Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells

Willem W Overwijk, Marc R Theoret, Steven E Finkelstein, Deborah R Surman, Laurina A de Jong, Florry A Vyth-Dreese, Trees A Dellemijn, Paul A Antony, Paul J Spiess, Douglas C Palmer, David M Heimann, Christopher A Klebanoff, Zhiya Yu, Leroy N Hwang, Lionel Feigenbaum, Ada M Kruisbeek, Steven A Rosenberg, Nicholas P Restifo, Willem W Overwijk, Marc R Theoret, Steven E Finkelstein, Deborah R Surman, Laurina A de Jong, Florry A Vyth-Dreese, Trees A Dellemijn, Paul A Antony, Paul J Spiess, Douglas C Palmer, David M Heimann, Christopher A Klebanoff, Zhiya Yu, Leroy N Hwang, Lionel Feigenbaum, Ada M Kruisbeek, Steven A Rosenberg, Nicholas P Restifo

Abstract

Many tumor-associated antigens are derived from nonmutated "self" proteins. T cells infiltrating tumor deposits recognize self-antigens presented by tumor cells and can be expanded in vivo with vaccination. These T cells exist in a functionally tolerant state, as they rarely result in tumor eradication. We found that tumor growth and lethality were unchanged in mice even after adoptive transfer of large numbers of T cells specific for an MHC class I-restricted epitope of the self/tumor antigen gp100. We sought to develop new strategies that would reverse the functionally tolerant state of self/tumor antigen-reactive T cells and enable the destruction of large (with products of perpendicular diameters of >50 mm2), subcutaneous, unmanipulated, poorly immunogenic B16 tumors that were established for up to 14 d before the start of treatment. We have defined three elements that are all strictly necessary to induce tumor regression in this model: (a) adoptive transfer of tumor-specific T cells; (b) T cell stimulation through antigen-specific vaccination with an altered peptide ligand, rather than the native self-peptide; and (c) coadministration of a T cell growth and activation factor. Cells, vaccination, or cyto-kine given alone or any two in combination were insufficient to induce tumor destruction. Autoimmune vitiligo was observed in mice cured of their disease. These findings illustrate that adoptive transfer of T cells and IL-2 can augment the function of a cancer vaccine. Furthermore, these data represent the first demonstration of complete cures of large, established, poorly immunogenic, unmanipulated solid tumors using T cells specific for a true self/tumor antigen and form the basis for a new approach to the treatment of patients with cancer.

Figures

Figure 1.
Figure 1.
Despite large numbers of gp100-specific T cells, B16 melanoma grows normally in pmel-1 TCR transgenic mice. (A) Generation and characterization of pmel-1 TCR transgenic mice. Single cell suspensions of spleens from a 6-wk-old pmel-1 mouse and a nontransgenic C57BL/6 mouse littermate as well as pmel-1 splenocytes cultured with hgp10025–33 peptide were stained for CD8, Vβ13, and the activation markers CD25, CD44, CD62L, and CD69, and analyzed by FACS®. (B) Recognition of gp100 peptide by pmel-1 T cells. PBL from pmel-1 “D8” founder were cultured for 7 d with mgp10025–33 peptide, washed, and incubated with titrated doses of mgp10025–33 (native) or hgp10025–33 (altered) peptide. IFN-γ production was measured by ELISA on culture supernatants. (C) Pmel-1 T cells specifically recognize B16 melanoma. Cultured pmel-1 splenocytes (gray bars) or clone 9 (black bars) were coincubated with B16 melanoma, EL-4 thymoma, MCA207 sarcoma, MC38 colon carcinoma cells, or CM. Supernatants were assessed for IFN-γ production by ELISA. (D) B16 melanoma grows progressively in pmel-1 TCR transgenic mice. B16 cells were implanted in 6-wk-old pmel-1 T cell receptor transgenic mice and littermates not expressing the transgene. All experiments shown were performed independently at least two times with similar results.
Figure 2.
Figure 2.
