The Food and Drug Administration (FDA) approval of sipuleucel-T1 as the first therapeutic cancer vaccine represents a major stride for this field. Sipuleucel-T consists of autologous peripheral blood mononuclear cells, including antigen-presenting cells that have been activated ex vivo with a recombinant fusion protein. In addition to this vaccine, numerous other vaccine platforms are currently demonstrating evidence of patient benefit in multicenter, randomized Phase II and Phase III studies in a range of human cancers.
This article will review several of these recent findings as well as describe those areas of research that will most impact on improving the ultimate use of cancer vaccines either as monotherapy or in combination therapy. Also described will be the hurdles that must be overcome in dealing with immune regulatory and inhibitory entities and the lessons learned for appropriate clinical trial design.
While the vast majority of clinical studies that involve therapeutic cancer vaccines have been performed in patients with metastatic melanoma, it is interesting to note that the first FDA-approved vaccine has shown benefit in patients with metastatic prostate cancer. Several characteristics render prostate cancer an excellent disease for evaluation of a cancer vaccine. Prostate cancer is generally an indolent disease with a long interval from disease detection to metastasis. The serum marker prostate-specific antigen (PSA) can also be used to help identify patients' response to therapy.
The initial Phase III study of sipuleucel-T2 in patients with minimally symptomatic metastatic prostate cancer did not meet its primary end point of improved disease progression, but subsequently showed evidence of enhanced patient survival in the vaccine arm. A second Phase III trial1 was then conducted with overall survival as the end point, which again demonstrated an improvement in median overall survival of 25.8 months in the vaccine arm vs 21.7 months in the control arm (p=0.03). This vaccine platform involves three leukaphereses, each of which is shipped to a central facility where antigen-presenting cells are incubated with a prostate antigen (PAP)/granulocyte macrophage colony-stimulating factor (GM-CSF) fusion protein; the “vaccine” is then shipped back for infusion into patients.
A second prostate cancer vaccine has also shown promising results in the same patient population. This “off-the-shelf” vaccine platform (PROSTVAC) consists of recombinant viral vectors that contain transgenes for the prostate antigen PSA and three immune-stimulating molecules (designated TRICOM). A multicenter, placebo-controlled Phase II trial3 demonstrated an improved median overall survival of 25.1 months in the vaccine arm vs 16.6 months in the control arm (p=0.006). A Phase III study has been initiated with the PROSTVAC vaccine.
An important point to consider with both of the vaccines described above, as well as with other vaccines to be described below, is the extremely low level of toxicity. In addition to the quality of life consideration, this also renders vaccines amenable to combination therapies without the issue of compounding toxicities.
Successful Phase III studies have also recently been reported with two additional vaccine platforms. An anti-idiotype vaccine4 in patients with follicular lymphoma demonstrated a median time to relapse of 44.2 months in the vaccine arm vs 30 months in the control arm (p=0.045). Compared with two other anti-idiotype vaccine trials5 that failed to meet end points, the patients in the successful trial had lower tumor burden. Vaccines in these trials were also produced differently. A modified GP100 peptide vaccine in adjuvant plus high-dose interleukin (IL)-26 demonstrated a longer median overall survival compared with the control IL-2 group only (17.8 months vs 11.1 months, p=0.06). There are several other ongoing Phase III trials using additional vaccine platforms in a range of human cancers, including emepepimut-S (Stimuvax®)7 (liposomal MUC-1 peptide) and belagenpumatucel-L (Lucanix™),8,9 which is an allogeneic whole tumor cell vaccine that contains a transforming growth factor (TGF)-β antisense transgene, both in patients with non-small cell lung cancer, and a MAGE-A3 protein vaccine in metastatic melanoma.10
Several issues are emerging from experience with multiple randomized vaccine clinical studies. These include: (a) there appears to be a greater vaccine efficacy in patients with low grade or more indolent disease compared with what one would observe with other forms of therapy.11 One potential reason for this is that it often takes multiple vaccinations and thus time to enhance the host T-cell response to tumor antigen(s) to a point at which it can effectively control tumor growth.12 Often patients have been taken off vaccine therapy before receiving optimal boosting regimens. (b) Patients who have had a longer duration since their last chemotherapy or have received fewer regimens of chemotherapy will respond better to vaccine.13,14 (c) The mechanism of action and kinetics of clinical response with vaccine compared with other forms of therapy appear to be quite different. Cytotoxic and even small molecule targeted therapies directly affect tumor only during the period of administration. If a drug is discontinued because of toxicity or drug resistance, all antitumor activity ceases and tumor growth rate will increase.
