Skip to main content
Top
Published in: Annals of Surgical Oncology 12/2008

01-12-2008 | Melanomas

Evaluation of the Sentinel Immunized Node for Immune Monitoring of Cancer Vaccines

Authors: Craig L. Slingluff Jr, MD, Galina V. Yamshchikov, MS, Kevin T. Hogan, PhD, Sarah C. Hibbitts, BA, Gina R. Petroni, PhD, Eric A. Bissonette, MS, James W. Patterson, MD, Patrice Y. Neese, MS, William W. Grosh, MD, Kimberly A. Chianese-Bullock, PhD, Andrea Czarkowski, BS, Patrice K. Rehm, MD, Jayashree Parekh, MD

Published in: Annals of Surgical Oncology | Issue 12/2008

Login to get access

Abstract

Background

We hypothesized that lymph nodes draining sites of cutaneous vaccination could be identified by sentinel node biopsy techniques, and that measuring T-cell response with lymphocytes obtained from these lymph nodes would provide a more sensitive measure of immunogenicity than would the same measurement made with peripheral blood lymphocytes (PBL).

Methods

ELISpot analysis was used to determine the magnitude of vaccine-specific T-cell response in the sentinel immunized nodes (SIN), random lymph nodes, and peripheral blood lymphocytes (PBL) obtained from patients enrolled in clinical trials of experimental melanoma vaccines.

Results

The SIN biopsy was successful in 97% of cases and morbidity was very low. The T-cell response to vaccination was detected with greater sensitivity in the SIN (57%) than in PBL (39%), and evaluation of T-cell responses in the SIN and the PBL together yielded T-cell responses in 63% of patients. When the T-cell responses from a SIN and a random lymph node were compared in four patients, immune responses were detected to one of the vaccine peptides in three of these four patients. In all of those cases, responses were present in the SIN but absent from the random lymph node.

Conclusion

Measurements of T-cell responsiveness to cutaneous immunization are more frequently positive in the SIN than they are in the PBL, however evaluation of both the SIN and PBL permit a more sensitive measure of T-cell immunogenicity than use of either single source.
Literature
1.
go back to reference Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med 2004; 10:909–15PubMedCrossRef Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med 2004; 10:909–15PubMedCrossRef
2.
go back to reference Drake CG, Jaffee E, Pardoll DM. Mechanisms of immune evasion by tumors. Adv Immunol 2006; 90:51–81PubMedCrossRef Drake CG, Jaffee E, Pardoll DM. Mechanisms of immune evasion by tumors. Adv Immunol 2006; 90:51–81PubMedCrossRef
3.
go back to reference Slingluff CL, Chianese-Bullock KA, Bullock TNJ, et al. Immunity to melanoma antigens: from self-tolerance to immunotherapy. Adv Immunol 2006; 90:243–95PubMedCrossRef Slingluff CL, Chianese-Bullock KA, Bullock TNJ, et al. Immunity to melanoma antigens: from self-tolerance to immunotherapy. Adv Immunol 2006; 90:243–95PubMedCrossRef
4.
go back to reference Marchand M, van Baren N, Weynants P, et al. 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 1999; 80:219–30PubMedCrossRef Marchand M, van Baren N, Weynants P, et al. 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 1999; 80:219–30PubMedCrossRef
5.
go back to reference Coulie PG, Karanikas V, Colau D, et al. A monoclonal cytolytic T-lymphocyte response observed in a melanoma patient vaccinated with a tumor-specific antigenic peptide encoded by gene MAGE-3. Proc Natl Acad Sci USA 2001; 98:10290–5PubMedCrossRef Coulie PG, Karanikas V, Colau D, et al. A monoclonal cytolytic T-lymphocyte response observed in a melanoma patient vaccinated with a tumor-specific antigenic peptide encoded by gene MAGE-3. Proc Natl Acad Sci USA 2001; 98:10290–5PubMedCrossRef
6.
go back to reference Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat Med 1998; 4:321–7PubMedCrossRef Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat Med 1998; 4:321–7PubMedCrossRef
7.
go back to reference Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Impact of cytokine administration on the generation of antitumor reactivity in patients with metastatic melanoma receiving a peptide vaccine. J Immunol 1999; 163:1690–5PubMed Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Impact of cytokine administration on the generation of antitumor reactivity in patients with metastatic melanoma receiving a peptide vaccine. J Immunol 1999; 163:1690–5PubMed
8.
go back to reference Yamshchikov GV, Barnd DL, Eastham S, et al. Evaluation of peptide vaccine immunogenicity in draining lymph nodes and peripheral blood of melanoma patients. Int J Cancer 2001; 92:703–11PubMedCrossRef Yamshchikov GV, Barnd DL, Eastham S, et al. Evaluation of peptide vaccine immunogenicity in draining lymph nodes and peripheral blood of melanoma patients. Int J Cancer 2001; 92:703–11PubMedCrossRef
9.
go back to reference Slingluff CL Jr, Petroni GR, Yamshchikov GV, et al. Clinical and immunologic results of a randomized phase II trial of vaccination using four melanoma peptides either administered in granulocyte-macrophage colony-stimulating factor in adjuvant or pulsed on dendritic cells. J Clin Oncol 2003; 21:4016–26PubMedCrossRef Slingluff CL Jr, Petroni GR, Yamshchikov GV, et al. Clinical and immunologic results of a randomized phase II trial of vaccination using four melanoma peptides either administered in granulocyte-macrophage colony-stimulating factor in adjuvant or pulsed on dendritic cells. J Clin Oncol 2003; 21:4016–26PubMedCrossRef
10.
go back to reference Slingluff CL Jr, Petroni GR, Yamshchikov GV, et al. Immunologic and clinical outcomes of vaccination with a multiepitope melanoma peptide vaccine plus low-dose interleukin-2 administered either concurrently or on a delayed schedule. J Clin Oncol 2004; 22:4474–85PubMedCrossRef Slingluff CL Jr, Petroni GR, Yamshchikov GV, et al. Immunologic and clinical outcomes of vaccination with a multiepitope melanoma peptide vaccine plus low-dose interleukin-2 administered either concurrently or on a delayed schedule. J Clin Oncol 2004; 22:4474–85PubMedCrossRef
11.
go back to reference Slingluff CL Jr, Petroni GR, Chianese-Bullock KA, et al. Immunologic and clinical outcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting. Clin Cancer Res 2007; 13:6386–95PubMedCrossRef Slingluff CL Jr, Petroni GR, Chianese-Bullock KA, et al. Immunologic and clinical outcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting. Clin Cancer Res 2007; 13:6386–95PubMedCrossRef
12.
go back to reference Storkus WJ, Salter RD, Cresswell P, et al. Peptide-induced modulation of target cell sensitivity to natural killing. J Immunol 1992; 149:1185–90PubMed Storkus WJ, Salter RD, Cresswell P, et al. Peptide-induced modulation of target cell sensitivity to natural killing. J Immunol 1992; 149:1185–90PubMed
13.
go back to reference Anderson KS, Alexander J, Wei M, et al. Intracellular transport of class I MHC molecules in antigen processing mutant cell lines. J Immunol 1993; 151:3407–19PubMed Anderson KS, Alexander J, Wei M, et al. Intracellular transport of class I MHC molecules in antigen processing mutant cell lines. J Immunol 1993; 151:3407–19PubMed
14.
go back to reference Kittlesen DJ, Thompson LW, Gulden PH, et al. Human melanoma patients recognize an HLA-A1-restricted CTL epitope from tyrosinase containing two cysteine residues: implications for tumor vaccine development. J Immunol 1998; 160:2099–106PubMed Kittlesen DJ, Thompson LW, Gulden PH, et al. Human melanoma patients recognize an HLA-A1-restricted CTL epitope from tyrosinase containing two cysteine residues: implications for tumor vaccine development. J Immunol 1998; 160:2099–106PubMed
15.
go back to reference Skipper JC, Hendrickson RC, Gulden PH, et al. An HLA-A2-restricted tyrosinase antigen on melanoma cells results from posttranslational modification and suggests a novel pathway for processing of membrane proteins. J Exp Med 1996; 183:527–34PubMedCrossRef Skipper JC, Hendrickson RC, Gulden PH, et al. An HLA-A2-restricted tyrosinase antigen on melanoma cells results from posttranslational modification and suggests a novel pathway for processing of membrane proteins. J Exp Med 1996; 183:527–34PubMedCrossRef
16.
go back to reference Cox AL, Skipper J, Chen Y, et al. Identification of a peptide recognized by five melanoma-specific human cytotoxic T cell lines. Science 1994; 264:716–9PubMedCrossRef Cox AL, Skipper J, Chen Y, et al. Identification of a peptide recognized by five melanoma-specific human cytotoxic T cell lines. Science 1994; 264:716–9PubMedCrossRef
17.
go back to reference Skipper JC, Kittlesen DJ, Hendrickson RC, et al. Shared epitopes for HLA-A3-restricted melanoma-reactive human CTL include a naturally processed epitope from Pmel-17/gp100. J Immunol 1996; 157:5027–33PubMed Skipper JC, Kittlesen DJ, Hendrickson RC, et al. Shared epitopes for HLA-A3-restricted melanoma-reactive human CTL include a naturally processed epitope from Pmel-17/gp100. J Immunol 1996; 157:5027–33PubMed
18.
go back to reference Slingluff CL Jr, Yamshchikov G, Neese P, et al. Phase I trial of a melanoma vaccine with gp100(280–288) peptide and tetanus helper peptide in adjuvant: immunologic and clinical outcomes. Clin Cancer Res 2001; 7:3012–24PubMed Slingluff CL Jr, Yamshchikov G, Neese P, et al. Phase I trial of a melanoma vaccine with gp100(280–288) peptide and tetanus helper peptide in adjuvant: immunologic and clinical outcomes. Clin Cancer Res 2001; 7:3012–24PubMed
19.
go back to reference Kawakami Y, Robbins PF, Wang X, et al. Identification of new melanoma epitopes on melanosomal proteins recognized by tumor infiltrating T lymphocytes restricted by HLA-A1, -A2, and -A3 alleles. J Immunol 1998; 161:6985–92PubMed Kawakami Y, Robbins PF, Wang X, et al. Identification of new melanoma epitopes on melanosomal proteins recognized by tumor infiltrating T lymphocytes restricted by HLA-A1, -A2, and -A3 alleles. J Immunol 1998; 161:6985–92PubMed
20.
