Skip to main content
Top
Published in: Familial Cancer 2/2013

01-06-2013 | Original Article

Towards a vaccine to prevent cancer in Lynch syndrome patients

Authors: Magnus von Knebel Doeberitz, Matthias Kloor

Published in: Familial Cancer | Issue 2/2013

Login to get access

Abstract

Germline mutations of DNA mismatch repair (MMR) genes predispose Lynch syndrome mutation carriers to the development of MMR-deficient tumors. MMR-deficient tumors show high-level microsatellite instability (MSI-H) and are typically characterized by a comparatively favorable prognosis and the absence of distant organ metastasis. Lynch syndrome-associated cancers are characterized by a pronounced local anti-tumoral immune response and usually display dense lymphocyte infiltration. This finding strongly suggested that the immune system may play an active role in the surveillance and biology of these cancers. The progression of MMR deficient cancers seems to be triggered by mutations in microsatellite sequences within gene-encoding regions. These mutations may cause shifts of the translational reading frame and thus give rise to the generation of potentially immunogenic frameshift peptides (FSP) at the carboxy terminal end of the respective gene products. FSP-specific immune responses are thought to represent one major mechanism by which the host’s adaptive immune system can recognize and potentially control Lynch syndrome-associated MSI-H cancers. Consequently, vaccination with FSP antigens represent a promising approach for treatment of Lynch syndrome-associated cancers, potentially also suitable for tumor prevention in so far tumor-free Lynch syndrome germ line mutation carriers. This review will summarize the molecular mechanisms contributing to the immunological phenotype of MSI-H cancers. In addition, clinical perspectives will be discussed, focusing on MSI-H cancer-associated FSP antigens as potential targets for immune therapy approaches.
Literature
1.
go back to reference Watson P, Lin KM, Rodriguez-Bigas MA et al (1998) Colorectal carcinoma survival among hereditary nonpolyposis colorectal carcinoma family members. Cancer 83:259–266PubMedCrossRef Watson P, Lin KM, Rodriguez-Bigas MA et al (1998) Colorectal carcinoma survival among hereditary nonpolyposis colorectal carcinoma family members. Cancer 83:259–266PubMedCrossRef
2.
go back to reference Popat S, Hubner R, Houlston RS (2005) Systematic review of microsatellite instability and colorectal cancer prognosis. J Clin Oncol 23:609–618PubMedCrossRef Popat S, Hubner R, Houlston RS (2005) Systematic review of microsatellite instability and colorectal cancer prognosis. J Clin Oncol 23:609–618PubMedCrossRef
3.
go back to reference Kim H, Jen J, Vogelstein B, Hamilton SR (1994) Clinical and pathological characteristics of sporadic colorectal carcinomas with DNA replication errors in microsatellite sequences. Am J Pathol 145:148–156PubMed Kim H, Jen J, Vogelstein B, Hamilton SR (1994) Clinical and pathological characteristics of sporadic colorectal carcinomas with DNA replication errors in microsatellite sequences. Am J Pathol 145:148–156PubMed
4.
go back to reference Risio M, Reato G, di Celle PF et al (1996) Microsatellite instability is associated with the histological features of the tumor in nonfamilial colorectal cancer. Cancer Res 56:5470–5474PubMed Risio M, Reato G, di Celle PF et al (1996) Microsatellite instability is associated with the histological features of the tumor in nonfamilial colorectal cancer. Cancer Res 56:5470–5474PubMed
5.
go back to reference Dolcetti R, Viel A, Doglioni C et al (1999) High prevalence of activated intraepithelial cytotoxic T lymphocytes and increased neoplastic cell apoptosis in colorectal carcinomas with microsatellite instability. Am J Pathol 154:1805–1813PubMedCrossRef Dolcetti R, Viel A, Doglioni C et al (1999) High prevalence of activated intraepithelial cytotoxic T lymphocytes and increased neoplastic cell apoptosis in colorectal carcinomas with microsatellite instability. Am J Pathol 154:1805–1813PubMedCrossRef
6.
