Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Perspective

Universal and Stemness-Related Tumor Antigens: Potential Use in Cancer Immunotherapy

Giorgio Parmiani, Vincenzo Russo, Andrea Marrari, Gianluca Cutolo, Chiara Casati, Lorenzo Pilla, Cristina Maccalli, Licia Rivoltini and Chiara Castelli
Giorgio Parmiani
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Vincenzo Russo
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Andrea Marrari
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Gianluca Cutolo
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Chiara Casati
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lorenzo Pilla
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Cristina Maccalli
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Licia Rivoltini
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Chiara Castelli
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1078-0432.CCR-07-0879 Published October 2007
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading
  • Universal cancer antigens
  • immunotherapy
  • cancer stem cells
  • Tumor Immunobiology
  • Cancer vaccines
  • Stem Cell Biology

After a decade of cancer immunotherapy (both active and adoptive), based on the use of well-defined tumor-associated antigens (TAA), and despite the large amount of new information that has been collected both in preclinical and clinical settings, the clinical outcome of immunotherapy trials has been altogether disappointing (1, 2). Several reasons have been put forward to explain such phenomenon like tumor escape from and alterations of the immune response caused by the presence of growing tumor cells (3).

Less attention has been paid, however, to the nature of TAAs to be used in vaccine formulation. In fact, practically all the trials of immunotherapy of cancer (by and large of vaccination) conducted during the last 10 years were based on the use of TAAs encompassing normal proteins or peptides, to generate antitumor T cells mostly restricted by class I HLA (4). These TAAs are now known to be weakly immunogenic owing to different forms of tolerance to them displayed by the patients' immune system. Moreover, several of these self-TAAs have been shown to be heterogeneously expressed within the tumor mass (5) and selected against by CTLs during tumor growth allowing an immunologically unrestricted progression of the neoplasia (6, 7). Thus, the search for new, tumor-specific and immunogenic TAAs constitutively linked to the neoplastic state and, therefore, unselectable by the patient immune reactions, remains an important issue in tumor immunotherapy.

Truly tumor-specific TAAs are those expressed only by tumor cells and not by whatsoever normal cells. Such TAAs may derive (a) from somatic gene mutations of tumor cells resulting in new TAA epitopes recognized by the host T lymphocytes, like the unique TAAs (8, 9), or less frequently, (b) from alterations in splicing (10–12). Unique TAAs are now being frequently found in several human tumors, although their use in clinical trials is difficult for the time required to characterize unique TAAs at a single patient level (8, 9, 13).

Universal and Cancer Stemness-Related TAAs as Potentially More Effective Targets in Immunotherapeutic Interventions

Recently, another group of TAAs was described that include antigens overexpressed by neoplastic and fetal cells, and was weakly expressed in a phase-specific way in a few normal cells (14, 15). Such TAAs have the remarkable feature of being indispensable for tumor growth and progression. Immune responses targeting these proteins could thus result in a limited or no generation of antigen-loss variants and, therefore, they may efficiently control in vivo tumor growth, thus meeting the requirements for an ideal TAA set forth above. These are the so-called universal TAAs and include molecules like human telomerase reverse transcriptase (hTERT) and the inhibitor of apoptosis proteins (IAP).

An additional group of TAAs, practically unknown at the moment, includes those that might be expressed by cancer stem cells (CSC), a minor population of tumor cells which, however, shows the features of stemness (see ref. 16) and, therefore, may represent the most important target for eradicating tumor lesions even in case of immunotherapy. It is, therefore, mandatory to identify TAAs or other cell surface molecules (e.g., natural killer cell–activating receptors) of CSCs in order to assess whether they can represent a new target for the host immune system. Our prediction is that at least some of the CSC antigens will match proteins already known to serve as TAAs and expressed by embryo-fetal tissues (e.g., survivin, telomerase, cancer/testis antigens) reflecting common functions between embryonic stem cells and CSCs, i.e., multipotent differentiation, plasticity, and self-renewal. However, because these TAAs are expressed even by the majority of cancer cells of a given tumor, while CSCs encompass a small subpopulation of the tumor mass, CSCc may represent a better target if specifically enriched in cancer/testis, IAP-derived and/or mutated (unique) antigens in parallel with stem cell markers (e.g., CD44, CD133), in comparison with other tumor cells showing a more disperse concentration of such TAAs (17). Therefore, immune-mediated tumor regression should reflect a qualitative rather than a quantitative targeting aimed at eliminating a specific, minor subpopulation of tumor cells, i.e., CSC, rather than the majority of neoplastic cells. If so, this will represent a new paradigm in the immunotherapy of human tumors.