Enhanced T cell and antitumor response by adoptively transferred T cells after vaccination with altered peptide ligand. (A) Enhanced antitumor efficacy upon vaccination with rVV encoding altered peptide ligand. Tumor growth was measured in C57BL/6 mice bearing 3-d B16 tumors that received pmel-1 splenocytes followed immediately by vaccination with rVVLacZ, rVVmgp100, rVVhgp100, or no treatment. (B) Increased numbers of pmel-1 T cells from mice immunized with rVV encoding altered peptide ligand. Lymphocytes from mice described in A were isolated from peripheral blood and stained for CD8 and Vβ13 before FACS® analysis. Numbers of CD8+Tm+ cells are depicted through time as percentage of total CD8+ cells. (C) Prolonged peptide/MHC dissociation time of altered peptide ligand. RMA-S cells were incubated with hgp10025–33 (closed symbols) or mgp10025–33 (open symbols, shown at corresponding concentrations) at 25°C overnight. The cells were washed three times, incubated at 37°C for the time designated, and added to 7-d cultured pmel-1 T cells for 24 h. IFN-γ in culture supernatants was quantified by ELISA. Experiments were performed independently at least twice with similar results.
Figure 3.
Figure 3.
Adoptive transfer of tumor-specific T cells combined with vaccination and IL-2 causes regression and cure of large, established tumors. B16 tumor was implanted subcutaneously into C57BL/6 mice treated by adoptive transfer of fresh pmel-1 splenocytes ± vaccination with rFPVhgp100 either 7 (A and B) or 14 d (C and D) after tumor inoculation. IL-2 was administered twice daily for six doses. Fresh or cultured splenocytes were effective in the treatment of large, established tumors. Splenocytes derived from an identically constructed TCR transgenic mouse with specificity for β-galactosidase were used as a control in some experiments (C and D) and were not therapeutic. Statistically significant tumor regression was seen in mice treated with pmel-1 cells given in combination with rFPVhgp100 and IL-2 in >20 independently performed experiments. There were at least five mice/group in all experiments. Mouse survival consistently correlated with tumor growth reduction.
Figure 4.
Figure 4.
Long-term (>1 yr) survival of mice bearing large, established B16 tumors after treatment with adoptive transfer of tumor-specific T cells combined with vaccination and IL-2 is associated with the development of vitiligo. C57BL/6 mice were treated with adoptive transfer of fresh or cultured pmel-1 transgenic splenocytes 14 d after inoculation with B16 melanoma and vaccinated with rFPVhgp100. IL-2 was administered twice daily for six doses. Mice treated with fresh naive or cultured transgenic T cells were cured of B16, and vitiligo was observed, which started at the former tumor site. At >1-yr after therapy, these mice remain tumor-free with progressive vitiligo. To illustrate the autoimmune vitiligo, a photograph of the cohort of 5/5 surviving mice treated with cultured pmel-1 cells from A is shown in B.
Figure 5.
Figure 5.
Endogenous host B or T lymphocytes are not required for the treatment of established B16 tumors. C57BL/6 Rag-1−/− knockout mice bearing subcutaneous B16 tumors established for 14 d were treated with 106 cultured pmel-1 cells given in combination with rFPVhgp100 and IL-2 as described previously. Similar results were obtained in five independently completed experiments.
Figure 6.
Figure 6.
Persistence and function of gp100 reactive T cells after adoptive transfer, vaccination, and administration of IL-2 in mice bearing subcutaneous B16 tumors. (A–C) Data derived from the same, representative experiment. (A) Representative treatment experiment using C57BL/6 mice bearing B16 tumors established for 3 d were treated with fresh pmel-1 T cells plus vaccination with rVVLacZ, rVVmgp100, or rVVhgp100 with or without IL-2. (B) Vaccine-induced specific T cells in peripheral blood. Numbers of CD8+ Tm+ cells isolated from PBL of groups from A are depicted as percentage of total CD8+ cells. (C) Effect of boost with vaccine and IL-2. Mice surviving to day 30 (continuation of B) were retreated by heterologous boosting with rFPVhgp100 plus IL-2. Numbers of CD8+Vβ13+ cells isolated from blood are depicted as percentage of total CD8+ cells. (D) Effect of IL-2 on the number and function of vaccine-induced gp100-specific T cells in tumor tissue. The absolute numbers used to calculate the ratios shown were obtained 7 d after treatment (14 d after tumor implantation) and are normalized for 300 mg of tumor tissue (the average weight of excised tumors on day 14). These numbers are as follows (IL-2/PBS): total lymphocyte gate (1,758:945), CD8+ T cells (1,271:619), CD8+Vβ13+ T cells (890:266), and CD8+Vβ13+ IFN-γ+ T cells (261:8). After a 4-h restimulation with 1 μM mgp10025–33 peptide, the number of CD8+Vβ13+ IFN-γ+ T cells measured from this group were (754:147) to give a ratio of 5:1 (not depicted).