Recent vaccine studies have demonstrated that even in the absence of reduction of tumor size and/or time to progression, a slower growth rate of tumor can lead to enhanced survival.12 This phenomenon is most likely because vaccines induce a dynamic process of host immunity that can persist long after vaccine therapy is terminated and thus continue to control tumor growth. The same phenomenon may also be associated with the still anecdotal findings that some patients for whom vaccine therapy has failed have better than expected responses to subsequent therapies. A multicenter, randomized trial has now been initiated to evaluate this hypothesis. Patients with metastatic prostate cancer will be randomized in a multicenter trial to receive docetaxel vs PROSTVAC vaccine followed by docetaxel.15
It is anticipated that patient benefit may well occur with the use of vaccine combination therapies. This area of research/clinical trial design can be divided into two major segments: Vaccines in combination with other immune therapies and in combination with nonimmune therapeutic modalities. Preclinical and early clinical studies have demonstrated that a variety of immune stimulants can enhance vaccine efficacy. These include the use of one of numerous cytokines such as GM-CSF, IL-2, IL-15, IL-7, and interferons, all of which can enhance different components of the immune response. A range of toll-like receptor agonists as well as more “classical” adjuvants, such as incomplete Freund's, Monophosphoryl Lipid A (MPL), and chitosan, all have the potential to enhance vaccine efficacy.
Vaccines may also enhance immune and antitumor responses to monoclonal antibodies that are mediated by antibody-dependent cell-mediated cytotoxicity, as well as following adoptive T-cell transfer regimens.16 One area of investigation that is often overlooked is the use of two or more diverse vaccine platform combinations, either used concomitantly or in diversified prime-boost schemas. Preclinical studies have clearly demonstrated that diverse vaccine platforms can activate different components of the immune system and thus additively or synergistically enhance vaccine efficacy. Combination chemotherapy is standard of care for numerous tumor types, and combinations of small molecule targeted therapies are demonstrating enhanced patient benefit. There is also a strong rationale for the use of combinations of vaccines, and unlike combinations of chemotherapies and small molecule targeted therapies, there should be minimal issues with compounding toxicities.
Inhibitors of immune suppressive entities should also lead to enhanced vaccine efficacy. While the immune checkpoint inhibitor ipilimumab (anti-cytotoxic T-lymphocyte antigen (CTLA) 4 MAb) has now been approved by the FDA for use in metastatic melanoma,17 it and other immune checkpoint inhibitors, such as antiprogrammed death (PD)1 and anti-PDL1,18,19 will most certainly be evaluated in the future in combination with vaccines. Preclinical studies have demonstrated that the use of carcinoembryonic antigen (CEA)-TRICOM vaccine to enhance costimulation, in combination with the checkpoint inhibitor anti-CTLA4, can actually enhance T-cell avidity and subsequent antitumor activity.20 A single arm clinical trial has recently provided some preliminary evidence21,22 of patient benefit in the use of PROSTVAC vaccine with anti-CTLA4.
The impact of the tumor microenvironment remains an important consideration in obtaining optimal vaccine efficacy.23–27 In addition to cell-associated immune checkpoint inhibitors, numerous soluble immune suppressive factors such as TGF-β and IL-8 are found in the tumor microenvironment. While inhibitors of some of these molecules are currently in development, control of the tumor microenvironment to enhance vaccine efficacy will undoubtedly require combination therapies.
