence of mannan. These observations provide insights into the design of novel vaccine strategies and might be important for the future application of antigens identified in other diseases.
Collapse
Affiliation(s)
- Z-Y Ding
- Department of Oncology, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, West China Medical School, Sichuan University, Sichuan, The People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Abstract
Human telomerase reverse transcriptase (hTERT) represents a universal tumor-associated antigen to activate specific immune response in cancer immune therapy. Peptides derived from hTERT are presented by major histocompatibility complex (MHC) class I alleles to T lymphocytes, and CD8+ cytotoxic T lymphocytes (CTLs) specific for the hTERT-derived antigenic epitopes lyse hTERT-positive tumors from multiple histologies. These findings identify hTERT as an important tumor antigen widely applicable for anti-cancer immunotherapeutic strategies. The hTERT antigen-specific immunotherapy involves both active vaccination and adoptive immunotherapy approaches. Most importantly, the anti-tumor immune responses have been observed in the absence of toxicity, underlying the ongoing endeavors to develop immunotherapy directed against hTERT antigen. This chapter discusses most promising results and the approaches for investigation to target hTERT peptides as tumor antigens.
Collapse
Affiliation(s)
- He Li
- Centre for Functional Genomics and Human Disease, Monash University, Melbourne, Australia
| | | | | |
Collapse
|
61
|
Bendle GM, Holler A, Downs AM, Xue SA, Stauss HJ. Broadly expressed tumour-associated proteins as targets for cytotoxic T lymphocyte-based cancer immunotherapy. Expert Opin Biol Ther 2006; 5:1183-92. [PMID: 16120049 DOI: 10.1517/14712598.5.9.1183] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
T cell-based antigen-specific immunotherapy targeting self-proteins aberrantly expressed in many tumours offers the potential for widely applicable cancer immunotherapy, but carries the risk of autoimmunity. Immunological tolerance represents an inherent limitation of cancer vaccines targeting such broadly expressed tumour-associated proteins. Therefore, strategies to circumvent T cell tolerance have been developed and, when combined with T cell receptor (TCR) gene transfer technology, can generate highly avid tumour-reactive patient cytotoxic T lymphocytes (CTLs) specific for peptide epitopes of tumour-associated proteins. This review analyses the level of tolerance to broadly expressed tumour-associated proteins in the autologous T cell repertoire, assesses strategies that have been developed to circumvent T cell tolerance to such antigens, and evaluates the prospects for effective immunotherapy targeting broadly expressed tumour-associated proteins.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Clinical Trials as Topic
- Genetic Therapy
- Humans
- Immune Tolerance
- Immunotherapy/methods
- Models, Animal
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/therapy
- Proto-Oncogene Proteins c-mdm2/immunology
- Proto-Oncogene Proteins c-mdm2/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/transplantation
Collapse
Affiliation(s)
- Gavin M Bendle
- Royal Free Hospital, Department of Immunology and Molecular Pathology, University College London, London, NW3 2QG, UK
| | | | | | | | | |
Collapse
|
62
|
Chen L, Liang GP, Tang XD, Chen T, Cai YG, Fang DC, Yu ST, Luo YH, Yang SM. In vitro anti-tumor immune response induced by dendritic cells transfected with hTERT recombinant adenovirus. Biochem Biophys Res Commun 2006; 351:927-34. [PMID: 17097054 DOI: 10.1016/j.bbrc.2006.10.165] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2006] [Accepted: 10/25/2006] [Indexed: 02/04/2023]
Abstract
Transduction with recombinant, replication-defective adenoviral (Ad) vectors encoding a transgene is an efficient method for gene transfer into human dendritic cells (DC). Several studies have demonstrated that epitopes of the human telomerase reverse transcriptase gene (hTERT) can produce CTLs specific for malignant tumors. In this study, we constructed an hTERT recombinant adenovirus (rAd-hTERT) using DNA recombination. We found that human dendritic cells transduced with rAd-hTERT could effectively induce hTERT-specific cytotoxic T lymphocytes (CTLs) in vitro against various tumor cell lines, which were hTERT-positive and HLA-A2 matched. We also found that these hTERT-specific CTLs could not lyse autologous lymphocytes with low telomerase activity. Further studies revealed that rAd-hTERT transduced DCs could increase secretion of IFN-gamma by effector cells when they were co-cultured with hTERT-positive and HLA-A2 matched tumor cell lines. These data suggest that an hTERT vaccine can induce anti-tumor immunity against various tumor cells expressing hTERT in a HLA-A2-restricted fashion in vitro. The transduction of DCs with rAd-hTERT offers a great opportunity in cancer immunotherapy.
Collapse
Affiliation(s)
- Ling Chen
- Institute of Gastroenterology of PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Canales BK, Li Y, Thompson MG, Gleason JM, Chen Z, Malaeb B, Corey DR, Herbert BS, Shay JW, Koeneman KS. Small molecule, oligonucleotide-based telomerase template inhibition in combination with cytolytic therapy in an in vitro androgen-independent prostate cancer model. Urol Oncol 2006; 24:141-51. [PMID: 16520278 DOI: 10.1016/j.urolonc.2005.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE Determine the efficacy and timing of small molecule oligonucleotide-based inhibitors to the enzyme telomerase in an in vitro model of androgen-independent, osseous prostate cancer. MATERIALS AND METHODS Telomerase was inhibited in prostate cancer cell lines C4-2/C4-2B and in controls by using small molecule antisense oligonucleotide-based inhibitors alone or in various combinations of small-dose Taxotere (sanofi-aventis, Bridgewater, NJ) and/or conditionally replication competent adenovirus (AD-BSP-E1a). After transfection and proliferation, telomerase telomeric repeat amplification protocol and telomere restriction fragment assays were performed, with specific times for evaluating telomere length. Specimens were stained for analysis with hematoxylin and eosin (H&E), terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL), and prostate-specific antigen (PSA). RESULTS C4-2/C4-2B cell lines had the shortest initial mean telomere length (approximately 2.5 kilobase [kb]) compared to PC-3 (approximately 5.5 kb). Dose-dependent inhibition of telomerase activity was seen using match oligonucleotide-based inhibitors to telomerase (50% inhibitory concentration 3-5 nm), whereas mismatch compound showed no telomerase inhibition. Significant growth delay and apoptosis in cell lines occurred after > 50 days of treatment. Cells treated with combination "triple therapy" (i.e., telomerase inhibitors, adenovirus, and Taxotere) had the highest amount of apoptosis. Compared to controls, all combination treatment groups had statistically significant reductions in prostate-specific antigen in the conditioned media. CONCLUSIONS Combining cytotoxic regimens with small molecule inhibitors to telomerase with oligonucleotide-based agents could be beneficial in controlling osseous hormone refractory prostate cancer, as evidenced by these in vitro, preclinical investigations. Telomerase inhibition needs to move into in vivo models and human studies.