go back to reference Michael-Robinson JM, Biemer-Hüttmann A et al (2001) Tumour infiltrating lymphocytes and apoptosis are independent features in colorectal cancer stratified according to microsatellite instability status. Gut 48:360–366PubMedCrossRef Michael-Robinson JM, Biemer-Hüttmann A et al (2001) Tumour infiltrating lymphocytes and apoptosis are independent features in colorectal cancer stratified according to microsatellite instability status. Gut 48:360–366PubMedCrossRef
7.
go back to reference Shia J, Ellis NA, Paty PB et al (2003) Value of histopathology in predicting microsatellite instability in hereditary nonpolyposis colorectal cancer and sporadic colorectal cancer. Am J Surg Pathol 27:1407–1417PubMedCrossRef Shia J, Ellis NA, Paty PB et al (2003) Value of histopathology in predicting microsatellite instability in hereditary nonpolyposis colorectal cancer and sporadic colorectal cancer. Am J Surg Pathol 27:1407–1417PubMedCrossRef
8.
go back to reference Smyrk TC, Watson P, Kaul K et al (2001) Tumor-infiltrating lymphocytes are a marker for microsatellite instability in colorectal carcinoma. Cancer 91:2417–2422PubMedCrossRef Smyrk TC, Watson P, Kaul K et al (2001) Tumor-infiltrating lymphocytes are a marker for microsatellite instability in colorectal carcinoma. Cancer 91:2417–2422PubMedCrossRef
9.
go back to reference Galon J, Costes A, Sanchez-Cabo F et al (2006) Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313:1960–1964PubMedCrossRef Galon J, Costes A, Sanchez-Cabo F et al (2006) Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313:1960–1964PubMedCrossRef
10.
go back to reference Phillips SM, Banerjea A, Feakins R et al (2004) Tumour-infiltrating lymphocytes in colorectal cancer with microsatellite instability are activated and cytotoxic. Br J Surg 91:469–475PubMedCrossRef Phillips SM, Banerjea A, Feakins R et al (2004) Tumour-infiltrating lymphocytes in colorectal cancer with microsatellite instability are activated and cytotoxic. Br J Surg 91:469–475PubMedCrossRef
11.
go back to reference Michel S, Benner A, Tariverdian M et al (2008) High density of FOXP3-positive T cells infiltrating colorectal cancers with microsatellite instability. Br J Cancer 99:1867–1873PubMedCrossRef Michel S, Benner A, Tariverdian M et al (2008) High density of FOXP3-positive T cells infiltrating colorectal cancers with microsatellite instability. Br J Cancer 99:1867–1873PubMedCrossRef
12.
go back to reference Bauer K, Michel S, Reuschenbach M et al (2011) Dendritic cell and macrophage infiltration in microsatellite-unstable and microsatellite-stable colorectal cancer. Fam Cancer 10:557–565PubMedCrossRef Bauer K, Michel S, Reuschenbach M et al (2011) Dendritic cell and macrophage infiltration in microsatellite-unstable and microsatellite-stable colorectal cancer. Fam Cancer 10:557–565PubMedCrossRef
13.
go back to reference Banerjea A, Ahmed S, Hands RE et al (2004) Colorectal cancers with microsatellite instability display mRNA expression signatures characteristic of increased immunogenicity. Mol Cancer 3:21PubMedCrossRef Banerjea A, Ahmed S, Hands RE et al (2004) Colorectal cancers with microsatellite instability display mRNA expression signatures characteristic of increased immunogenicity. Mol Cancer 3:21PubMedCrossRef
14.