Figure 7.
Figure 7.
Histological analysis reveals presence of Vβ13+ T cells in tumors and demonstrates the activating effects of IL-2. T cells and tissue architecture within tumors were visualized after vaccination with or without IL-2. C57BL/6 mice bearing 14-d B16 tumors received 107 pmel-1 splenocytes and vaccination with hgp10025–33 peptide in IFA and anti-CD40 with or without IL-2. As was the case for vaccination with rVVs or rFPVs encoding hgp100, vaccination with hgp10025–33 peptide in IFA and anti-CD40 after the adoptive transfer of pmel-1 cells was greatly enhanced with the addition of IL-2 in repeated experiments (not depicted). Tumors were excised 7 d after the vaccination and 4-μm frozen sections were stained with anti-Vβ13 mAb (top, arrows) or IgG control (bottom), counterstained with hematoxylin, and photographed at an original magnification of 200. (left) pmel-1 cells plus hgp100 vaccination. (right) pmel-1 cells plus hgp100 vaccination followed by IL-2 administration.

References

    1. Houghton, A.N., J.S. Gold, and N.E. Blachere. 2001. Immunity against cancer: lessons learned from melanoma. Curr. Opin. Immunol. 13:134–140.
    1. Weber, L.W., W.B. Bowne, J.D. Wolchok, R. Srinivasan, J. Qin, Y. Moroi, R. Clynes, P. Song, J.J. Lewis, and A.N. Houghton. 1998. Tumor immunity and autoimmunity induced by immunization with homologous DNA. J. Clin. Invest. 102:1258–1264.
    1. Pardoll, D.M. 2002. Spinning molecular immunology into successful immunotherapy. Nat. Rev. Immunol. 2:227–238.
    1. Rosenberg, S.A. 2001. Progress in human tumour immunology and immunotherapy. Nature. 411:380–384.
    1. Overwijk, W.W., D.S. Lee, D.R. Surman, K.R. Irvine, C.E. Touloukian, C.C. Chan, M.W. Carroll, B. Moss, S.A. Rosenberg, and N.P. Restifo. 1999. Vaccination with a recombinant vaccinia virus encoding a “self” antigen induces autoimmune vitiligo and tumor cell destruction in mice: requirement for CD4(+) T lymphocytes. Proc. Natl. Acad. Sci. USA. 96:2982–2987.
    1. Overwijk, W.W., and N.P. Restifo. 2000. Autoimmunity and the immunotherapy of cancer: targeting the “self” to destroy the “other.” Crit. Rev. Immunol. 20:433–450.
    1. Marchand, M., N. van Baren, P. Weynants, V. Brichard, B. Dreno, M.H. Tessier, E. Rankin, G. Parmiani, F. Arienti, Y. Humblet, et al. 1999. Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1. Int. J. Cancer 80:219–230.
    1. Nestle, F.O., S. Alijagic, M. Gilliet, Y. Sun, S. Grabbe, R. Dummer, G. Burg, and D. Schadendorf. 1998. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat. Med. 4:328–332.
    1. Rosenberg, S.A., J.C. Yang, D.J. Schwartzentruber, P. Hwu, F. Marincola, S.L. Topalian, N.P. Restifo, M.E. Dudley, S.L. Schwarz, P.J. Spiess, et al. 1998. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat. Med. 4:321–327.
    1. Nguyen, L.T., A.R. Elford, K. Murakami, K.M. Garza, S.P. Schoenberger, B. Odermatt, D.E. Speiser, and P.S. Ohashi. 2002. Tumor growth enhances cross-presentation leading to limited T cell activation without tolerance. J. Exp. Med. 195:423–435.