The Universal and CSC-Derived TAAs: Tissue Distribution and Immune Functions

The universal TAAs have been well-defined because of their wide distribution in human tumors of different histology, and due to the general and fundamental functions they exert in tumor biology, i.e., that of maintaining an effective proliferation and resistance to apoptosis, thus contributing to cancer cell progression and survival (Fig. 1 ). CSCs should also express universal TAAs together with most cells of the tumor mass, as suggested by immunohistochemical analysis, which reveals the presence of hTERT and IAPs (e.g., survivin) in the large majority of tumor cells (15, 18). However, whether CSCs are endowed with the ability to mount IAP-derived epitopes on their HLA is being tested in ongoing studies. Preliminary findings suggest that CSCs show a variable, but in general, low expression of HLA on their surface.3 Moreover, if the CSC target molecules are also present in normal stem cells, potential damage to normal tissues may occur. Therefore, CSC-selective elimination by immune responses should be based either on new CSC-specific targets resulting from somatic alterations/mutations of CSC genes that do not occur in normal stem cells, or in an enrichment of molecules, like the universal TAAs, that in normal stem cells cannot represent a valuable target whereas mediating a T cell cytotoxicity on neoplastic counterparts (14, 15).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Functions of survivin, livin, and telomerase in cancer cells. Survivin and livin are IAPs that interfere with the apoptotic process by blocking upstream caspase-9 activity and/or the final downstream effector step of caspase-3 and caspase-7 as well as by binding the mitochondrial protein Smac (left). Survivin can also favor tumor cell mitosis by localizing mostly to centrosomes and microtubules whose regular assembly is also dependent on the presence of this IAP (middle). Telomerase is a RNA-dependent polymerase that allows the maintenance of chromosome length and stability in tumor cells, which in turn can divide indefinitely. It includes repeated DNA sequences (TTAGGG) and associated proteins. RNA antisense and other inhibitors can block telomerase activity and allows only a limited number or divisions to tumor cells (right).

Universal TAAs have recently been assessed in preclinical in vitro studies for their ability to generate T cell–specific immune responses (see ref. 19). Repeated stimulations of peripheral blood lymphocytes from both healthy donors and cancer patients with APCs pulsed with HLA-I restricted epitopes, entire proteins, or mRNAs encoding the antigens has made it possible to detect CD8+ and CD4+ T cell spontaneous and/or induced responses against survivin, livin, hTERT, and Bcl-2 (19–25). Importantly, these effectors recognize HLA-matched tumor cells, which endogenously process the antigens, but not normal cells (e.g., CD34+ hematopoietic precursors, activated T and B cells; ref. 18). Moreover, ex vivo studies have shown a significant frequency of functional CTLs directed against hTERT among peripheral blood lymphocytes of a proportion of cancer patients as well as the presence of survivin-specific CTLs in tumor-invaded lymph nodes (26, 27). All these features make universal TAAs suitable targets for cancer immunotherapy, although a screening of single patients to analyze their capacity to mount an anti-hTERT immune response seems advisable (26).

By applying the reverse immunology approach, several peptides binding different HLA alleles have been recently identified in survivin, livin, and hTERT proteins. Some of these peptides have been shown to function as T cell epitopes being able to induce peptide-specific T cells in patients' peripheral blood mononuclear cells (see ref. 19). However, an additional crucial issue that needs to be addressed in order to consider a defined peptide as useful for a therapeutic approach is its ability to elicit T cells that can recognize the naturally processed peptide on tumor cells expressing the target protein and the appropriate HLA molecule. Although this requirement has been met for a majority of the identified peptides, for some, doubts on the efficiency of their presentation by tumor cells still remain (28, 29). An example of such a phenomenon has been reported for the hTERT peptide 540-548, which although being processed by human cells when transfected with hTERT encoding cDNA, was only marginally produced by tumor-derived proteasomes (29). In view of these observations, the functional properties of T cells elicited by immunogenic peptides should always be analyzed on a large panel of tumor cells of different histologies.