go back to reference Bernal M, García-Alcalde F, Concha A et al (2012) Genome-wide differential genetic profiling characterizes colorectal cancers with genetic instability and specific routes to HLA class I loss and immune escape. Cancer Immunol Immunother 61:803–816PubMedCrossRef Bernal M, García-Alcalde F, Concha A et al (2012) Genome-wide differential genetic profiling characterizes colorectal cancers with genetic instability and specific routes to HLA class I loss and immune escape. Cancer Immunol Immunother 61:803–816PubMedCrossRef
15.
go back to reference de Miranda NF, Goudkade D, Jordanova ES et al (2012) Infiltration of Lynch colorectal cancers by activated immune cells associates with early staging of the primary tumor and absence of lymph node metastases. Clin Cancer Res 18:1237–1245PubMedCrossRef de Miranda NF, Goudkade D, Jordanova ES et al (2012) Infiltration of Lynch colorectal cancers by activated immune cells associates with early staging of the primary tumor and absence of lymph node metastases. Clin Cancer Res 18:1237–1245PubMedCrossRef
16.
17.
go back to reference Duval A, Hamelin R (2002) Genetic instability in human mismatch repair deficient cancers. Ann Genet 45:71–75PubMedCrossRef Duval A, Hamelin R (2002) Genetic instability in human mismatch repair deficient cancers. Ann Genet 45:71–75PubMedCrossRef
18.
go back to reference Markowitz S, Wang J, Myeroff L et al (1995) Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science 268:1336–1338PubMedCrossRef Markowitz S, Wang J, Myeroff L et al (1995) Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science 268:1336–1338PubMedCrossRef
19.
go back to reference Ionov Y, Yamamoto H, Krajewski S et al (2000) Mutational inactivation of the proapoptotic gene BAX confers selective advantage during tumor clonal evolution. Proc Natl Acad Sci U S A 97:10872–10877PubMedCrossRef Ionov Y, Yamamoto H, Krajewski S et al (2000) Mutational inactivation of the proapoptotic gene BAX confers selective advantage during tumor clonal evolution. Proc Natl Acad Sci U S A 97:10872–10877PubMedCrossRef
20.
go back to reference Markowitz SD, Roberts AB (1996) Tumor suppressor activity of the TGF-beta pathway in human cancers. Cytokine Growth Factor Rev 7:93–102PubMedCrossRef Markowitz SD, Roberts AB (1996) Tumor suppressor activity of the TGF-beta pathway in human cancers. Cytokine Growth Factor Rev 7:93–102PubMedCrossRef
21.
go back to reference Woerner SM, Kloor M, von Knebel Doeberitz M et al (2006) Microsatellite instability in the development of DNA mismatch repair deficient tumors. Cancer Biomark 2:69–86PubMed Woerner SM, Kloor M, von Knebel Doeberitz M et al (2006) Microsatellite instability in the development of DNA mismatch repair deficient tumors. Cancer Biomark 2:69–86PubMed
22.
23.
go back to reference Saeterdal I, Bjørheim J, Lislerud K et al (2001) Frameshift-mutation-derived peptides as tumor-specific antigens in inherited and spontaneous colorectal cancer. Proc Natl Acad Sci U S A 98:13255–13260PubMedCrossRef Saeterdal I, Bjørheim J, Lislerud K et al (2001) Frameshift-mutation-derived peptides as tumor-specific antigens in inherited and spontaneous colorectal cancer. Proc Natl Acad Sci U S A 98:13255–13260PubMedCrossRef
24.
go back to reference Linnebacher M, Gebert J, Rudy W et al (2001) Frameshift peptide-derived T-cell epitopes: a source of novel tumor-specific antigens. Int J Cancer 93:6–11PubMedCrossRef Linnebacher M, Gebert J, Rudy W et al (2001) Frameshift peptide-derived T-cell epitopes: a source of novel tumor-specific antigens. Int J Cancer 93:6–11PubMedCrossRef
25.