Collapse
Affiliation(s)
- Benjamin K Canales
- Department of Urology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Lin X, Zhou C, Wang S, Wang D, Ma W, Liang X, Lin C, Wang Z, Li J, Guo S, Zhang Y, Zhang S. Enhanced antitumor effect against human telomerase reverse transcriptase (hTERT) by vaccination with chemotactic-hTERT gene-modified tumor cell and the combination with anti-4-1BB monoclonal antibodies. Int J Cancer 2006; 119:1886-96. [PMID: 16708388 DOI: 10.1002/ijc.22048] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Human telomerase reverse transcriptase (hTERT) represents an attractive target for cancer immunotherapy because hTERT is reactivated in most human tumors. In an attempt to develop an effective vaccine against most human cancers, we constructed chemotactic-hTERT vaccine. Two hTERT fragments encoding multiple cytotoxic T lymphocyte and T helper cell epitopes were fused as a tumor antigen (named Te). The plasmid based DNA vaccine (pCCL21-Te-Fc) was constructed by linking human CCL21 and IgG Fc gene sequences to each end of Te. In poorly immunogenic B16F10 mouse melanoma model, DNA (pCCL21-Te-Fc) vaccination significantly inhibited tumor growth and all of the mice were dead by day 52. The immunization with pCCL21-Te-Fc-modified tumor cells (B16/CCL21-Te-Fc) resulted in a higher antitumor effect than DNA vaccination and 25% of tumor-bearing mice achieved long-term survival (> 120 days). The combined therapy of B16/CCL21-Te-Fc plus anti-4-1BB MAbs further enhanced the immune response, resulting in 75% of tumor-bearing mice achieved long-term survival (> 120 days) in subcutaneous model and few lung nodules in pulmonary metastasis model. Rechallenge experiment showed that a persistent memory response was successfully induced by the combined therapy. In vivo depletion of lymphocytes indicated that CD8+ T cells were essential in the antitumor activity induced by B16/CCL21-Te-Fc plus anti-4-1BB MAbs, whereas NK cells and CD4+ T cells played substantial roles. The CTL activity induced by pCCL21-Te-Fc-transfected PBMCs specifically lysed a variety of human leukocyte antigen-matched and hTERT-positive human tumor cells, suggesting pCCL21-Te-Fc could serve as a vaccine against most human cancers.
Collapse
Affiliation(s)
- Xiaoyan Lin
- Department of Immunology, Cancer Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Abstract
The progression from the cloning of human telomerase reverse transcriptase (hTERT) in 1997 to the first clinical trials of hTERT as an antitumour immunotherapy target has been swift. hTERT is overexpressed in the vast majority of human cancers yet has limited expression in normal adult tissue. It plays a critical role in oncogenesis and may be expressed by cancer stem cells. However, despite being a self antigen, hTERT is immunogenic both in vitro and in vivo. Several Phase I studies of hTERT immunotherapy have been completed in patients with breast, prostate, lung and other cancers, and clinical and immunological results are encouraging. Immunotherapy induces functional, antitumour T cells in patients in the absence of clinical toxicity. The opportunity for vaccinating individuals as an immunoprevention strategy can also be envisioned for hTERT-based therapies.
Collapse
Affiliation(s)
- Erica L Carpenter
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, 551 BRBII/III, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | | |
Collapse
|
66
|
Chen YJ, Sheng WY, Huang PR, Wang TCV. Potent inhibition of human telomerase by U-73122. J Biomed Sci 2006; 13:667-74. [PMID: 16850179 DOI: 10.1007/s11373-006-9100-z] [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: 04/28/2006] [Accepted: 06/14/2006] [Indexed: 02/02/2023] Open
Abstract
Telomerase activity is repressed in normal human somatic cells, but is activated in most cancers, suggesting that telomerase may be an important target for cancer therapy. In this study, we report that U-73122, an amphiphilic alkylating agent that is commonly used as an inhibitor for phospholipase C, is also a potent and selective inhibitor of human telomerase. The inhibition of telomerase by U-73122 was attributed primarily to the pyrrole-2,5-dione group, since its structural analog U-73343 did not inhibit telomerase. In confirmation, we observed that telomerase was inhibited by N-ethylmaleimide, but not N-ethylsuccinimide. The IC(50) value of U-73122 for the in vitro inhibition of telomerase activity is 0.2 microM, which is comparable to or slightly more sensitive than that for phospholipase C. The inhibitory action of U-73122 on telomerase appears to be rather selective since the presence of externally added proteins did not protect the inhibition and the IC(50) values for the other enzymes tested in this study were at least an order of magnitude higher than that for telomerase. Furthermore, we demonstrate that U-73122 can inhibit telomerase in hematopoietic cancer cells. The potent and selective inhibition of telomerase by U-73122 raises the potential exploitation of this drug and other alkylating agents as telomerase inhibitor.