    1. Touloukian, C.E., W.W. Leitner, R.E. Schnur, P.F. Robbins, Y. Li, S. Southwood, A. Sette, S.A. Rosenberg, and N.P. Restifo. 2003. Normal tissue depresses while tumor tissue enhances human T cell responses in vivo to a novel self/tumor melanoma antigen, OA1. J. Immunol. 170:1579–1585.
    1. Touloukian, C.E., W.W. Leitner, P.F. Robbins, Y.F. Li, X. Kang, R. Lapointe, P. Hwu, S.A. Rosenberg, and N.P. Restifo. 2002. Expression of a “self-”antigen by human tumor cells enhances tumor antigen-specific CD4(+) T-cell function. Cancer Res. 62:5144–5147.
    1. Speiser, D.E., R. Miranda, A. Zakarian, M.F. Bachmann, K. McKall-Faienza, B. Odermatt, D. Hanahan, R.M. Zinkernagel, and P.S. Ohashi. 1997. Self antigens expressed by solid tumors do not efficiently stimulate naive or activated T cells: implications for immunotherapy. J. Exp. Med. 186:645–653.
    1. Staveley-O'Carroll, K., E. Sotomayor, J. Montgomery, I. Borrello, L. Hwang, S. Fein, D. Pardoll, and H. Levitsky. 1998. Induction of antigen-specific T cell anergy: an early event in the course of tumor progression. Proc. Natl. Acad. Sci. USA. 95:1178–1183.
    1. Spiotto, M.T., P. Yu, D.A. Rowley, M.I. Nishimura, S.C. Meredith, T.F. Gajewski, Y.X. Fu, and H. Schreiber. 2002. Increasing tumor antigen expression overcomes “ignorance” to solid tumors via crosspresentation by bone marrow-derived stromal cells. Immunity. 17:737–747.
    1. Wang, M., V. Bronte, P.W. Chen, L. Gritz, D. Panicali, S.A. Rosenberg, and N.P. Restifo. 1995. Active immunotherapy of cancer with a nonreplicating recombinant fowlpox virus encoding a model tumor-associated antigen. J. Immunol. 154:4685–4692.
    1. Ochsenbein, A.F., S. Sierro, B. Odermatt, M. Pericin, U. Karrer, J. Hermans, S. Hemmi, H. Hengartner, and R.M. Zinkernagel. 2001. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature. 411:1058–1064.
    1. Tham, E.L., P. Shrikant, and M.F. Mescher. 2002. Activation-induced nonresponsiveness: a Th-dependent regulatory checkpoint in the CTL response. J. Immunol. 168:1190–1197.
    1. Hanson, H.L., D.L. Donermeyer, H. Ikeda, J.M. White, V. Shankaran, L.J. Old, H. Shiku, R.D. Schreiber, and P.M. Allen. 2000. Eradication of established tumors by CD8+ T cell adoptive immunotherapy. Immunity. 13:265–276.
    1. Wick, M., P. Dubey, H. Koeppen, C.T. Siegel, P.E. Fields, L. Chen, J.A. Bluestone, and H. Schreiber. 1997. Antigenic cancer cells grow progressively in immune hosts without evidence for T cell exhaustion or systemic anergy. J. Exp. Med. 186:229–238.
    1. Melief, C.J., S.H. Van Der Burg, R.E. Toes, F. Ossendorp, and R. Offringa. 2002. Effective therapeutic anticancer vaccines based on precision guiding of cytolytic T lymphocytes. Immunol. Rev. 188:177–182.
    1. Dranoff, G., E. Jaffee, A. Lazenby, P. Golumbek, H. Levitsky, K. Brose, V. Jackson, H. Hamada, D. Pardoll, and R.C. Mulligan. 1993. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc. Natl. Acad. Sci. USA. 90:3539–3543.
    1. Seliger, B., U. Wollscheid, F. Momburg, T. Blankenstein, and C. Huber. 2001. Characterization of the major histocompatibility complex class I deficiencies in B16 melanoma cells. Cancer Res. 61:1095–1099.
    1. Kawakami, Y., N. Dang, X. Wang, J. Tupesis, P.F. Robbins, R.F. Wang, J.R. Wunderlich, J.R. Yannelli, and S.A. Rosenberg. 2000. Recognition of shared melanoma antigens in association with major HLA-A alleles by tumor infiltrating T lymphocytes from 123 patients with melanoma. J. Immunother. 23:17–27.
    1. Sarma, S., Y. Guo, Y. Guilloux, C. Lee, X.F. Bai, and Y. Liu. 1999. Cytotoxic T lymphocytes to an unmutated tumor rejection antigen P1A: normal development but restrained effector function in vivo. J. Exp. Med. 189:811–820.
    1. Colella, T.A., T.N. Bullock, L.B. Russell, D.W. Mullins, W.W. Overwijk, C.J. Luckey, R.A. Pierce, N.P. Restifo, and V.H. Engelhard. 2000. Self-tolerance to the murine homologue of a tyrosinase-derived melanoma antigen. Implications for tumor immunotherapy. J. Exp. Med. 191:1221–1232.
    1. Van Parijs, L., and A.K. Abbas. 1998. Homeostasis and self-tolerance in the immune system: turning lymphocytes off. Science. 280:243–248.
    1. Irvine, K.R., M.R. Parkhurst, E.P. Shulman, J.P. Tupesis, M. Custer, C.E. Touloukian, P.F. Robbins, A.G. Yafal, P. Greenhalgh, R.P. Sutmuller, et al. 1999. Recombinant virus vaccination against “self” antigens using anchor-fixed immunogens. Cancer Res. 59:2536–2540.
    1. Slansky, J.E., F.M. Rattis, L.F. Boyd, T. Fahmy, E.M. Jaffee, J.P. Schneck, D.H. Margulies, and D.M. Pardoll. 2000. Enhanced antigen-specific antitumor immunity with altered peptide ligands that stabilize the MHC-peptide-TCR complex. Immunity. 13:529–538.
    1. Dyall, R., W.B. Bowne, L.W. Weber, J. LeMaoult, P. Szabo, Y. Moroi, G. Piskun, J.J. Lewis, A.N. Houghton, and J. Nikolic-Zugic. 1998. Heteroclitic immunization induces tumor immunity. J. Exp. Med. 188:1553–1561.
    1. Overwijk, W.W., A. Tsung, K.R. Irvine, M.R. Parkhurst, T.J. Goletz, K. Tsung, M.W. Carroll, C. Liu, B. Moss, S.A. Rosenberg, and N.P. Restifo. 1998. gp100/pmel 17 is a murine tumor rejection antigen: induction of “self”-reactive, tumoricidal T cells using high-affinity, altered peptide ligand. J. Exp. Med. 188:277–286.
    1. Zhai, Y., J.C. Yang, P. Spiess, M.I. Nishimura, W.W. Overwijk, B. Roberts, N.P. Restifo, and S.A. Rosenberg. 1997. Cloning and characterization of the genes encoding the murine homologues of the human melanoma antigens MART1 and gp100. J. Immunother. 20:15–25.
    1. Kouskoff, V., K. Signorelli, C. Benoist, and D. Mathis. 1995. Cassette vectors directing expression of T cell receptor genes in transgenic mice. J. Immunol. Methods. 180:273–280.
    1. Overwijk, W.W., D.R. Surman, K. Tsung, and N.P. Restifo. 1997. Identification of a Kb-restricted CTL epitope of beta-galactosidase: potential use in development of immunization protocols for “self” antigens. Methods. 12:117–123.
    1. Earl, P.L., N. Cooper, and B. Moss. 1991. Preparation of cell cultures and vaccinia virus stocks. In Current Protocols in Molecular Biology. F.M. Asubel, R. Brent, R.E. Kingston, D.D. Moore, J.G. Seidman, and L.A. Smith, editors. Greene Publishing Associates and Wiley Interscience, New York. 16.16.1–16.16.17.
    1. Vyth-Dreese, F.A., H. Boot, T.A. Dellemijn, D.M. Majoor, L.C. Oomen, J.D. Laman, M. Van Meurs, R.A. De Weger, and D. De Jong. 1998. Localization in situ of costimulatory molecules and cytokines in B-cell non-Hodgkin's lymphoma. Immunology. 94:580–586.