go back to reference Ripberger E, Linnebacher M, Schwitalle Y et al (2003) Identification of an HLA-A0201-restricted CTL epitope generated by a tumor-specific frameshift mutation in a coding microsatellite of the OGT gene. J Clin Immunol 23:415–423PubMedCrossRef Ripberger E, Linnebacher M, Schwitalle Y et al (2003) Identification of an HLA-A0201-restricted CTL epitope generated by a tumor-specific frameshift mutation in a coding microsatellite of the OGT gene. J Clin Immunol 23:415–423PubMedCrossRef
26.
go back to reference Schwitalle Y, Linnebacher M, Ripberger E et al (2004) Immunogenic peptides generated by frameshift mutations in DNA mismatch repair-deficient cancer cells. Cancer Immun 4:14PubMed Schwitalle Y, Linnebacher M, Ripberger E et al (2004) Immunogenic peptides generated by frameshift mutations in DNA mismatch repair-deficient cancer cells. Cancer Immun 4:14PubMed
27.
go back to reference Garbe Y, Maletzki C, Linnebacher M (2011) An MSI tumor specific frameshift mutation in a coding microsatellite of MSH3 encodes for HLA-A0201-restricted CD8+ cytotoxic T cell epitopes. PLoS ONE 6:e26517PubMedCrossRef Garbe Y, Maletzki C, Linnebacher M (2011) An MSI tumor specific frameshift mutation in a coding microsatellite of MSH3 encodes for HLA-A0201-restricted CD8+ cytotoxic T cell epitopes. PLoS ONE 6:e26517PubMedCrossRef
28.
go back to reference Schwitalle Y, Kloor M, Eiermann S et al (2008) Immune response against frameshift-induced neopeptides in HNPCC patients and healthy HNPCC mutation carriers. Gastroenterology 134:988–997PubMedCrossRef Schwitalle Y, Kloor M, Eiermann S et al (2008) Immune response against frameshift-induced neopeptides in HNPCC patients and healthy HNPCC mutation carriers. Gastroenterology 134:988–997PubMedCrossRef
29.
go back to reference Bauer K, Nelius N, Reuschenbach M et al (2012) T cell responses against microsatellite instability-induced frameshift peptides and influence of regulatory T cells in colorectal cancer. Cancer Immunol Immunother 62:27–37 Bauer K, Nelius N, Reuschenbach M et al (2012) T cell responses against microsatellite instability-induced frameshift peptides and influence of regulatory T cells in colorectal cancer. Cancer Immunol Immunother 62:27–37
30.
go back to reference Ribic CM, Sargent DJ, Moore MJ et al (2003) Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med 349:247–257PubMedCrossRef Ribic CM, Sargent DJ, Moore MJ et al (2003) Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med 349:247–257PubMedCrossRef
31.
go back to reference Sargent DJ, Marsoni S, Monges G et al (2010) Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer. J Clin Oncol 28:3219–3226PubMedCrossRef Sargent DJ, Marsoni S, Monges G et al (2010) Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer. J Clin Oncol 28:3219–3226PubMedCrossRef
32.
go back to reference Sinicrope FA, Foster NR, Thibodeau SN et al (2011) DNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst 103:863–875PubMedCrossRef Sinicrope FA, Foster NR, Thibodeau SN et al (2011) DNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst 103:863–875PubMedCrossRef
33.
go back to reference Burn J, Gerdes AM, Macrae F et al (2011) Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet 378:2081–2087PubMedCrossRef Burn J, Gerdes AM, Macrae F et al (2011) Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet 378:2081–2087PubMedCrossRef
34.
go back to reference Czéh M, Loddenkemper C, Shalapour S et al (2010) The immune response to sporadic colorectal cancer in a novel mouse model. Oncogene 29:6591–6602PubMedCrossRef Czéh M, Loddenkemper C, Shalapour S et al (2010) The immune response to sporadic colorectal cancer in a novel mouse model. Oncogene 29:6591–6602PubMedCrossRef
35.