Collapse
Affiliation(s)
- Yi-Jui Chen
- Department of Molecular and Cellular Biology, Chang Gung University, Kwei-San, Tao-Yuan, 333, Taiwan
| | | | | | | |
Collapse
|
67
|
Mendrzyk F, Korshunov A, Benner A, Toedt G, Pfister S, Radlwimmer B, Lichter P. Identification of gains on 1q and epidermal growth factor receptor overexpression as independent prognostic markers in intracranial ependymoma. Clin Cancer Res 2006; 12:2070-9. [PMID: 16609018 DOI: 10.1158/1078-0432.ccr-05-2363] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Pathogenesis of ependymomas is still poorly understood and molecular markers for risk-adapted patient stratification are not available. Our aim was to screen for novel genomic imbalances and prognostic markers in ependymal tumors. EXPERIMENTAL DESIGN We analyzed 68 sporadic tumors by matrix-based comparative genomic hybridization using DNA microarrays containing >6,400 genomic DNA fragments. Novel recurrent genomic gains were validated by fluorescence in situ hybridization using a tissue microarray consisting of 170 intracranial ependymomas. Candidate genes were also tested for mRNA expression by quantitative real-time PCR, and protein expression was determined by immunohistochemistry on the tissue microarray. RESULTS Chromosomal gain of 1q correlated with pediatric patients (P = 0.004), intracranial ependymomas (P = 0.05), and tumors of grade III (P = 0.002). Gain of 1q21.1-32.1 was associated with tumor recurrence in intracranial ependymomas (P < 0.001). Furthermore, gain of 1q25 as determined by fluorescence in situ hybridization represented an independent prognostic marker for either recurrence-free survival (P < 0.001) or overall survival (P = 0.003). Recurrent gains at 5p15.33 covering hTERT were validated by immunohistochemistry, and elevated protein levels correlated with adverse prognosis (P = 0.01). In addition to frequent gains and high-level amplification of epidermal growth factor receptor (EGFR) at 7p11.2, immunohistochemistry revealed protein overexpression to be correlated with poor prognosis (P = 0.002). EGFR protein status subdivides intracranial grade II ependymomas into two different risk groups (P = 0.03) as shown by multivariate analysis. CONCLUSIONS Thus, the states of 1q25 and EGFR represent independent prognostic markers for intracranial ependymomas to identify patient subgroups with different risk profiles in further clinical investigations. Moreover, EGFR might serve as therapeutic target for more specific chemotherapy applications.
Collapse
Affiliation(s)
- Frank Mendrzyk
- Division of Molecular Genetics and Central Unit Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
68
|
Shay JW, Wright WE. Telomerase therapeutics for cancer: challenges and new directions. Nat Rev Drug Discov 2006; 5:577-84. [PMID: 16773071 DOI: 10.1038/nrd2081] [Citation(s) in RCA: 310] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It has been approximately a decade since telomerase was described as an almost universal marker for human cancer. Most human tumours not only express telomerase but also have very short telomeres, whereas telomerase activity is either reduced or absent in normal tissues, making the inhibition of telomerase an attractive target for cancer therapeutics. Here we review the current status of telomerase therapeutics and discuss future opportunities and challenges for telomerase research, including a possible relationship with cancer stem cells that could be a source of chemo-/radioresistance development in many advanced cancers.
Collapse
Affiliation(s)
- Jerry W Shay
- Department of Cell Biology, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9039, USA.
| | | |
Collapse
|
69
|
Abstract
Senescence has been considered a programmed cellular response, parallel to apoptosis, that is turned on when a cell reaches Hayflick's limit. Once cells enter the senescence program, they cease to proliferate and undergo a series of morphological and functional changes. Studies support a central role for Rb protein in controlling this process after it receives senescent signals from the p53 and p16 pathways. Cellular senescence is considered an essential contributor to the aging process and has been shown to be an important tumor suppression mechanism. In addition, emerging evidence suggests that senescence may also be involved in the pathogenesis of stem cell dysfunction and chronic human diseases. Under these circumstances cells undergo stress-induced premature senecence, which has several specific features. Focusing on endothelial cells, we discuss recent advances in our understanding of the stresses and their pathways that prompt the premature senescence response, evaluate their correlation with the apoptotic response, and examine their links to the development of chronic diseases and the impaired function of endothelial progenitor cells, with the emphasis on vasculopathy. Emerging novel therapeutic interventions based on recent experimental findings are also reviewed.
Collapse
Affiliation(s)
- Jun Chen
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, NY 10595, USA.
| | | |
Collapse
|
70
|
Adotévi O, Mollier K, Neuveut C, Cardinaud S, Boulanger E, Mignen B, Fridman WH, Zanetti M, Charneau P, Tartour E, Lemonnier F, Langlade-Demoyen P. Immunogenic HLA-B*0702-Restricted Epitopes Derived from Human Telomerase Reverse Transcriptase That Elicit Antitumor Cytotoxic T-Cell Responses. Clin Cancer Res 2006; 12:3158-67. [PMID: 16707616 DOI: 10.1158/1078-0432.ccr-05-2647] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The human telomerase reverse transcriptase (hTERT) is considered as a potential target for cancer immunotherapy because it is preferentially expressed in tumor cells. To increase the applicability of hTERT-based immunotherapy, we set out to identify CTL epitopes in hTERT restricted by HLA-B*0702 molecule, a common MHC class I allele. EXPERIMENTAL DESIGN HLA-B*0702-restricted peptides from hTERT were selected by using a method of epitope prediction and tested for their immunogenicity in human (in vitro) and HLA-B*0702 transgenic mice (in vivo). RESULTS All the six hTERT peptides that were predicted to bind to HLA-B*0702 molecule were found to induce primary human CTL responses in vitro. The peptide-specific CD8+ CTL lines were tested against various hTERT+ tumor cells. Although differences were observed according to the tumor origin, only three CTL lines specific for p277, p342, and p351 peptides exhibited cytotoxicity against tumor cells in a HLA-B*0702-restricted manner. In addition, this cytotoxicity was inhibited by the addition of peptide-loaded cold target cells and indicated that these epitopes are naturally processed and presented on the tumor cells. Further, in vivo studies using humanized HLA-B*0702 transgenic mice showed that all the candidate peptides were able to induce CTL responses after peptide immunization. Furthermore, vaccination with a plasmid DNA encoding full-length hTERT elicited peptide-specific CTL responses, indicating that these epitopes are efficiently processed in vivo. CONCLUSIONS Together with previously reported hTERT epitopes, the identification of new CTL epitopes presented by HLA-B*0702 increases the applicability of hTERT-based immunotherapy to treating cancer.