Clinical Trials with Universal TAAs

Although the central role of universal TAAs in malignant transformation and cancer progression and their immunogenicity have been established, only a limited number of vaccination protocols targeting IAPs have been and are being carried out (Table 1 ). The following is a brief summary of such clinical trials.

View this table:
  • View inline
  • View popup
Table 1.

Clinical trials involving the use of universal TAAs

In a pilot study, patients with chemotherapy-resistant metastatic breast cancer or hormone-resistant prostate cancer received ex vivo–generated autologous dendritic cells (DC) pulsed with the HLA-A*0201–restricted hTERT I540 peptide together with keyhole limpet hemocyanin (28). Among six of the seven evaluable patients, a mixed clinical response was seen. In these patients, prevaccination and postvaccination immunohistochemical analyses of skin nodules showed a lymphoid infiltrate predominantly made of CD8+ T cells, but no information on the function of these tumor-infiltrating lymphocytes was reported. A specific T cell response against hTERT was detected in the uncultured peripheral blood mononuclear cells of three patients. After 1 week of in vitro peptide sensitization, CD8+ tetramer+ T cells were seen in four patients. These CTL populations contained clones that were able to recognize and lyse hTERT expressing HLA-A*0201 cancer cells.

A peptide-based approach of immunotherapy was carried out by Parkhurst and coworkers using the same epitope emulsified in Montanide ISA51 but achieving opposite results (29). None of the 14 patients with metastatic melanoma developed a CTL response recognizing endogenously processed telomerase target cell, even though high-avidity T cell clones were obtained. Furthermore, no clinical benefits were observed in these patients.

Other hTERT-derived class I and class II epitopes given together with different adjuvants have been tested in patients with progressive and chemotherapy-unresponsive tumors. Both native and modified peptides have proved to be safe, showing their ability to induce a specific immune response without causing any sign of hematological toxicity on the blood stem cell compartment. For example, Gaudernack and coworkers treated lung and pancreatic cancer patients with HR2822 (540-548) and GV1001 (611-626) hTERT-derived epitopes (30, 31). The latter is a promiscuous HLA class II epitope that has been shown to induce a strong CD4+ T cell response able to generate and sustain a CTL response. The immunologic responder patients showed a tendency for a prolonged survival as compared with historical controls. This seems to be important considering the aggressiveness and the immune-suppressive activity that characterizes these tumors.

In the effort to overcome the problems of altered antigen processing, hTERT mRNA was used to transfect metastatic prostate cancer–derived DCs (22). In a subgroup of patients, a chimeric transcript made of the lysosomal-associated membrane antigen-1 was fused to the target antigen, thus improving the generation of CD4+ T response. Although no clinical benefits were achieved in either group, transient and short-lasting effects on prostate-specific antigen doubling times were obtained in some patients.

In the attempt to develop a polyvalent vaccine targeting both patient-specific tumor and stromal targets, total RNA derived from renal tumor tissue was used to transfect patients' autologous DCs (32). Although no assessment of the therapeutic effect could be made, a strong immunologic response against hTERT was elicited.

Clinical experience on the use of survivin in tumor vaccination trials is limited to few studies (19), despite the development of a spontaneous immune response to such an antigen, which seems to be quite common in patients with cancer. A modified HLA-A*0201 epitope of survivin (96-104, 2M) has been used to vaccinate stage IV melanoma patients with DC as adjuvant (33). Four out of five patients mounted a specific antisurvivin immune response detected both in the peripheral blood and in metastatic lesions showing the ability of the vaccine-induced CD8+ T cell population to home into peripheral tumor tissues. At the same time, these patients experienced an unexpected long survival.

In a phase I trial, eight patients with hormone-refractory prostate cancer received autologous DCs pulsed with HLA-A*0201–restricted epitopes derived from prostate cancer–associated antigens and survivin (95-104; ref. 34). In this trial, the strongest reduction of prostate-specific antigen plasma levels was observed in the four patients (one with partial remission and three with stable disease) developing specific CD8+ T cell–mediated immune response, only two of whom, however, directed against the survivin epitope.