The next frontier for vaccine therapy will be the use of vaccines in combination with certain chemotherapeutic agents, radiation, hormone therapy, and certain small molecule targeted therapies. It has been elegantly shown that certain chemotherapeutic agents such as oxaliplatin and doxorubicin can induce “immunologic cell death,” which results in enhanced uptake of dead and dying tumor cells by dendritic cells resulting in activation of T cells to tumor antigens.28–30 Other chemotherapeutic agents such as docetaxel, as well as radiation of tumor, have been shown to alter the phenotype of tumor cells in terms of enhancing expression of tumor antigens, peptide major histocompatibility complexes (MHC) and death receptors, to render tumor cells more susceptible to vaccine-mediated T-cell killing.31–33 These two phenomena of immunologic tumor cell death and altering phenotype of tumor are not mutually exclusive, and both can lead to enhanced patient benefit.
Certain chemotherapeutic regimens such as cisplatin, vinorelbine, or cyclophosphamide have also been shown to preferentially reduce the number of immune suppressive cells such as regulatory T cells and myeloid-derived suppressor cells.34,35 This has also been observed with the use of small molecule targeted therapies such as a BCL-2 inhibitor and the tyrosine kinase inhibitor sunitinib, both of which have been shown in preclinical models to enhance the ratio of tumor-specific T cells to regulatory cells, resulting in enhanced vaccine antitumor efficacy.36–39
Antiandrogen hormonal therapy is standard of care for several stages of prostate cancer and has been shown to induce thymic regeneration, resulting in the induction of naïve T cells40,41; this phenomenon could potentially render greater vaccine-induced T-cell responses during a period of hormone therapy.42 Clinical trials have been completed, are ongoing, and are planned using vaccines in combination with hormonal therapy in different stages of prostate cancer.43–45 The challenge in all of the above combination therapies will be the appropriate scheduling of vaccine in relationship to the other therapeutic modalities as well as the appropriate patient population and clinical trial end point.
There are some 14 different vaccine platforms currently in Phase II and Phase III clinical studies, each of which encompasses a wide range of targeted tumor-associated antigens. One of the major findings in recent years is the realization that one can target, with vaccine-induced T cells, molecules that are responsible for tumor initiation or progression; these molecules need not be found on the tumor cell surface but are presented in peptide MHC complexes for vaccine-mediated T-cell recognition. Thus, one can potentially also target with vaccine therapy molecules involved in processes such as cancer cell “stemness,” epithelial-to-mesenchymal transition, and drug resistance.46–51 The era of therapeutic cancer vaccine therapy is thus now entering a new stage.
Disclosure Statement
The Laboratory of Tumor Immunology and Biology, NCI, has Collaborative Research and Development Agreements (CRADAs) with BN ImmunoTherapeutics, Inc.; GlobeImmune, Inc.; and EMD Serono/Merck.
References
- 1.Kantoff PW. Higano CS. Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. New Engl J Med. 2010;363:411. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
- 2.Higano CS. Schellhammer PF. Small EJ, et al. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer. 2009;115:3670. doi: 10.1002/cncr.24429. [DOI] [PubMed] [Google Scholar]