    1. Sotomayor, E.M., I. Borrello, E. Tubb, F.M. Rattis, H. Bien, Z. Lu, S. Fein, S. Schoenberger, and H.I. Levitsky. 1999. Conversion of tumor-specific CD4+ T-cell tolerance to T-cell priming through in vivo ligation of CD40. Nat. Med. 5:780–787.
    1. Bronte, V., K. Tsung, J.B. Rao, P.W. Chen, M. Wang, S.A. Rosenberg, and N.P. Restifo. 1995. IL-2 enhances the function of recombinant poxvirus-based vaccines in the treatment of established pulmonary metastases. J. Immunol. 154:5282–5292.
    1. Tamura, Y., P. Peng, K. Liu, M. Daou, and P.K. Srivastava. 1997. Immunotherapy of tumors with autologous tumor-derived heat shock protein preparations. Science. 278:117–120.
    1. Kannan, K., N.E. Sharpless, J. Xu, R.C. O'Hagan, M. Bosenberg, and L. Chin. 2003. Components of the Rb pathway are critical targets of UV mutagenesis in a murine melanoma model. Proc. Natl. Acad. Sci. USA. 100:1221–1225.
    1. Noonan, F.P., J.A. Recio, H. Takayama, P. Duray, M.R. Anver, W.L. Rush, E.C. De Fabo, and G. Merlino. 2001. Neonatal sunburn and melanoma in mice. Nature. 413:271–272.
    1. Lee, P.P., C. Yee, P.A. Savage, L. Fong, D. Brockstedt, J.S. Weber, D. Johnson, S. Swetter, J. Thompson, P.D. Greenberg, et al. 1999. Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients. Nat. Med. 5:677–685.
    1. Gold, J.S., C.R. Ferrone, J.A. Guevara-Patino, W.G. Hawkins, R. Dyall, M.E. Engelhorn, J.D. Wolchok, J.J. Lewis, and A.N. Houghton. 2003. A single heteroclitic epitope determines cancer immunity after xenogeneic DNA immunization against a tumor differentiation antigen. J. Immunol. 170:5188–5194.
    1. Wherry, E.J., J.N. Blattman, K. Murali-Krishna, M.R. van der, and R. Ahmed. 2003. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J. Virol. 77:4911–4927.
    1. Dudley, M.E., J.R. Wunderlich, P.F. Robbins, J.C. Yang, P. Hwu, D.J. Schwartzentruber, S.L. Topalian, R. Sherry, N.P. Restifo, A.M. Hubicki, et al. 2002. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science. 298:850–854.
    1. Kjaergaard, J., L. Peng, P.A. Cohen, J.A. Drazba, A.D. Weinberg, and S. Shu. 2001. Augmentation versus inhibition: effects of conjunctional OX-40 receptor monoclonal antibody and IL-2 treatment on adoptive immunotherapy of advanced tumor. J. Immunol. 167:6669–6677.
    1. Overwijk, W.W., M.R. Theoret, and N.P. Restifo. 2000. The future of interleukin-2: enhancing therapeutic anticancer vaccines. Cancer J. Sci. Am. 6:S76–S80.
    1. Blattman, J.N., J.M. Grayson, E.J. Wherry, S.M. Kaech, K.A. Smith, and R. Ahmed. 2003. Therapeutic use of IL-2 to enhance antiviral T-cell responses in vivo. Nat. Med. 9:540–547.
    1. Trcka, J., Y. Moroi, R.A. Clynes, S.M. Goldberg, A. Bergtold, M.A. Perales, M. Ma, C.R. Ferrone, M.C. Carroll, J.V. Ravetch, and A.N. Houghton. 2002. Redundant and alternative roles for activating Fc receptors and complement in an antibody-dependent model of autoimmune vitiligo. Immunity. 16:861–868.
    1. Bowne, W.B., R. Srinivasan, J.D. Wolchok, W.G. Hawkins, N.E. Blachere, R. Dyall, J.J. Lewis, and A.N. Houghton. 1999. Coupling and uncoupling of tumor immunity and autoimmunity. J. Exp. Med. 190:1717–1722.
    1. Rosenberg, S.A., and D.E. White. 1996. Vitiligo in patients with melanoma: normal tissue antigens can be targets for cancer immunotherapy. J. Immunother. Emphasis Tumor Immunol. 19:81–84.

Source: PubMed

3
Prenumerera