In our centers (Istituto Nazionale Tumori, San Raffaele Scientific Institute), clinical trials involving vaccination with peptides derived from survivin are presently ongoing. These trials are based on the administration of multiple HLA-A*0201–restricted peptides derived from differentiation antigens and cyclophosphamide aimed at reducing the in vivo effect of regulatory T cells. This approach is under evaluation in early stage melanoma, prostate cancer patients with biochemical failure after conventional treatments (receiving PSMA and survivin-derived peptides), and in locally recurrent rectal carcinoma (with carcinoembryonic antigen and survivin-derived epitopes). Preliminary immunologic analysis shows that a significant boost of T cells specific for all the administered peptides, including survivin, could be detected in both PBLs and in draining lymph nodes.4

Altogether, these early clinical trials have enrolled a limited number of patients and are too heterogeneous in terms of vaccine formulation and tumor targets to allow any meaningful conclusion on the clinical efficacy of immunizations based on universal TAAs. Further results of ongoing phase II studies with a larger number of patients are eagerly awaited in order to assess the importance of this approach for cancer therapy.

Perspectives

From the studies on universal TAAs, we have learned that such antigens are, more than any other TAAs, frequently expressed in many different human tumors and, therefore, they apparently provide the most common, neoplasia-related and easily identifiable immune target. Upon closer examination, however, it remains to be established whether hTERT is more immunogenic for T cells of cancer patients as compared with other TAAs obtained from the same tumor as suggested by Su and coworkers in renal cell carcinoma patients vaccinated with tumor RNA pulsed DCs (22). In fact, a different study found that the frequency of HLA-A2–restricted T cell precursors against hTERT peptide was similar in cancer patients and in normal donors (35).

As for survivin, cancer patients were shown to have spontaneous T cell responses against survivin peptides (27) and we have also shown that anti-survivin T cell precursors are detectable in cancer patients but not in normal individuals (24); importantly, such T cells were found to recognize tumor cells expressing survivin. These findings suggest that survivin may be strongly immunogenic in at least a fraction of patients.

Whether these antigens might also actually represent a significant target for CSCs is unknown at the moment. However, the study of the immunogenic potential of universal TAAs is worth pursuing because it may pave the way for in vitro and in vivo characterization of similar, if not identical, TAAs expressed by CSCs; thus allowing their use as targets in future trials of cancer immunotherapy. The presence of CSCs in the tumor mass may change the whole paradigm of cancer immunotherapy with the need for preparing T and/or natural killer cell effectors and/or antibodies able to recognize and preferentially destroy the small CSC subpopulation, rather than a huge number of lymphocyte effectors aimed at targeting as many tumor cells as possible. Because the markers (antigens?) which have thus far been reported to characterize different types of CSC (e.g., ESA, CD44, CD133; refs. 16, 36–47) seem to be normal molecules involved in the process of cell adhesion and migration, it is likely that such molecules may be more easily targeted by antibodies than by antigen-specific T cells. In fact, an antibody against CD44 has recently been shown to eradicate xenografted human acute myelogenous leukemia stem cells, possibly by interfering with the CSC-supporting microenvironment (48). The anti-CSC function of T cells, however, cannot be completely discarded. In fact, one of CSC's most common markers, ESA (Ep-CAM; refs. 16, 39), has been shown to contain epitopes that can be recognized by class I HLA–restricted T cells (49), and which has been used to vaccinate colorectal cancer patients, although with limited success (50). In this context, vaccines containing T cell–defined universal and CSC TAAs and their combination with antibodies targeting the cell surface molecules expressed by CSCs (e.g., CD44) may be much more effective than those including self-TAAs previously used in cancer vaccination.

Footnotes

  • ↵3 Maccalli C, Galli R, Parmiani G, unpublished results.

  • ↵4 Rivoltini L, Castelli C, Parmiani G, unpublished results.

  • Grant support: Associazione Italiana Ricerche sul Cancro (Milan), the European Community FP5 (Brussels), the Italian Ministry of Health (Rome).

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    • Accepted July 2, 2007.
    • Received April 12, 2007.
    • Revision received June 15, 2007.