- 3.Kantoff PW. Schuetz TJ. Blumenstein BA, et al. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol. 2010;28:1099. doi: 10.1200/JCO.2009.25.0597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Schuster SJ. Neelapu SS. Gause BL, et al. Vaccination with patient-specific tumor-derived antigen in first remission improves disease-free survival in follicular lymphoma. J Clin Oncol. 2011;29:2787. doi: 10.1200/JCO.2010.33.3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Freedman A. Neelapu SS. Nichols C, et al. Placebo-controlled phase III trial of patient-specific immunotherapy with mitumprotimut-T and granulocyte-macrophage colony-stimulating factor after rituximab in patients with follicular lymphoma. J Clin Oncol. 2009;27:3036. doi: 10.1200/JCO.2008.19.8903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Schwartzentruber DJ. Lawson DH. Richards JM, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med. 2011;364:2119. doi: 10.1056/NEJMoa1012863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Butts C. Murray N. Maksymiuk A, et al. Randomized phase IIB trial of BLP25 liposome vaccine in stage IIIB and IV non-small-cell lung cancer. J Clin Oncol. 2005;23:6674. doi: 10.1200/JCO.2005.13.011. [DOI] [PubMed] [Google Scholar]
- 8.Mellstedt H. Vansteenkiste J. Thatcher N. Vaccines for the treatment of non-small cell lung cancer: Investigational approaches and clinical experience. Lung Cancer. 2011;73:11. doi: 10.1016/j.lungcan.2011.02.023. [DOI] [PubMed] [Google Scholar]
- 9.Nemunaitis J. Nemunaitis M. Senzer N, et al. Phase II trial of Belagenpumatucel-L, a TGF-beta2 antisense gene modified allogeneic tumor vaccine in advanced non-small cell lung cancer (NSCLC) patients. Cancer Gene Ther. 2009;16:620. doi: 10.1038/cgt.2009.15. [DOI] [PubMed] [Google Scholar]
- 10.Vansteenkiste J. Zielinski H. Linder A, et al. Final results of a muilti-center, double-blind, randomized, placebo-controlled Phase II study to assess the efficacy of MAGE-A3 immunotherapeutic as adjuvant therapy in stage Ib/II non-small cell lung cancer (NCSLC) [abst] J Clin Oncol. 2007;25(suppl18) [Google Scholar]
- 11.Gulley JL. Arlen PM. Madan RA, et al. Immunologic and prognostic factors associated with overall survival employing a poxviral-based PSA vaccine in metastatic castrate-resistant prostate cancer. Cancer Immunol Immunother. 2010;59:663. doi: 10.1007/s00262-009-0782-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Stein WD. Gulley JL. Schlom J, et al. Tumor regression and growth rates determined in five intramural NCI prostate cancer trials: The growth rate constant as an indicator of therapeutic efficacy. Clin Cancer Res. 2011;17:907. doi: 10.1158/1078-0432.CCR-10-1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.von Mehren M. Arlen P. Gulley J, et al. The influence of granulocyte macrophage colony-stimulating factor and prior chemotherapy on the immunological response to a vaccine (ALVAC-CEA B7.1) in patients with metastatic carcinoma. Clin Cancer Res. 2001;7:1181. [PubMed] [Google Scholar]
- 14.von Mehren M. Arlen P. Tsang KY, et al. Pilot study of a dual gene recombinant avipox vaccine containing both carcinoembryonic antigen (CEA) and B7.1 transgenes in patients with recurrent CEA-expressing adenocarcinomas. Clin Cancer Res. 2000;6:2219. [PubMed] [Google Scholar]
- 15.Docetaxel and prednisone with or without vaccine therapy in treating patients with metastatic hormone-resistant prostate cancer. www.clinicaltrials.gov/ct2/show/NCT01145508?term=McNeel&rank=5. [Dec 26;2011 ]. www.clinicaltrials.gov/ct2/show/NCT01145508?term=McNeel&rank=5
- 16.Bristol JA. Schlom J. Abrams SI. Persistence, immune specificity, and functional ability of murine mutant ras epitope-specific CD4(+) and CD8(+) T lymphocytes following in vivo adoptive transfer. Cell Immunol. 1999;194:78. doi: 10.1006/cimm.1999.1489. [DOI] [PubMed] [Google Scholar]
- 17.Hodi FS. O'Day SJ. McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Brahmer JR. Drake CG. Wollner I, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: Safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167. doi: 10.1200/JCO.2009.26.7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kline J. Gajewski TF. Clinical development of mAbs to block the PD1 pathway as an immunotherapy for cancer. Curr Opin Investig Drugs. 2010;11:1354. [PubMed] [Google Scholar]