Collapse
Affiliation(s)
- Olivier Adotévi
- Institut National de la Sante et de la Recherche Medicale U255, Université René Descartes, Unité d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Assistance-Public Hôpitaux de Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Filaci G, Fravega M, Setti M, Traverso P, Millo E, Fenoglio D, Negrini S, Ferrera F, Romagnoli A, Basso M, Contini P, Rizzi M, Ghio M, Benatti U, Damonte G, Ravetti JL, Carmignani G, Zanetti M, Indiveri F. Frequency of telomerase-specific CD8+ T lymphocytes in patients with cancer. Blood 2006; 107:1505-12. [PMID: 16249379 DOI: 10.1182/blood-2005-01-0258] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Telomerase is considered a universal tumor-associated antigen (TAA) due to its high rate of expression by cancers (≈90%), and clinical trials are in progress to test the immunotherapeutical efficacy of antitelomerase immunization in patients with cancer. However, the data concerning frequency and functional activity of telomerase-specific cytotoxic T lymphocytes (CTLs) in patients with cancer are few and conflicting, although their knowledge would be mandatory to predict the efficacy of telomerase-specific immunotherapy in selected patients. We performed this study to analyze frequency and cytolytic function of circulating CD8+ T lymphocytes specific for the p540 telomerase peptide in a series of human leukocyte antigen (HLA)–A2+ cancer patients. The results show that most patients with cancer have circulating telomerase-specific CD8+ T lymphocytes, but a high frequency of telomerase-specific CTLs are present only in a fraction of them. Furthermore, CTL lines able to kill telomerase-positive tumor cells, including autologous cancer cells, can be expanded ex vivo from some, but not all, patients with cancer. In conclusion, the results of the study support the development of clinical protocols using telomerase peptides as an immunizing agent. However, they underline the necessity to study single patients immunologically before undergoing vaccination, to select the patients adequately, and to eventually adapt the immunization schedule to the patient's immunologic status.
Collapse
Affiliation(s)
- Gilberto Filaci
- Department of Internal Medicine and Center of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV no. 6, 16132 Genova, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Guo H, Hao J, Wu C, Fang DC. Construction of virus-like particle peptide-nucleic acid vaccine of human telomerase reverse transcriptase and identification of its immunogenicity. Shijie Huaren Xiaohua Zazhi 2005; 13:2645-2649. [DOI: 10.11569/wcjd.v13.i22.2645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct a novel virus-like particle peptide-nucleic acid vaccine (VPNV) of human telomerase reverse transcriptase (hTERT), and to identify its imm-unogenicity and transfection activity.
METHODS: Cationic antigenic peptide K18P9 was syn-thesized and purified, then human GM-CSF and TERT gene were cloned into eukaryotic expression vector pTCAE. The peptide was combined with the nucleic acid vaccine to make VPNV, which were transfected into eukaryotic cells COS-7. The immunogenicity of hGM-CSF and hTERT were detected by enzyme linked imm-unosorbent assay (ELISA) and Western blotting.
RESULTS: Restriction enzyme digestion and sequen-ce analysis confirmed that hGM-CSF and hTERT were cloned into pTCAE and the nucleic acid vaccine of hTERT gene was constructed successfully. Under ele-ctronic microscopy, nucleic acid was packaged by pep-tide, forming into virus-like particle. Furthermore, the transfection activity of VPNV and the immunogenic-ity of hGM-CSF and hTERT could reach 78.5% as co-mpared with the positive controls.
CONCLUSION: The VPNV is successfully constructed, and its immunogenicity is also identified.
Collapse
|
73
|
Curigliano G, Spitaleri G, Pietri E, Rescigno M, de Braud F, Cardillo A, Munzone E, Rocca A, Bonizzi G, Brichard V, Orlando L, Goldhirsch A. Breast cancer vaccines: a clinical reality or fairy tale? Ann Oncol 2005; 17:750-62. [PMID: 16293674 DOI: 10.1093/annonc/mdj083] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The characterization of tumor antigens recognized by immune effector cells has opened the perspective of developing therapeutic vaccines in the field of breast cancer. The potential advantages of the vaccines are: (i) the induction of a robust immune response against tumors that are spontaneously weekly immunogenic; (ii) the tumor specificity for some antigens; (iii) the good tolerance and safety profile and (iv) the long-term immune memory, critical to prevent efficiently tumor recurrence. Most trials evaluating breast cancer vaccines have been carried out in patients with extended metastatic breast cancer, characterized by aggressive tumors, resistant to standard cytotoxic treatments, so that clinical efficacy was difficult to achieve. However, some significant immune responses against tumor antigens induced upon vaccinations were recorded. The aim of this review is to analyze the activity of vaccination strategies in current clinical trials. Data of clinical activity have been observed by using vaccines targeting HER2/neu protein, human telomerase reverse transcriptase, carcinoembryonic antigen and carbohydrate antigen given after stem cell rescue. The review discusses possible future directions for vaccine development and applications in the adjuvant setting.
Collapse
Affiliation(s)
- G Curigliano
- Department of Medicine, Clinical Pharmacology and New Drugs Development Unit, Division of Experimental Oncology, Laboratory of Experimental Immunology, European Institute of Oncology, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Dikmen ZG, Gellert GC, Jackson S, Gryaznov S, Tressler R, Dogan P, Wright WE, Shay JW. In vivo inhibition of lung cancer by GRN163L: a novel human telomerase inhibitor. Cancer Res 2005; 65:7866-73. [PMID: 16140956 DOI: 10.1158/0008-5472.can-05-1215] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Differential regulation of telomerase activity in normal and tumor cells provides a rationale for the design of new classes of telomerase inhibitors. The telomerase enzyme complex presents multiple potential sites for the development of inhibitors. GRN163L, a telomerase enzyme antagonist, is a lipid-modified 13-mer oligonucleotide N3' --> P5'-thio-phosphoramidate, complementary to the template region of telomerase RNA (hTR). We evaluated both the in vitro and in vivo effects of GRN163L using A549-luciferase (A549-Luc) human lung cancer cells expressing a luciferase reporter. GRN163L (1 micromol/L) effectively inhibits telomerase activity of A549-Luc cells, resulting in progressive telomere shortening. GRN163L treatment also reduces colony formation in soft agar assays. Surprisingly, after only 1 week of treatment with GRN163L, A549-Luc cells were unable to form robust colonies in the clonal efficiency assay, whereas the mismatch control compound had no effect. Finally, we show that in vivo treatment with GRN163L is effective in preventing lung metastases in xenograft animal models. These in vitro and in vivo data support the development of GRN163L as a therapeutic for the treatment of cancer.
Collapse
Affiliation(s)
- Z Gunnur Dikmen
- University of Hacettepe, Faculty of Medicine, Department of Biochemistry, Ankara, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Danet-Desnoyers GAH, Luongo JL, Bonnet DA, Domchek SM, Vonderheide RH. Telomerase vaccination has no detectable effect on SCID-repopulating and colony-forming activities in the bone marrow of cancer patients. Exp Hematol 2005; 33:1275-80. [PMID: 16263411 DOI: 10.1016/j.exphem.2005.07.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Revised: 06/30/2005] [Accepted: 07/12/2005] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The telomerase reverse transcriptase hTERT is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes (CTL). We have previously shown that vaccination of cancer patients against hTERT induces functional anti-tumor CTL in vivo, but it is not known whether hTERT vaccination harms normal cells expressing the enzyme, especially hematopoietic stem cells and progenitors. PATIENTS AND METHODS We employed colony-forming cell (CFC) assays, long-term in vitro cultures, and nonobese diabetic/severe combined immunodeficient (NOD/SCID) repopulation studies to evaluate the effects of hTERT vaccination on hematopoietic progenitors and stem cells in cancer patients following treatment. RESULTS Using bone marrow samples obtained from cancer patients before and after vaccination, we found that there was no significant decline in the frequency of granulocyte, macrophage or erythroid CFCs using CFC assays or long-term in vitro cultures. In NOD/SCID mice, human hematopoietic reconstitution was easily detected, without quantitative or qualitative differences between pre- and postvaccine samples. CONCLUSION These findings suggest that induction of tumor-lytic hTERT-specific T cells in vivo by vaccination does not result in a detectable decline in hematopoietic potential despite the expression of hTERT and major histocompatibility complex class I in bone marrow progenitors and stem cells. Thus, even for self-antigens such as telomerase, tumor immunity does not necessarily involve autoimmunity in normal tissues that share the target.
Collapse
|
76
|
de Grey ADNJ, Alvarez PJJ, Brady RO, Cuervo AM, Jerome WG, McCarty PL, Nixon RA, Rittmann BE, Sparrow JR. Medical bioremediation: prospects for the application of microbial catabolic diversity to aging and several major age-related diseases. Ageing Res Rev 2005; 4:315-38. [PMID: 16040282 DOI: 10.1016/j.arr.2005.03.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2005] [Accepted: 03/09/2005] [Indexed: 11/30/2022]
Abstract
Several major diseases of old age, including atherosclerosis, macular degeneration and neurodegenerative diseases are associated with the intracellular accumulation of substances that impair cellular function and viability. Moreover, the accumulation of lipofuscin, a substance that may have similarly deleterious effects, is one of the most universal markers of aging in postmitotic cells. Reversing this accumulation may thus be valuable, but has proven challenging, doubtless because substances resistant to cellular catabolism are inherently hard to degrade. We suggest a radically new approach: augmenting humans' natural catabolic machinery with microbial enzymes. Many recalcitrant organic molecules are naturally degraded in the soil. Since the soil in certain environments - graveyards, for example - is enriched in human remains but does not accumulate these substances, it presumably harbours microbes that degrade them. The enzymes responsible could be identified and engineered to metabolise these substances in vivo. Here, we survey a range of such substances, their putative roles in age-related diseases and the possible benefits of their removal. We discuss how microbes capable of degrading them can be isolated, characterised and their relevant enzymes engineered for this purpose and ways to avoid potential side-effects.
Collapse
Affiliation(s)
- Aubrey D N J de Grey
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Schreurs MWJ, Kueter EWM, Scholten KBJ, Kramer D, Meijer CJLM, Hooijberg E. Identification of a potential human telomerase reverse transcriptase-derived, HLA-A1-restricted cytotoxic T-lymphocyte epitope. Cancer Immunol Immunother 2005; 54:703-12. [PMID: 15726360 PMCID: PMC11032920 DOI: 10.1007/s00262-004-0611-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2004] [Accepted: 08/23/2004] [Indexed: 12/18/2022]
Abstract
The catalytic subunit of human telomerase reverse transcriptase (hTERT) is expressed in the majority of tumor cells of different histological origins as opposed to most normal somatic cells. This implicates hTERT as a widely expressed tumor-associated antigen and an attractive candidate for antigen-specific tumor immunotherapy. T lymphocytes specific for hTERT-derived epitopes have been isolated and shown reactive with hTERT-expressing tumor cells. To further increase the applicability of hTERT as a target antigen for immunotherapy, we set out to identify potential hTERT-derived, HLA-A1-restricted cytotoxic T-lymphocyte (CTL) epitopes. The "reverse immunology" approach, involving computer-assisted epitope prediction, in vitro CTL induction, and tetramer-guided CTL isolation, resulted in specific CTLs against hTERT-derived, HLA-A1-binding peptides. Intermediate- to low-avidity CTLs were induced against the hTERT325-333 peptide and recognized endogenously processed hTERT. Recognition of endogenous hTERT depended on an increase of hTERT expression above normal levels in tumor cells through hTERT transduction, most probably as a result of limited CTL avidity. The altered peptide ligand hTERT699T-707 was designed to increase HLA-A1-binding affinity of the hTERT699-707 peptide and was used to induce CTLs. However, these CTLs poorly cross-recognized native hTERT699-707 and failed to recognize endogenously processed hTERT. In conclusion, our study has identified the hTERT325-333 peptide as a potential hTERT-derived epitope that may prove useful for induction and monitoring of hTERT-specific, HLA-A1-restricted CTL responses.
Collapse
Affiliation(s)
- Marco W. J. Schreurs
- Department of Pathology, VU University Medical Center, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Esther W. M. Kueter
- Department of Pathology, VU University Medical Center, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Kirsten B. J. Scholten
- Department of Pathology, VU University Medical Center, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Duco Kramer
- Department of Pathology, VU University Medical Center, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Chris J. L. M. Meijer
- Department of Pathology, VU University Medical Center, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Erik Hooijberg
- Department of Pathology, VU University Medical Center, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
78
|
Abstract
Telomerase is a ribonucleoprotein that directs the synthesis of telomeric sequence. It is detected in majority of malignant tumors, but not in most normal somatic cells. Because telomerase plays a critical role in cell immortality and tumor formation, it has been one of the targets for anti-cancer and regeneration drug development. In this review, we will discuss therapeutic approaches based mainly on small molecules that have been developed to inhibit telomerase activity, modulate telomerase expression, and telomerase directed gene therapy.
Collapse
Affiliation(s)
- Yi-hsin Hsu
- Institute of Biopharmaceutical Science, National Yang-Ming University, Taipei, Taiwan, China
| | | |
Collapse
|
79
|
|
80
|
|
81
|
Abstract
Although tumor vaccines have been studied for decades, there is no vaccine approved as a clinical product. Nevertheless, recent advances in immunology and tumor biology justify a renewed interest. First, cancer cells express many antigens that can be recognized by the immune system, some with high tumor selectivity. Second, knowledge about immune regulation, including the importance of costimulatory signals, has been successfully applied to the studies of tumors. Third, mechanisms of how tumors can escape from immunological control have been identified, setting the stage to discover agents to decrease their impact. Rejection of established mouse tumors has been accomplished as a result of therapeutic tumor vaccination and there are encouraging findings from vaccine trials in humans.
Collapse
|
82
|
Spiropoulou T, Ferekidou L, Angelopoulou K, Stathopoulou A, Talieri M, Lianidou ES. Effect of antineoplastic agents on the expression of human telomerase reverse transcriptase beta plus transcript in MCF-7 cells. Clin Biochem 2004; 37:299-304. [PMID: 15003732 DOI: 10.1016/j.clinbiochem.2003.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2003] [Revised: 12/10/2003] [Accepted: 12/11/2003] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To evaluate the effect of antineoplastic agents on the expression of human telomerase reverse transcriptase (hTERT) splice variants in MCF-7 cells. DESIGN AND METHODS We have developed a luminometric hybridization assay for hTERT beta plus transcript. MCF-7 cells were isolated before and after treatment with antineoplastic agents. A combination of nested RT-PCR and the developed luminometric hybridization assay was used for the specific detection of hTERT beta plus transcript in treated and untreated MCF-7 cells. Amplification of all hTERT splicing variants by nested PCR in the same samples was also performed. RESULTS MCF-7 cells treated with taxol and etoposide were found positive for all hTERT splicing variants, while the expression of hTERT beta plus transcript did not differ significantly before and after exposure. MCF-7 cells treated with doxorubicin and 5-fluorouracil did not express any of hTERT splicing variants. In the presence of cisplatin, three splicing variants of hTERT were detected. CONCLUSIONS The developed hybridization assay is highly sensitive and specific for the detection of hTERT beta plus transcript in clinical samples.
Collapse
Affiliation(s)
- Tonia Spiropoulou
- Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, 15771 Athens, Greece
| | | | | | | | | | | |
Collapse
|
83
|
Vonderheide RH, Domchek SM, Schultze JL, George DJ, Hoar KM, Chen DY, Stephans KF, Masutomi K, Loda M, Xia Z, Anderson KS, Hahn WC, Nadler LM. Vaccination of cancer patients against telomerase induces functional antitumor CD8+ T lymphocytes. Clin Cancer Res 2004; 10:828-39. [PMID: 14871958 DOI: 10.1158/1078-0432.ccr-0620-3] [Citation(s) in RCA: 203] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE High-level expression of the telomerase reverse transcriptase (hTERT) in >85% of human cancers, in contrast with its restricted expression in normal adult tissues, points to hTERT as a broadly applicable molecular target for anticancer immunotherapy. CTLs recognize peptides derived from hTERT and kill hTERT+ tumor cells of multiple histologies in vitro. Moreover, because survival of hTERT+ tumor cells requires functionally active telomerase, hTERT mutation or loss as a means of escape may be incompatible with sustained tumor growth. EXPERIMENTAL DESIGN A Phase I clinical trial was performed to evaluate the clinical and immunological impact of vaccinating advanced cancer patients with the HLA-A2-restricted hTERT I540 peptide presented with keyhole limpet hemocyanin by ex vivo generated autologous dendritic cells. RESULTS As measured by peptide/MHC tetramer, enzyme-linked immunospot, and cytotoxicity assays, hTERT-specific T lymphocytes were induced in 4 of 7 patients with advanced breast or prostate carcinoma after vaccination with dendritic cells pulsed with hTERT peptide. Tetramer-guided high-speed sorting and polyclonal expansion achieved highly enriched populations of hTERT-specific cells that killed tumor cells in an MHC- restricted fashion. Despite concerns of telomerase activity in rare normal cells, no significant toxicity was observed. Partial tumor regression in 1 patient was associated with the induction of CD8+ tumor infiltrating lymphocytes. CONCLUSIONS These results demonstrate the immunological feasibility of vaccinating patients against telomerase and provide rationale for targeting self-antigens with critical roles in oncogenesis.
Collapse
Affiliation(s)
- Robert H Vonderheide
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
|
85
|
Lane C, Leitch J, Tan X, Hadjati J, Bramson JL, Wan Y. Vaccination-induced autoimmune vitiligo is a consequence of secondary trauma to the skin. Cancer Res 2004; 64:1509-14. [PMID: 14973051 DOI: 10.1158/0008-5472.can-03-3227] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A major concern for cancer vaccines targeting self-tumor antigens is the risk of autoimmune sequelae. Although antitumor immunity correlates with autoimmune disease in some preclinical models, the mechanism(s) linking antitumor immunity and subsequent autoimmune pathology remain(s) to be determined. In the current study, we demonstrated that intradermal (i.d.) immunization with a recombinant adenovirus (Ad) expressing the murine melanoma antigen tyrosinase-related protein 2 (AdmTrp-2) results in a moderate level of tumor protection against the B16F10 murine melanoma without any vitiligo. Similar immunization with an Ad encoding human Trp-2 (AdhTrp-2) resulted in 50-fold greater protective immunity and produced vitiligo in all of the mice, suggesting that the development of autoimmunity may reflect the potency of the vaccine. Interestingly, delivery of AdhTrp-2 by i.m. injection generated protective immunity comparable with that seen in mice that received the vaccine by the i.d. route, but none of the recipients in the i.m. group developed vitiligo. The cellular and humoral responses in the i.m. immunized mice were greater than in the i.d. group; therefore, the lack of vitiligo was not caused by reduced efficacy of the vaccine. These results led us to hypothesize that vaccine-induced vitiligo was associated with local inflammatory responses. Mice immunized i.m. with AdhTrp-2 did develop vitiligo when they subsequently were injected i.d. with either a control Ad vector or complete Freund's adjuvant, suggesting that vitiligo is initiated by some form of trauma within the skin. Our data demonstrated that autoimmune pathology is not an unavoidable outcome of effective cancer vaccines directed against self-tumor antigens.
Collapse
Affiliation(s)
- Cecilia Lane
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, Hamilton, Ontario, Canada L8N 3Z5
| | | | | | | | | | | |
Collapse
|
86
|
Vonderheide RH, June CH. A translational bridge to cancer immunotherapy: exploiting costimulation and target antigens for active and passive T cell immunotherapy. Immunol Res 2004; 27:341-56. [PMID: 12857980 DOI: 10.1385/ir:27:2-3:341] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Building on significant advances in basic tumor immunology over the past decade, current translational efforts to develop novel antitumor T cell therapeutics continue to accelerate. Both passive T cell immunotherapy (e.g., adoptive T cell transfusions) and active immunotherapy (e.g., vaccination) may eventually become part of the arsenal to treat cancer. Successful approaches will need to repair host immunoincompetence in T cell function, circumvent immunosuppressive factors of the tumor microenvironment, and optimize target antigens with regard to clinical applicability, autoimmunity, and risk of antigen mutation. Here, we characterize two model systems for the ex vivo activation and expansion of human T lymphocytes and describe the potential for providing broadly applicable antitumor specificity by targeting universal tumor antigens. Polyclonal CD4+ T lymphocytes can be activated and expanded using anti-CD3 and anti-CD28 antibodies presented on magnetic beads, and CD8+T lymphocytes can be successfully expanded using a novel genetically engineered cell-based technology that presents anti-CD3 and anti-CD28 along with the costimulatory molecule CD137 (4-1BBL). As the prototypical and best-described universal tumor antigen, the human telomerase reverse transcriptase hTERT is vastly overexpressed in human tumors but absent in most normal tissues. Cytotoxic T lymphocytes (CTL) recognize peptides derived from hTERT and kill hTERT-positive tumor cells of multiple histologies. Phase I trials translating these discoveries to novel active and passive T cell therapies have been initiated, with an eye toward combining these strategies once safety is established.
Collapse
Affiliation(s)
- Robert H Vonderheide
- Abramson Family Cancer Research Institute, University of Pennsylvania Cancer Center and the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
87
|
Hahn WC. Targeting cancer with telomerase: commentary re Q. Huang et al., a novel conditionally replicative adenovirus vector targeting telomerase-positive tumor cells. Clin. Cancer Res., 10: 1439-1445, 2004. Clin Cancer Res 2004; 10:1203-5. [PMID: 14977815 DOI: 10.1158/1078-0432.ccr-04-0060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
88
|
Adotévi O, Pons FG, Langlade-Demoyen P. La télomérase : un antigène tumoral universel pour l’immunothérapie anti-cancéreuse. Med Sci (Paris) 2004; 20:19-21. [PMID: 14770360 DOI: 10.1051/medsci/200420119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
89
|
Affiliation(s)
- Freda K Stevenson
- Molecular Immunology Group, Tenovus Laboratory, Cancer Sciences Division Southampton University Hospitals Trust, Southampton SO16 6YD, United Kingdom
| | | | | |
Collapse
|
90
|
Abstract
Gastric cancer is common in China, and its early diagnosis and treatment are difficult. In recent years great progress has been achieved in gene therapy, and a wide array of gene therapy systems for gastric cancer has been investigated. The present article deals with the general principles of gene therapy and then focuses on how these principles may be applied to gastric cancer.
Collapse
Affiliation(s)
- Chao Zhang
- Department of General Surgery, Southwest Hospital, Third Military Medical University, Gaotan Yan, Chongqing 400038, China.
| | | |
Collapse
|
91
|
Abstract
Recent advances in basic medical sciences have led to a deeper understanding of the molecular characteristics of soft-tissue sarcomas. Likewise, novel technologies have led to a better appreciation of the relationship between an antigenic stimulus and the subsequent immune response against the antigen. In the past few years, the intersection of the understanding of the immune system and the knowledge of sarcoma biology has become apparent. As seen with other forms of cancer, there is a detectable autologous immune response against sarcomas. It is the hope of many investigators that the hints of a tumor-specific immune response will be enough to generate a signal that can be amplified and directed against the host sarcoma. The data regarding the initial evidence of immune responses against sarcomas are reviewed in the context of current or potential clinical studies.
Collapse
Affiliation(s)
- Robert G Maki
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 223, New York, NY 10021-6007, USA.
| |
Collapse
|
92
|
Knutson KL, Salazar L, Schiffman K, Disis ML. Immunotherapy of glioblastoma multiforme. Expert Rev Neurother 2003; 3:511-23. [PMID: 19810935 DOI: 10.1586/14737175.3.4.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glioblastoma multiforme is immunogenic and several glioblastoma multiforme-related antigens have now been identified. In addition, the immunologic characteristics of the tumor microenvironment that may affect tumor growth are becoming increasingly understood. The type of immune-based approach selected to treat glioblastoma multiforme will depend on the tumor burden. For minimal disease states, active vaccination may be useful for generating adequate protection from relapse. However, for more advanced stage disease states, more rigorous strategies may need to be applied, such as adoptive T-cell therapy, antibody therapy or a combination of different techniques. The immunosuppressive environment observed during advanced malignancy may need to be reversed for improved efficacy of immune-based therapies.
Collapse
Affiliation(s)
- Keith L Knutson
- Tumor Vaccine Group, 1959 NE Pacific St, Box 356527, HSB BB1321, University of Washington, Seattle, WA 98195 USA.
| | | | | | | |
Collapse
|
93
|
Abstract
Prostate cancer is the second leading cause of cancer death in the US, largely because of the limitations of our current therapeutic options, especially once the cancer has metastasized. Investigators have long sought new therapeutic modalities such as angiogenesis inhibitors, vaccines, and gene therapy, among others. It appears that a combination approach will be required to cure the majority of malignancies. Immunotherapy for prostate cancer appears feasible and a likely therapeutic modality in the armamentarium. Unfortunately, further research in basic immunology and the interaction of the immune system with other forms of therapy is needed. Many obstacles exist in immunotherapy, including vector design, tumouricidal specificity, and tumor evasion, which will have to be overcome in order to realize the maximum therapeutic benefit from this treatment modality.
Collapse
Affiliation(s)
- Joseph M Kaminski
- Department of Radiology, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|
94
|
Abstract
A number of different approaches have been developed to inhibit telomerase activity in human cancer cells. Different components and types of inhibitors targeting various regulatory levels have been regarded as useful for telomerase inhibition. Most methods, however, rely on successive telomere shortening. This process is very slow and causes a long time lag between the onset of inhibition and the occurrence of senescence or apoptosis as a reversal of the immortal phenotype. Many telomerase inhibitors seem to be most efficient when combined with conventional chemotherapeutics. There are some promising approaches that seem to circumvent the slow way of telomere shortening and induce fast apoptosis in treated tumor cells. It has been demonstrated that telomerase may be involved in triggering apoptosis, but the underlying molecular mechanism remains unclear.
Collapse
|
95
|
Abstract
Specialized nucleoprotein structures, termed telomeres, cap the ends of human chromosomes. These terminal structures, composed of repetitive arrays of guanine-rich hexameric DNA together with specific telomere-binding proteins, play essential roles in protecting the chromosome from damage and degradation. In addition, several lines of evidence implicate telomere maintenance as an important regulator of cell life span. Activation of telomerase, a dedicated reverse transcriptase that synthesizes telomeric sequences, is strongly associated with cancer, and recent observations confirm that telomeres and telomerase perform important roles in both suppressing and facilitating malignant transformation. These dual functions of telomere biology are evident in the clinical manifestations of the multisystem syndrome, dyskeratosis congenita, forms of which display defects in telomerase function. Recent advances in our understanding of telomere biology indicate that the manipulation of telomeres and telomerase will lead to clinically significant applications in the diagnosis, prevention, and treatment of neoplastic disease.
Collapse
Affiliation(s)
- William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
96
|
Abstract
The unique biology of telomeres and telomerase plays important roles in many aspects of mammalian cell physiology. Over the past decade, several lines of evidence have confirmed that the maintenance of telomeres and telomerase participate actively in the pathogenesis of human cancer. Specifically, activation of telomerase is strongly associated with cancer, and recent observations confirm that telomeres and telomerase perform important roles in both suppressing and facilitating malignant transformation by regulating genomic stability and cell lifespan. In addition, recent evidence suggests that telomerase activation contributes to tumorigenesis independently of its role in maintaining telomere length. Here we review recent developments in our understanding of the relationships among telomeres, telomerase, and cancer.
Collapse
Affiliation(s)
- Kenkichi Masutomi
- Department of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney Street, Dana 710C, Boston, MA 02115, USA
| | | |
Collapse
|
97
|
Cunto-Amesty G, Monzavi-Karbassi B, Luo P, Jousheghany F, Kieber-Emmons T. Strategies in cancer vaccines development. Int J Parasitol 2003; 33:597-613. [PMID: 12782058 DOI: 10.1016/s0020-7519(03)00054-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The recent definition of tumour-specific immunity in cancer patients and the identification of tumour-associated antigens have generated renewed enthusiasm for the application of immune-based therapies for the treatment of malignancies. Recent developments in cancer vaccines have also been based on an improved understanding of the cellular interactions required to induce a specific anti-tumour immune response. Consequently, a number of cancer vaccines have entered clinical trials. Targeting broad-spectrum tumour-associated antigens has emerged as a strategy to lower the risk of tumour escape due to the loss of specific nominal antigen. Amongst the most challenging of tumour-associated antigens to which to target in active specific immunotherapy applications are carbohydrate antigens. As carbohydrates are intrinsically T-cell-independent antigens, more novel approaches are perhaps needed to drive specific-T-cell-dependent immune responses to carbohydrate antigens. In this context peptide mimetics of core structures of tumour-associated carbohydrate antigens might be developed to augment immune responses to these broad-spectrum antigens.
Collapse
Affiliation(s)
- Gina Cunto-Amesty
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
98
|
Abstract
BACKGROUND Clinically successful Ag-specific cancer immunotherapy depends on the identification of tumor-rejection Ags. Historically, tumor Ags have been identified by analyzing cancer patients' own T-cell or Ab responses. METHODS The unveiling of the human genome and optimized immunological analytical tools, particularly 'reverse immunology', have made it possible to screen any given protein for immunogenic epitopes. These advances enable the immunological characterization of universal tumor-associated gene products that mediate critical functions for tumor growth and development. RESULTS Four examples of candidate universal tumor Ags reviewed here include the telomerase reverse transcriptase (hTERT), the inhibitor of apoptosis survivin, the p53-interacting protein MDM2, and the cytochrome P450 isoform 1B1--each at various levels of preclinical and clinical development. DISCUSSION The cardinal feature of universal TAA is that they are expressed in (nearly) all tumors and in no normal tissues. They are directly involved in the malignant phenotype of the tumor. Certain peptides derived from such Ags are expressed on the tumor-cell surface, as evidenced by Ag-specific, MHC-restricted T-cell anti-tumor reactivity in vitro. It is hoped that these features imply a pre-existing, high-affinity T-cell pool that can be activated in vivo in patients, without immunoselection of variant tumor cells no longer expressing the Ag of choice.
Collapse
Affiliation(s)
- J D Gordan
- Abramson Family Cancer Research Institute, University of Pennsylvania Cancer Center and Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
99
|
Qu Y, Wang Z, Huang X, Wan C, Yang CL, Liu B, Cornelissen G, Halberg F. Circadian telomerase activity and DNA synthesis for timing peptide administration. Peptides 2003; 24:363-9. [PMID: 12732333 DOI: 10.1016/s0196-9781(03)00050-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
DNA synthesis and telomerase activity were assessed in nude mice transplanted with hepatic carcinoma. Hepatic cancer cells (SMMC-7721) were implanted into both flanks of each of 14 BALB/C mice synchronized in 12 h of light alternating with 12 h of darkness (LD12:12) for 4 weeks. At 7 timepoints, tumor samples were collected for measurement of cellular DNA content by flow cytometry and telomerase activity by PCR-ELISA assay. Cosinor analyses determine a 24-h rhythm for all variables, showing a similar timing for the DNA-synthesis phase and telomerase activity. These results provide a model for exploring optimal timing of chronotherapy with peptides, especially for treatment with telomerase inhibitors.
Collapse
Affiliation(s)
- Yi Qu
- Medical Center, Sichuan University, Sichuan, PR China
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
The continuous growth of advanced malignancies almost universally correlates with the reactivation of telomerase. While there is still a great deal of basic and applied research to be done, telomerase remains a very attractive novel target for cancer therapeutics. In this review, we will discuss the challenges and the pros and cons of the most promising antitelomerase approaches currently being investigated.
Collapse
Affiliation(s)
- Jerry W Shay
- The University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75309, USA.
| | | |
Collapse
|