References

  1. ↵
    Parmiani G, Castelli C, Dalerba P, et al. Cancer immunotherapy with peptide-based vaccines: what have we achieved? Where are we going? J Natl Cancer Inst 2002;94:805–18.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med 2004;10:909–15.
    OpenUrlCrossRefPubMed
  3. ↵
    Rivoltini L, Canese P, Huber V, et al. Escape strategies and reasons for failure in the interaction between tumour cells and the immune system: how can we tilt the balance towards immune-mediated cancer control? Expert Opin Biol Ther 2005;5:463–76.
    OpenUrlCrossRefPubMed
  4. ↵
    Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother 2005;54:187–207.
    OpenUrlCrossRefPubMed
  5. ↵
    Anichini A, Fossati G, Parmiani G. Heterogeneity of clones from a human metastatic melanoma detected by autologous cytotoxic T lymphocyte clones. J Exp Med 1986;163:215–20.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Maeurer MJ, Gollin SM, Martin D, et al. Tumor escape from immune recognition: lethal recurrent melanoma in a patient associated with downregulation of the peptide transporter protein TAP-1 and loss of expression of the immunodominant MART-1/Melan-A antigen. J Clin Invest 1996;98:1633–41.
    OpenUrlCrossRefPubMed
  7. ↵
    Boon T, Coulie PG, Van den Eynde BJ, van der Bruggen P. Human T cell responses against melanoma. Annu Rev Immunol 2006;24:175–208.
    OpenUrlCrossRefPubMed
  8. ↵
    Sensi M, Anichini A. Unique tumor antigens: evidence for immune control of genome integrity and immunogenic targets for T cell-mediated patient-specific immunotherapy. Clin Cancer Res 2006;12:5023–32.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Parmiani G, De Filippo A, Novellino L, Castelli C. Unique human tumor antigens: immunobiology and use in clinical trials. J Immunol 2007;178:1975–9.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Guilloux Y, Lucas S, Brichard VG, et al. A peptide recognized by human cytolytic T lymphocytes on HLA-A2 melanomas is encoded by an intron sequence of the N-acetylglucosaminyltransferase V gene. J Exp Med 1996;183:1173–83.
    OpenUrlAbstract/FREE Full Text
  11. Robbins PF, El-Gamil M, Li YF, Fitzgerald EB, Kawakami Y, Rosenberg SA. The intronic region of an incompletely spliced gp100 gene transcript encodes an epitope recognized by melanoma-reactive tumor-infiltrating lymphocytes. J Immunol 1997;159:303–8.
    OpenUrlAbstract
  12. ↵
    Lupetti R, Pisarra P, Verrecchia A, et al. Translation of a retained intron in tyrosinase-related protein (TRP) 2 mRNA generates a new cytotoxic T lymphocyte (CTL)-defined and shared human melanoma antigen not expressed in normal cells of the melanocytic lineage. J Exp Med 1998;188:1005–16.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Lennerz V, Fatho M, Gentilini C, et al. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc Natl Acad Sci U S A 2005;102:16013–8.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Adida C, Crotty PL, McGrath J, Berrebi D, Diebold J, Altieri DC. Developmentally regulated expression of the novel cancer anti-apoptosis gene survivin in human and mouse differentiation. Am J Pathol 1998;152:43–9.
    OpenUrlPubMed
  15. ↵
    Altieri DC, Marchisio PC. Survivin apoptosis: an interloper between cell death and cell proliferation in cancer. Lab Invest 1999;79:1327–33.
    OpenUrlPubMed
  16. ↵
    Dalerba P, Cho RW, Clarke MF. Cancer stem cells: models and concepts. Annu Rev Med 2007;58:267–84.
    OpenUrlCrossRefPubMed
  17. ↵
    Gedye C, Quirk J, Browning J, et al. Melanoma stem-like cells can be eliminated by targeting cancer/testis antigens. Keystone Symposia: Stem Cells and Cancer 2007;147:150.
    OpenUrl
  18. ↵
    Vonderheide RH, Hahn WC, Schultze JL, Nadler LM. The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity 1999;10:673–9.
    OpenUrlCrossRefPubMed
  19. ↵
    Andersen MH, Becker JC, Straten P. Regulators of apoptosis: suitable targets for immune therapy of cancer. Nat Rev Drug Discov 2005;4:399–409.
    OpenUrlCrossRefPubMed