- 20.Chakraborty M. Schlom J. Hodge JW. The combined activation of positive costimulatory signals with modulation of a negative costimulatory signal for the enhancement of vaccine-mediated T-cell responses. Cancer Immunol Immunother. 2007;56:1471. doi: 10.1007/s00262-007-0291-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Madan RA. Mohebtash M. Arlen PM, et al. A phase I trial of ipilimumab and a poxviral vaccine targeting PSA in metastatic castration-resistant prostate cancer: analysis of toxicity, response, and survival. Lancet Oncol. doi: 10.1016/S1470-2045(12)70006-2. Accepted for publication. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.van den Eertwegh AJM. Versluis J. van den Berg HP, et al. A phase I trial of combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells (GVAX) and ipilimumab in patients with metastatic castration-resistant prostate cancer. Lancet Oncol. doi: 10.1016/S1470-2045(12)70007-4. Accepted for publication. [DOI] [PubMed] [Google Scholar]
- 23.Carmeliet P. Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat Rev Drug Discov. 2011;10:417. doi: 10.1038/nrd3455. [DOI] [PubMed] [Google Scholar]
- 24.Condamine T. Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32:19. doi: 10.1016/j.it.2010.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gajewski TF. Failure at the effector phase: Immune barriers at the level of the melanoma tumor microenvironment. Clin Cancer Res. 2007;13:5256. doi: 10.1158/1078-0432.CCR-07-0892. [DOI] [PubMed] [Google Scholar]
- 26.Gajewski TF. Meng Y. Blank C, et al. Immune resistance orchestrated by the tumor microenvironment. Immunol Rev. 2006;213:131. doi: 10.1111/j.1600-065X.2006.00442.x. [DOI] [PubMed] [Google Scholar]
- 27.Gajewski TF. Meng Y. Harlin H. Immune suppression in the tumor microenvironment. J Immunother. 2006;29:233. doi: 10.1097/01.cji.0000199193.29048.56. [DOI] [PubMed] [Google Scholar]
- 28.Kepp O. Galluzzi L. Martins I, et al. Molecular determinants of immunogenic cell death elicited by anticancer chemotherapy. Cancer Metastasis Rev. 2011;30:61. doi: 10.1007/s10555-011-9273-4. [DOI] [PubMed] [Google Scholar]
- 29.Tesniere A. Schlemmer F. Boige V, et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 2010;29:482. doi: 10.1038/onc.2009.356. [DOI] [PubMed] [Google Scholar]
- 30.Zitvogel L. Kepp O. Senovilla L, et al. Immunogenic tumor cell death for optimal anticancer therapy: the calreticulin exposure pathway. Clin Cancer Res. 2010;16:3100. doi: 10.1158/1078-0432.CCR-09-2891. [DOI] [PubMed] [Google Scholar]
- 31.Chakraborty M. Abrams SI. Coleman CN, et al. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res. 2004;64:4328. doi: 10.1158/0008-5472.CAN-04-0073. [DOI] [PubMed] [Google Scholar]
- 32.Garnett CT. Schlom J. Hodge JW. Combination of docetaxel and recombinant vaccine enhances T-cell responses and antitumor activity: Effects of docetaxel on immune enhancement. Clin Cancer Res. 2008;14:3536. doi: 10.1158/1078-0432.CCR-07-4025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gelbard A. Garnett CT. Abrams SI, et al. Combination chemotherapy and radiation of human squamous cell carcinoma of the head and neck augments CTL-mediated lysis. Clin Cancer Res. 2006;12:1897. doi: 10.1158/1078-0432.CCR-05-1761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Emens LA. Asquith JM. Leatherman JM, et al. Timed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: A chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol. 2009;27:5911. doi: 10.1200/JCO.2009.23.3494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gameiro SR. Caballero JA. Higgins JP, et al. Exploitation of differential homeostatic proliferation of T-cell subsets following chemotherapy to enhance the efficacy of vaccine-mediated antitumor responses. Cancer Immunol Immunother. 2011;60:1227–1242. doi: 10.1007/s00262-011-1020-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Farsaci B. Higgins JP. Hodge JW. Consequence of dose scheduling of sunitinib on host immune response elements and vaccine combination therapy. Int J Cancer. 2011 Jun 1; doi: 10.1002/ijc.26219. E-pub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Farsaci B. Sabzevari H. Higgins JP, et al. Effect of a small molecule BCL-2 inhibitor on immune function and use with a recombinant vaccine. Int J Cancer. 2010;127:1603. doi: 10.1002/ijc.25177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Finke JH. Rini B. Ireland J, et al. Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients. Clin Cancer Res. 2008;14:6674. doi: 10.1158/1078-0432.CCR-07-5212. [DOI] [PubMed] [Google Scholar]
- 39.Ko JS. Zea AH. Rini BI, et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res. 2009;15:2148. doi: 10.1158/1078-0432.CCR-08-1332. [DOI] [PubMed] [Google Scholar]
- 40.Arredouani MS. Tseng-Rogenski SS. Hollenbeck BK, et al. Androgen ablation augments human HLA2.1-restricted T cell responses to PSA self-antigen in transgenic mice. Prostate. 2010;70:1002. doi: 10.1002/pros.21134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lee DK. Hakim FT. Gress RE. The thymus and the immune system: Layered levels of control. J Thorac Oncol. 2010;5:S273. doi: 10.1097/JTO.0b013e3181f20474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mercader M. Bodner BK. Moser MT, et al. T cell infiltration of the prostate induced by androgen withdrawal in patients with prostate cancer. Proc Natl Acad Sci U S A. 2001;98:14565. doi: 10.1073/pnas.251140998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bilusic M. Gulley J. Heery C, et al. A randomized phase II study of flutamide with or without PSA-TRICOM in nonmetastatic castration-resistant prostate cancer [abst] J Clin Oncol. 2011;29(suppl7) [Google Scholar]
- 44.Madan RA. Gulley JL. Schlom J, et al. Analysis of overall survival in patients with nonmetastatic castration-resistant prostate cancer treated with vaccine, nilutamide, and combination therapy. Clin Cancer Res. 2008;14:4526. doi: 10.1158/1078-0432.CCR-07-5048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Sanda MG. Smith DC. Charles LG, et al. Recombinant vaccinia-PSA (PROSTVAC) can induce a prostate-specific immune response in androgen-modulated human prostate cancer. Urology. 1999;53:260. doi: 10.1016/s0090-4295(98)00539-1. [DOI] [PubMed] [Google Scholar]
- 46.Dhodapkar KM. Feldman D. Matthews P, et al. Natural immunity to pluripotency antigen OCT4 in humans. Proc Natl Acad Sci U S A. 2010;107:8718. doi: 10.1073/pnas.0915086107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Fernando RI. Litzinger M. Trono P, et al. The T-box transcription factor Brachyury promotes epithelial-mesenchymal transition in human tumor cells. J Clin Invest. 2010;120:533. doi: 10.1172/JCI38379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hua W. Yao Y. Chu Y, et al. The CD133+ tumor stem-like cell-associated antigen may elicit highly intense immune responses against human malignant glioma. J Neurooncol. 2011 Apr 11; doi: 10.1007/s11060-011-0572-y. E-pub ahead of print. [DOI] [PubMed] [Google Scholar]
- 49.Mine T. Matsueda S. Li Y, et al. Breast cancer cells expressing stem cell markers CD44+ CD24 lo are eliminated by Numb-1 peptide-activated T cells. Cancer Immunol Immunother. 2009;58:1185. doi: 10.1007/s00262-008-0623-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Palena C. Polev DE. Tsang KY, et al. The human T-box mesodermal transcription factor Brachyury is a candidate target for T-cell-mediated cancer immunotherapy. Clin Cancer Res. 2007;13:2471. doi: 10.1158/1078-0432.CCR-06-2353. [DOI] [PubMed] [Google Scholar]
- 51.Polyak K. Weinberg RA. Transitions between epithelial and mesenchymal states: Acquisition of malignant and stem cell traits. Nat Rev Cancer. 2009;9:265. doi: 10.1038/nrc2620. [DOI] [PubMed] [Google Scholar]