go back to reference Traversari C, van der Bruggen P, Luescher IF, et al. A nonapeptide encoded by human gene MAGE-1 is recognized on HLA-A1 by cytolytic T lymphocytes directed against tumor antigen MZ2-E. J Exp Med 1992; 176:1453–7PubMedCrossRef Traversari C, van der Bruggen P, Luescher IF, et al. A nonapeptide encoded by human gene MAGE-1 is recognized on HLA-A1 by cytolytic T lymphocytes directed against tumor antigen MZ2-E. J Exp Med 1992; 176:1453–7PubMedCrossRef
21.
go back to reference Gaugler B, Van den Eynde B, van der Bruggen P, et al. Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes. J Exp Med 1994; 179:921–30PubMedCrossRef Gaugler B, Van den Eynde B, van der Bruggen P, et al. Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes. J Exp Med 1994; 179:921–30PubMedCrossRef
22.
go back to reference Parkhurst MR, Salgaller ML, Southwood S, et al. Improved induction of melanoma-reactive CTL with peptides from the melanoma antigen gp100 modified at HLA-A*0201-binding residues. J Immunol 1996; 157:2539–48PubMed Parkhurst MR, Salgaller ML, Southwood S, et al. Improved induction of melanoma-reactive CTL with peptides from the melanoma antigen gp100 modified at HLA-A*0201-binding residues. J Immunol 1996; 157:2539–48PubMed
23.
go back to reference Huang LQ, Brasseur F, Serrano A, et al. Cytolytic T lymphocytes recognize an antigen encoded by MAGE-A10 on a human melanoma. J Immunol 1999; 162:6849–54PubMed Huang LQ, Brasseur F, Serrano A, et al. Cytolytic T lymphocytes recognize an antigen encoded by MAGE-A10 on a human melanoma. J Immunol 1999; 162:6849–54PubMed
24.
go back to reference Chaux P, Luiten R, Demotte N, et al. Identification of five MAGE-A1 epitopes recognized by cytolytic T lymphocytes obtained by in vitro stimulation with dendritic cells transduced with MAGE-A1. J Immunol 1999; 163:2928–36PubMed Chaux P, Luiten R, Demotte N, et al. Identification of five MAGE-A1 epitopes recognized by cytolytic T lymphocytes obtained by in vitro stimulation with dendritic cells transduced with MAGE-A1. J Immunol 1999; 163:2928–36PubMed
25.
go back to reference Wang RF, Johnston SL, Southwood S, et al. Recognition of an antigenic peptide derived from tyrosinase-related protein-2 by CTL in the context of HLA-A31 and -A33. J Immunol 1998; 160:890–7PubMed Wang RF, Johnston SL, Southwood S, et al. Recognition of an antigenic peptide derived from tyrosinase-related protein-2 by CTL in the context of HLA-A31 and -A33. J Immunol 1998; 160:890–7PubMed
26.
go back to reference Renkvist N, Castelli C, Robbins PF, et al. A listing of human tumor antigens recognized by T cells. Cancer Immunol Immunother 2001; 50:3–15PubMedCrossRef Renkvist N, Castelli C, Robbins PF, et al. A listing of human tumor antigens recognized by T cells. Cancer Immunol Immunother 2001; 50:3–15PubMedCrossRef
27.
go back to reference Blum-Tirouvanziam U, Servis C, Habluetzel A, et al. Localization of HLA-A2.1-restricted T cell epitopes in the circumsporozoite protein of Plasmodium falciparum. J Immunol 1995; 154:3922–31PubMed Blum-Tirouvanziam U, Servis C, Habluetzel A, et al. Localization of HLA-A2.1-restricted T cell epitopes in the circumsporozoite protein of Plasmodium falciparum. J Immunol 1995; 154:3922–31PubMed
28.
go back to reference Johnson RP, Trocha A, Yang L, et al. HIV-1 gag-specific cytotoxic T lymphocytes recognize multiple highly conserved epitopes. Fine specificity of the gag-specific response defined by using unstimulated peripheral blood mononuclear cells and cloned effector cells. J Immunol 1991; 147:1512–21PubMed Johnson RP, Trocha A, Yang L, et al. HIV-1 gag-specific cytotoxic T lymphocytes recognize multiple highly conserved epitopes. Fine specificity of the gag-specific response defined by using unstimulated peripheral blood mononuclear cells and cloned effector cells. J Immunol 1991; 147:1512–21PubMed
29.
go back to reference Goulder PJ, Sewell AK, Lalloo DG, et al. Patterns of immunodominance in HIV-1-specific cytotoxic T lymphocyte responses in two human histocompatibility leukocyte antigens (HLA)-identical siblings with HLA-A*0201 are influenced by epitope mutation. J Exp Med 1997; 185:1423–33PubMedCrossRef Goulder PJ, Sewell AK, Lalloo DG, et al. Patterns of immunodominance in HIV-1-specific cytotoxic T lymphocyte responses in two human histocompatibility leukocyte antigens (HLA)-identical siblings with HLA-A*0201 are influenced by epitope mutation. J Exp Med 1997; 185:1423–33PubMedCrossRef
30.
go back to reference Brander C, Hartman KE, Trocha AK, et al. Lack of strong immune selection pressure by the immunodominant, HLA-A*0201-restricted cytotoxic T lymphocyte response in chronic human immunodeficiency virus-1 infection. J Clin Invest 1998; 101:2559–66PubMedCrossRef Brander C, Hartman KE, Trocha AK, et al. Lack of strong immune selection pressure by the immunodominant, HLA-A*0201-restricted cytotoxic T lymphocyte response in chronic human immunodeficiency virus-1 infection. J Clin Invest 1998; 101:2559–66PubMedCrossRef
31.
go back to reference Morton DL, Thompson JF, Cochran AJ, et al. Sentinel-node biopsy or nodal observation in melanoma. N Engl J Med 2006; 355:1307–17PubMedCrossRef Morton DL, Thompson JF, Cochran AJ, et al. Sentinel-node biopsy or nodal observation in melanoma. N Engl J Med 2006; 355:1307–17PubMedCrossRef
32.
go back to reference Sondak VK, Zager JS, Messina JL, et al. Sentinel lymph node biopsy as the standard of care for cutaneous melanoma. Clin Adv Hematol Oncol 2007; 5:483–90PubMed Sondak VK, Zager JS, Messina JL, et al. Sentinel lymph node biopsy as the standard of care for cutaneous melanoma. Clin Adv Hematol Oncol 2007; 5:483–90PubMed
33.
go back to reference Cimmino VM, Brown AC, Szocik JF, et al. Allergic reactions to isosulfan blue during sentinel node biopsy–a common event. Surgery 2001; 130:439–42PubMedCrossRef Cimmino VM, Brown AC, Szocik JF, et al. Allergic reactions to isosulfan blue during sentinel node biopsy–a common event. Surgery 2001; 130:439–42PubMedCrossRef
34.
go back to reference Leong SP, Donegan E, Heffernon W, et al. Adverse reactions to isosulfan blue during selective sentinel lymph node dissection in melanoma. Ann Surg Oncol 2000; 7:361–6PubMedCrossRef Leong SP, Donegan E, Heffernon W, et al. Adverse reactions to isosulfan blue during selective sentinel lymph node dissection in melanoma. Ann Surg Oncol 2000; 7:361–6PubMedCrossRef
35.
go back to reference Macatonia SE, Knight SC, Edwards AJ, et al. Localization of antigen on lymph node dendritic cells after exposure to the contact sensitizer fluorescein isothiocyanate. Functional and morphological studies. J Exp Med 1987; 166:1654–67PubMedCrossRef Macatonia SE, Knight SC, Edwards AJ, et al. Localization of antigen on lymph node dendritic cells after exposure to the contact sensitizer fluorescein isothiocyanate. Functional and morphological studies. J Exp Med 1987; 166:1654–67PubMedCrossRef
36.
go back to reference Rosato A, Zambon A, Macino B, et al. Anti-L-selectin monoclonal antibody treatment in mice enhances tumor growth by preventing CTL sensitization in peripheral lymph nodes draining the tumor area. Int J Cancer 1996; 65:847–51PubMedCrossRef Rosato A, Zambon A, Macino B, et al. Anti-L-selectin monoclonal antibody treatment in mice enhances tumor growth by preventing CTL sensitization in peripheral lymph nodes draining the tumor area. Int J Cancer 1996; 65:847–51PubMedCrossRef
37.
go back to reference Yoshizawa H, Chang AE, Shu S. Specific adoptive immunotherapy mediated by tumor-draining lymph node cells sequentially activated with anti-CD3 and IL-2. J Immunol 1991; 147:729–37PubMed Yoshizawa H, Chang AE, Shu S. Specific adoptive immunotherapy mediated by tumor-draining lymph node cells sequentially activated with anti-CD3 and IL-2. J Immunol 1991; 147:729–37PubMed
Metadata
Title
Evaluation of the Sentinel Immunized Node for Immune Monitoring of Cancer Vaccines
Authors
Craig L. Slingluff Jr, MD
Galina V. Yamshchikov, MS
Kevin T. Hogan, PhD
Sarah C. Hibbitts, BA
Gina R. Petroni, PhD
Eric A. Bissonette, MS
James W. Patterson, MD
Patrice Y. Neese, MS
William W. Grosh, MD
Kimberly A. Chianese-Bullock, PhD
Andrea Czarkowski, BS
Patrice K. Rehm, MD
Jayashree Parekh, MD
Publication date
01-12-2008
Publisher
Springer-Verlag
Published in
Annals of Surgical Oncology / Issue 12/2008
Print ISSN: 1068-9265
Electronic ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-008-0046-4

Other articles of this Issue 12/2008

Annals of Surgical Oncology 12/2008 Go to the issue