  20. Minev B, Hipp J, Firat H, Schmidt JD, Langlade-Demoyen P, Zanetti M. Cytotoxic T cell immunity against telomerase reverse transcriptase in humans. Proc Natl Acad Sci U S A 2000;97:4796–801.
    OpenUrlAbstract/FREE Full Text
  21. Schmitz M, Diestelkoetter P, Weigle B, et al. Generation of survivin-specific CD8+ T effector cells by dendritic cells pulsed with protein or selected peptides. Cancer Res 2000;60:4845–9.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Su Z, Vieweg J, Weizer AZ, et al. Enhanced induction of telomerase-specific CD4(+) T cells using dendritic cells transfected with RNA encoding a chimeric gene product. Cancer Res 2002;62:5041–8.
    OpenUrlAbstract/FREE Full Text
  23. Schroers R, Huang XF, Hammer J, Zhang J, Chen SY. Identification of HLA DR7-restricted epitopes from human telomerase reverse transcriptase recognized by CD4+ T-helper cells. Cancer Res 2002;62:2600–5.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Casati C, Dalerba P, Rivoltini L, et al. The apoptosis inhibitor protein survivin induces tumor-specific CD8+ and CD4+ T cells in colorectal cancer patients. Cancer Res 2003;63:4507–15.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Schmidt SM, Schag K, Muller MR, et al. Survivin is a shared tumor-associated antigen expressed in a broad variety of malignancies and recognized by specific cytotoxic T cells. Blood 2003;102:571–6.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Filaci G, Fravega M, Setti M, et al. Frequency of telomerase-specific CD8+ T lymphocytes in patients with cancer. Blood 2006;107:1505–12.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Andersen MH, Pedersen LO, Capeller B, Brocker EB, Becker JC, thor Straten P. Spontaneous cytotoxic T-cell responses against survivin-derived MHC class I-restricted T-cell epitopes in situ as well as ex vivo in cancer patients. Cancer Res 2001;61:5964–8.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Vonderheide RH, Domchek SM, Schultze JL, et al. Vaccination of cancer patients against telomerase induces functional antitumor CD8+ T lymphocytes. Clin Cancer Res 2004;10:828–39.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Parkhurst MR, Riley JP, Igarashi T, Li Y, Robbins PF, Rosenberg SA. Immunization of patients with the hTERT:540–548 peptide induces peptide-reactive T lymphocytes that do not recognize tumors endogenously expressing telomerase. Clin Cancer Res 2004;10:4688–98.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Brunsvig PF, Aamdal S, Gjertsen MK, et al. Telomerase peptide vaccination: a phase I/II study in patients with non-small cell lung cancer. Cancer Immunol Immunother 2006;55:1553–64.
    OpenUrlCrossRefPubMed
  31. ↵
    Bernhardt SL, Gjertsen MK, Trachsel S, et al. Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: a dose escalating phase I/II study. Br J Cancer 2006;95:1474–82.
    OpenUrlCrossRefPubMed
  32. ↵
    Su Z, Dannull J, Yang BK, et al. Telomerase mRNA-transfected dendritic cells stimulate antigen-specific CD8+ and CD4+ T cell responses in patients with metastatic prostate cancer. J Immunol 2005;174:3798–807.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Otto K, Andersen MH, Eggert A, et al. Lack of toxicity of therapy-induced T cell responses against the universal tumour antigen survivin. Vaccine 2005;23:884–9.
    OpenUrlCrossRefPubMed
  34. ↵
    Fuessel S, Meye A, Schmitz M, et al. Vaccination of hormone-refractory prostate cancer patients with peptide cocktail-loaded dendritic cells: results of a phase I clinical trial. Prostate 2006;66:811–21.
    OpenUrlCrossRefPubMed
  35. ↵
    Vonderheide RH, Schultze JL, Anderson KS, et al. Equivalent induction of telomerase-specific cytotoxic T lymphocytes from tumor-bearing patients and healthy individuals. Cancer Res 2001;61:8366–70.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994;367:645–8.
    OpenUrlCrossRefPubMed
  37. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003;100:3983–8.
    OpenUrlAbstract/FREE Full Text