go back to reference Reuschenbach M, Kloor M, Morak M et al (2010) Serum antibodies against frameshift peptides in microsatellite unstable colorectal cancer patients with Lynch syndrome. Fam Cancer 9:173–179PubMedCrossRef Reuschenbach M, Kloor M, Morak M et al (2010) Serum antibodies against frameshift peptides in microsatellite unstable colorectal cancer patients with Lynch syndrome. Fam Cancer 9:173–179PubMedCrossRef
36.
go back to reference Marsh L, Coletta PL, Hull MA et al (2012) Altered intestinal epithelium-associated lymphocyte repertoires and function in ApcMin/+mice. Int J Oncol 40:243–250PubMed Marsh L, Coletta PL, Hull MA et al (2012) Altered intestinal epithelium-associated lymphocyte repertoires and function in ApcMin/+mice. Int J Oncol 40:243–250PubMed
37.
go back to reference Bicknell DC, Kaklamanis L, Hampson R et al (1996) Selection for beta 2-microglobulin mutation in mismatch repair-defective colorectal carcinomas. Curr Biol 6:1695–1697PubMedCrossRef Bicknell DC, Kaklamanis L, Hampson R et al (1996) Selection for beta 2-microglobulin mutation in mismatch repair-defective colorectal carcinomas. Curr Biol 6:1695–1697PubMedCrossRef
38.
go back to reference Kloor M, Becker C, Benner A et al (2005) Immunoselective pressure and human leukocyte antigen class I antigen machinery defects in microsatellite unstable colorectal cancers. Cancer Res 65:6418–6424PubMedCrossRef Kloor M, Becker C, Benner A et al (2005) Immunoselective pressure and human leukocyte antigen class I antigen machinery defects in microsatellite unstable colorectal cancers. Cancer Res 65:6418–6424PubMedCrossRef
39.
go back to reference Kloor M, Michel S, Buckowitz B et al (2007) Beta2-microglobulin mutations in microsatellite unstable colorectal tumors. Int J Cancer 121:454–458PubMedCrossRef Kloor M, Michel S, Buckowitz B et al (2007) Beta2-microglobulin mutations in microsatellite unstable colorectal tumors. Int J Cancer 121:454–458PubMedCrossRef
40.
go back to reference Dierssen JW, de Miranda NF, Ferrone S et al (2007) HNPCC versus sporadic microsatellite-unstable colon cancers follow different routes toward loss of HLA class I expression. BMC Cancer 7:33PubMedCrossRef Dierssen JW, de Miranda NF, Ferrone S et al (2007) HNPCC versus sporadic microsatellite-unstable colon cancers follow different routes toward loss of HLA class I expression. BMC Cancer 7:33PubMedCrossRef
41.
go back to reference Chang CC, Ferrone S (2007) Immune selective pressure and HLA class I antigen defects in malignant lesions. Cancer Immunol Immunother 56:227–236PubMedCrossRef Chang CC, Ferrone S (2007) Immune selective pressure and HLA class I antigen defects in malignant lesions. Cancer Immunol Immunother 56:227–236PubMedCrossRef
42.
go back to reference Koelzer VH, Baker K, Kassahn D et al (2012) Prognostic impact of β-2-microglobulin expression in colorectal cancers stratified by mismatch repair status. J Clin Pathol 65:996–1002 Koelzer VH, Baker K, Kassahn D et al (2012) Prognostic impact of β-2-microglobulin expression in colorectal cancers stratified by mismatch repair status. J Clin Pathol 65:996–1002
43.
go back to reference Tikidzhieva A, Benner A, Michel S et al (2012) Microsatellite instability and Beta2-Microglobulin mutations as prognostic markers in colon cancer: results of the FOGT-4 trial. Br J Cancer 106:1239–1245PubMedCrossRef Tikidzhieva A, Benner A, Michel S et al (2012) Microsatellite instability and Beta2-Microglobulin mutations as prognostic markers in colon cancer: results of the FOGT-4 trial. Br J Cancer 106:1239–1245PubMedCrossRef
44.
go back to reference Huang WC, Wu D, Xie Z et al (2006) Beta2-Microglobulin is a signaling and growth-promoting factor for human prostate cancer bone metastasis. Cancer Res 66:9108–9116PubMedCrossRef Huang WC, Wu D, Xie Z et al (2006) Beta2-Microglobulin is a signaling and growth-promoting factor for human prostate cancer bone metastasis. Cancer Res 66:9108–9116PubMedCrossRef
45.
go back to reference Josson S, Nomura T, Lin JT et al (2011) b2-Microglobulin induces epithelial to mesenchymal transition and confers cancer lethality and bone metastasis in human cancer cells. Cancer Res 71:2600–2610PubMedCrossRef Josson S, Nomura T, Lin JT et al (2011) b2-Microglobulin induces epithelial to mesenchymal transition and confers cancer lethality and bone metastasis in human cancer cells. Cancer Res 71:2600–2610PubMedCrossRef
46.
go back to reference Ma D, Luyten GP, Luyder TM et al (1995) Relationship between natural killer cell susceptibility and metastasis of human uveal melanoma cells in a murine model. Invest Ophthalmol Vis Sci 36:435–441PubMed Ma D, Luyten GP, Luyder TM et al (1995) Relationship between natural killer cell susceptibility and metastasis of human uveal melanoma cells in a murine model. Invest Ophthalmol Vis Sci 36:435–441PubMed
47.
go back to reference Jager MJ, Hurks HM, Levitskaya J et al (2002) HLA expression in uveal melanoma: there is no rule without some exception. Hum Immunol 63:444–451PubMedCrossRef Jager MJ, Hurks HM, Levitskaya J et al (2002) HLA expression in uveal melanoma: there is no rule without some exception. Hum Immunol 63:444–451PubMedCrossRef
48.
go back to reference Bernal M, Ruiz-Cabello F, Concha A et al (2012) Implication of the β2-microglobulin gene in the generation of tumor escape phenotypes. Cancer Immunol Immunother 61:1359–1371PubMedCrossRef Bernal M, Ruiz-Cabello F, Concha A et al (2012) Implication of the β2-microglobulin gene in the generation of tumor escape phenotypes. Cancer Immunol Immunother 61:1359–1371PubMedCrossRef
49.
go back to reference Michel S, Linnebacher M, Alcaniz J et al (2010) Lack of HLA class II antigen expression in microsatellite unstable colorectal carcinomas is caused by mutations in HLA class II regulatory genes. Int J Cancer 127:889–898PubMed Michel S, Linnebacher M, Alcaniz J et al (2010) Lack of HLA class II antigen expression in microsatellite unstable colorectal carcinomas is caused by mutations in HLA class II regulatory genes. Int J Cancer 127:889–898PubMed
51.
go back to reference Kloor M, Huth C, Voigt AY et al (2012) Prevalence of mismatch repair-deficient crypt foci in Lynch syndrome: a pathological study. Lancet Oncol 13:598–606PubMedCrossRef Kloor M, Huth C, Voigt AY et al (2012) Prevalence of mismatch repair-deficient crypt foci in Lynch syndrome: a pathological study. Lancet Oncol 13:598–606PubMedCrossRef
Metadata
Title
Towards a vaccine to prevent cancer in Lynch syndrome patients
Authors
Magnus von Knebel Doeberitz
Matthias Kloor
Publication date
01-06-2013
Publisher
Springer Netherlands
Published in
Familial Cancer / Issue 2/2013
Print ISSN: 1389-9600
Electronic ISSN: 1573-7292
DOI
https://doi.org/10.1007/s10689-013-9662-7

Other articles of this Issue 2/2013

Familial Cancer 2/2013 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine