Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Review
. 2017 Feb;66(2):259-271.
doi: 10.1007/s00262-016-1947-x. Epub 2016 Dec 31.

Rejection versus escape: the tumor MHC dilemma

Affiliations
Review

Rejection versus escape: the tumor MHC dilemma

Federico Garrido et al. Cancer Immunol Immunother. 2017 Feb.

Abstract

Most tumor cells derive from MHC-I-positive normal counterparts and remain positive at early stages of tumor development. T lymphocytes can infiltrate tumor tissue, recognize and destroy MHC class I (MHC-I)-positive cancer cells ("permissive" phase I). Later, MHC-I-negative tumor cell variants resistant to T-cell killing emerge. During this process, tumors first acquire a heterogeneous MHC-I expression pattern and finally become uniformly MHC-I-negative. This stage (phase II) represents a "non-permissive" encapsulated structure with tumor nodes surrounded by fibrous tissue containing different elements including leukocytes, macrophages, fibroblasts, etc. Molecular mechanisms responsible for total or partial MHC-I downregulation play a crucial role in determining and predicting the antigen-presenting capacity of cancer cells. MHC-I downregulation caused by reversible ("soft") lesions can be upregulated by TH1-type cytokines released into the tumor microenvironment in response to different types of immunotherapy. In contrast, when the molecular mechanism of the tumor MHC-I loss is irreversible ("hard") due to a genetic defect in the gene/s coding for MHC-I heavy chains (chromosome 6) or beta-2-microglobulin (B2M) (chromosome 15), malignant cells are unable to upregulate MHC-I, remain undetectable by cytotoxic T-cells, and continue to grow and metastasize. Based on the tumor MHC-I molecular analysis, it might be possible to define MHC-I phenotypes present in cancer patients in order to distinguish between non-responders, partial/short-term responders, and likely durable responders. This highlights the need for designing strategies to enhance tumor MHC-I expression that would allow CTL-mediated tumor rejection.

Keywords: Immune escape; MHC class I; PIVAC 15; Tumor rejection; Tumor tissue architecture; Tumor-infiltrating lymphocytes.

PubMed Disclaimer

Conflict of interest statement

Authors declare no conflict of interest.

Figures

Fig. 1
Fig. 1
Different patterns of HLA-I expression in human prostate cancer. Three different HLA-I immunolabeling patterns in prostate tumor tissues: homogeneously positive HLA-I expression, heterogeneous HLA-I staining and homogeneously HLA-I-negative pattern with a clear histological separation of HLA-I-negative tumor from HLA-positive stroma. W6/32 mAb directed against HLA-ABC/B2M complex have been used to characterize tumor tissue HLA-I expression patterns
Fig. 2
Fig. 2
Tissue architecture in HLA-I-positive and HLA-I-negative breast (a), lung (b) and bladder (c) carcinomas in relation with the patterns of tumor infiltration with CD3+ and CD8+ lymphocytes. HLA-I-positive tumor tissue architecture is “permissive,” with tumor infiltration by different mononuclear cells, including T-cells (phase I). At later stages, primary tumors are heterogeneous, composed of HLA-I-positive and HLA-I-negative tumor cells, and TILs. Finally, tumor masses become uniformly HLA-I-negative. Leukocytes and other cells are now located outside the tumor in the stroma with a clear morphological separation of tumor cells from the stroma generating an encapsulated “non-permissive” structure (phase II). W6/32 mAb were used to analyze the expression of HLA-I-ABC/B2M complex
Fig. 2
Fig. 2
Tissue architecture in HLA-I-positive and HLA-I-negative breast (a), lung (b) and bladder (c) carcinomas in relation with the patterns of tumor infiltration with CD3+ and CD8+ lymphocytes. HLA-I-positive tumor tissue architecture is “permissive,” with tumor infiltration by different mononuclear cells, including T-cells (phase I). At later stages, primary tumors are heterogeneous, composed of HLA-I-positive and HLA-I-negative tumor cells, and TILs. Finally, tumor masses become uniformly HLA-I-negative. Leukocytes and other cells are now located outside the tumor in the stroma with a clear morphological separation of tumor cells from the stroma generating an encapsulated “non-permissive” structure (phase II). W6/32 mAb were used to analyze the expression of HLA-I-ABC/B2M complex
Fig. 3
Fig. 3
HLA-I immunostaining patterns in primary melanoma (a) and colorectal tumor (b) and in corresponding autologous metastases. a Primary melanoma lesion has heterogeneous HLA-I labeling pattern with positive and negative areas surrounded by stroma. Autologous melanoma metastasis is uniformly negative for HLA-I expression, while the stroma is HLA-I-positive. b Primary colorectal cancer is clearly heterogeneous for HLA-I expression with positive and negative areas, while autologous liver metastasis is homogeneously HLA-I-negative
Fig. 4
Fig. 4
HLA-I-negative tumors encapsulated by surrounding fibrous stroma. a Breast carcinoma homogeneously negative for HLA-I with a clear separation from HLA-I-positive stroma surrounding tumor nests. CD3+ cells are outside the tumor tissue in the fibrous stromal capsule. b The “non-permissive” tumor tissue structure observed in HLA-I-negative tumors (lung, colorectal, laryngeal, bladder). HLA-I-negative tumor markedly contrasts with the HLA-I-positive stroma. Note that these tumor tissue examples represent primary lesions with a homogeneous negative HLA-I pattern. Tissue architecture with tumor nodes encapsulated by the stroma is clearly observed. W6/32 mAb were used to analyze the expression of HLA-I-ABC/B2M complex
Fig. 5
Fig. 5
Three-dimensional schematic demonstration of the two phases of tumor development with the corresponding tissue organization patterns: a phase I or “permissive” and b phase II or “non-permissive/encapsulated”. a In phase I, tumor cells are HLA-I-positive (green) and are surrounded and killed by CD8+ T lymphocytes (yellow). HLA-I-negative or HLA-I-deficient tumor cells (red) escape the recognition and destruction by TILs. The tumor is now heterogeneous and is composed of HLA-I-positive and HLA-I-negative cells. Cells other than T-cell tumor-infiltrating cells, such as macrophages (blue), can also be found within the tumor tissue. This pattern of the tumor/stroma tissue architecture is “permissive,” since it permits leukocytes and other cells to get in close contact with tumor cells. b In phase II as a result of T-cell immune selection, the tumor becomes homogeneously negative for HLA class I expression (red cells). These HLA-I-negative tumor cells are encapsulated by the stroma rich in tumor-specific T lymphocytes (yellow) and other cells (blue), such as Tregs and MDSCs. A clear physical separation between these two structures (tumor cells and the stroma) creates a “non-permissive” tissue structure which prevents immune cell infiltration into the tumor mass. Thus, due to this immunosuppressive tumor microenvironment, HLA-I-negative cells are in an immune privileged tumor structure
Fig. 6
Fig. 6
Frequency of HLA-I loss in different types of cancer. Summarized results of the immunohistological analysis of tumors using different antibodies directed against monomorphic, locus and allele-specific HLA determinants. For details, see Refs. [21, 22, 29, 39, 65, 67]
Fig. 7
Fig. 7
Recovery of HLA-I expression in melanoma cells: by IFN in the case of “soft” lesions and by adenovirus-mediated B2M gene transfer in cells with “hard” HLA-I lesions. a “soft” lesions—a selective HLA-B locus downregulation in ESTDAB-25 cells and total HLA-ABC downregulation in ESTDAB-42 cells—are corrected by IFN-γ (as demonstrated on flow cytometry plots); b resistance to IFN-γ-mediated HLA-ABC upregulation in melanoma cell line ESTDAB-004 (“hard” lesion); c recovery of HLA-I expression in melanoma cells with total loss of HLA-I expression due to B2M mutations (“hard” lesion) after adenovirus-mediated transfer of a wild-type human B2M gene (as demonstrated by confocal microscopy). W6/32 mAB were used to examine cell surface HLA-ABC/B2M complex; L368 mAb were used to detect B2M protein

Similar articles

Cited by

References

    1. Gorer PA. The genetic and antigenic basis of tumor transplantation. J Pathol Bacteriol. 1937;44:691–697. doi: 10.1002/path.1700440313. - DOI
    1. Foley EJ. Antigenic properties of methylcholanthrene-induced tumors in mice of the strain of origin. Cancer Res. 1953;13:835–837. - PubMed
    1. Baldwin RW. Immunity to methylcholanthrene-induced tumors in inbred rats following atrophy and regression of implanted tumors. Br J Cancer. 1955;9:652–657. doi: 10.1038/bjc.1955.70. - DOI - PMC - PubMed
    1. Prehn RT, Main JM. Immunity to methylcholanthrene-induce sarcomas. J Natl Cancer Inst. 1957;18:769–778. - PubMed
    1. Klein G, Sjogren HO, Klein E, Hellstrom KE. Demonstration of resistance against methylcholanthreneinduced sarcomas in the primary autochthonous host. Cancer Res. 1960;20:1561–1572. - PubMed

Substances