  38. Ponti D, Costa A, Zaffaroni N, et al. Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res 2005;65:5506–11.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Dalerba P, Dylla SJ, Park IK, et al. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci U S A 2007;104:10158–63.
    OpenUrlAbstract/FREE Full Text
  40. Fang D, Nguyen TK, Leishear K, et al. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res 2005;65:9328–37.
    OpenUrlAbstract/FREE Full Text
  41. Galli R, Binda E, Orfanelli U, et al. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res 2004;64:7011–21.
    OpenUrlAbstract/FREE Full Text
  42. Patrawala L, Calhoun T, Schneider-Broussard R, et al. Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene 2006;25:1696–708.
    OpenUrlCrossRefPubMed
  43. Szotek PP, Pieretti-Vanmarcke R, Masiakos PT, et al. Ovarian cancer side population defines cells with stem cell-like characteristics and Mullerian inhibiting substance responsiveness. Proc Natl Acad Sci U S A 2006;103:11154–9.
    OpenUrlAbstract/FREE Full Text
  44. Li C, Heidt DG, Dalerba P, et al. Identification of pancreatic cancer stem cells. Cancer Res 2007;67:1030–7.
    OpenUrlAbstract/FREE Full Text
  45. O'Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007;445:106–10.
    OpenUrlCrossRefPubMed
  46. Ricci-Vitiani L, Lombardi DG, Pilozzi E, et al. Identification and expansion of human colon-cancer-initiating cells. Nature 2007;445:111–5.
    OpenUrlCrossRefPubMed
  47. ↵
    Prince ME, Sivanandan R, Kaczorowski A, et al. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci U S A 2007;104:973–8.
    OpenUrlAbstract/FREE Full Text
  48. ↵
    Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med 2006;12:1167–74.
    OpenUrlCrossRefPubMed
  49. ↵
    Nagorsen D, Keilholz U, Rivoltini L, et al. Natural T-cell response against MHC class I epitopes of epithelial cell adhesion molecule, her-2/neu, and carcinoembryonic antigen in patients with colorectal cancer. Cancer Res 2000;60:4850–4.
    OpenUrlAbstract/FREE Full Text
  50. ↵
    Neidhart J, Allen KO, Barlow DL, et al. Immunization of colorectal cancer patients with recombinant baculovirus-derived KSA (Ep-CAM) formulated with monophosphoryl lipid A in liposomal emulsion, with and without granulocyte-macrophage colony-stimulating factor. Vaccine 2004;22:773–80.
    OpenUrlPubMed
PreviousNext
Back to top
Clinical Cancer Research: 13 (19)
October 2007
Volume 13, Issue 19
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Clinical Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Universal and Stemness-Related Tumor Antigens: Potential Use in Cancer Immunotherapy
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Universal and Stemness-Related Tumor Antigens: Potential Use in Cancer Immunotherapy
Giorgio Parmiani, Vincenzo Russo, Andrea Marrari, Gianluca Cutolo, Chiara Casati, Lorenzo Pilla, Cristina Maccalli, Licia Rivoltini and Chiara Castelli
Clin Cancer Res October 1 2007 (13) (19) 5675-5679; DOI: 10.1158/1078-0432.CCR-07-0879

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Universal and Stemness-Related Tumor Antigens: Potential Use in Cancer Immunotherapy
Giorgio Parmiani, Vincenzo Russo, Andrea Marrari, Gianluca Cutolo, Chiara Casati, Lorenzo Pilla, Cristina Maccalli, Licia Rivoltini and Chiara Castelli
Clin Cancer Res October 1 2007 (13) (19) 5675-5679; DOI: 10.1158/1078-0432.CCR-07-0879
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Universal and Cancer Stemness-Related TAAs as Potentially More Effective Targets in Immunotherapeutic Interventions
    • The Universal and CSC-Derived TAAs: Tissue Distribution and Immune Functions
    • Clinical Trials with Universal TAAs
    • Perspectives
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • ICML Workshop on the Design of Clinical Trials in Lymphomas
  • Cancer Lessons Learned from HIV
  • Pathogenesis of Renal Medullary Carcinoma
Show more Perspective
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement