Abstract
Although the adaptive immune system can detect and eliminate malignant cells, patients with intact and fully functional immune systems develop head and neck cancer. How is this paradox explained? Manuscripts published in the English language from 1975 to 2018 were reviewed using search inputs related to tumor cell antigenicity and immunogenicity, immunodominance, cancer immunoediting and genomic alterations present within carcinomas. Early in tumor development, T cell responses to immunodominant antigens may lead to the elimination of cancer cells expressing these antigens and a tumor composed to tumor cells expressing only immunorecessive antigens. Conversely, other tumor cells may acquire genomic or epigenetic alterations that result in an antigen processing or presentation defect or other inability to be detected or killed by T cells. Such T cell insensitive tumor cells may also be selected for in a progressing tumor. Tumors harboring subpopulations of cells that cannot be eliminated by T cells may require non-T cell-based treatments, such as NK cell immunotherapies. Recognition of such tumor cell populations within a heterogeneous cancer may inform the selection of treatment for HNSCC in the future.
Keywords: Immunodominance, immune escape, antigenicity, immunogenicity, immunotherapy
Introduction
Genomic rearrangement of T cell receptor (TCR) genes produces an estimated 1013 different T cell clonotypes giving the adaptive immune system the capacity to respond to a large number of unique antigens1. These antigens, presented to T cells via major histocompatibility complex (MHC) molecules, can be derived from foreign pathogens or, in the case of cancer, from altered endogenous proteins. When altered or mutated endogenous proteins are processed and presented on MHC with subsequent recognition by T cells, these antigens are termed neoantigens. Head and neck squamous cell carcinomas (HNSCCs) typically harbor many genomic alterations and thus, HNSCCs are predicted to harbor many neoantigens2–4. So how do cancers with many genomic alterations and neoantigens escape T cell recognition and elimination? One evasive mechanism employed by malignant cells is the expression of chemokines and cytokines that generate an immunosuppressive tumor microenvironment blocking the function of effector immune cells5–7. However, development of an immunosuppressive tumor microenvironment may primarily be a mechanism of immune suppression in later-stage tumors that have accumulated a mass of tumors cells that produce chemokines sufficient to mediate chemotaxis of immunosuppressive cells5.
Early in tumor development, it is likely that variable antigenicity of tumor cells drives the formation of a tumor composed of tumor cells that are able to escape immunity8. The genomic instability of malignant cells results in a high degree of cancer cell heterogeneity9. Some tumor cells harbor strong (immunodominant) T cell antigens that are likely eliminated by the host’s immune system early in tumor development10. This process, termed immunoediting, leads to the development of clinically relevant tumors that only harbor weaker (immunorecessive) antigens11. Complicating this further, antigen-specific T cells tend to develop against immunodominant and not immunorecessive antigens, even when the immunodominant antigen has been removed from the tumor via immunoediting12–14. Therefore, some tumors may be susceptible to immune detection and elimination if T cell responses against immunorecessive antigens develop, either naturally or after immunotherapy designed to activate T cell immunity.
Aside from differences in tumor cell antigenicity driving immune escape, differences in immunogenicity (or how well the tumor cell is recognized by T cells) may also be playing a role. Through genomic instability and selection pressure in the face of anti-tumor immunity, tumor cells that harbor genomic or epigenetic alterations that interfere with antigen processing and presentation, interferon responses or susceptibility to T cell killing may also be selected by the immune system15–18. Translationally, this is more ominous and means that some cancers may simply be resistant to immunotherapy designed to activate T cell immunity, even if they harbor T cell antigens. Here, we review the underlying genomic alterations present within tumor cells that lead to resistance to T cell recognition and killing. Recognizing and fully elucidating these intrinsic mechanisms of resistance is required to begin to devise means of reversing these processes to enhance the efficacy of therapies relying on cytotoxic T cell responses.
How should antigen-specific T cell immunity develop?
Analysis of how tumor cells evade T cell detection and elimination requires an understanding of the normal biology. The process of T cell detection of infected or cancerous cells begins when clonally expanded antigen-specific T cells recognize antigen presented on the surface of target cells via MHC class I (MHC I). To maximize chances that they see the antigen for which they are specific, activated T cells express interferon gamma (IFNγ) that in turn potently induces MHC class I expression on tumor cells through the IFN receptor/JAK/STAT signaling pathway19. Integrins and cell adhesion molecules strengthen the immunological synapse and “hold” T cells to target cells for a period of time to allow effector function and killing. Antigen:MHC complex binding to the antigen specific T cell receptor (TCR) triggers rapid release of preformed granules containing perforin and granzymes. Upon cell entry, granzymes mediate the early (first 8–16 hours) cytotoxic effects of T cells by acting as potent proteases and activating caspase-independent apoptosis20,21. Secondary mechanisms of T cell killing that occur after granule exocytosis include expression of tumor necrosis factor (TNF)-superfamily ligands such as TNFα, Fas ligand and TRAIL. The ligands bind different TNF family receptors to activate extrinsic apoptosis pathways. One or more genomic alterations or epigenetic expression defects in any of these biologic processes could act as a mechanism of resistance to T cell recognition and killing.
This review will categorically explore intrinsic mechanisms of resistance to T cell recognition and killing that have been identified in head and neck cancers:
-
1)
altered antigen processing and presentation
-
2)
absent or defective IFNγ receptor expression or downstream signaling
-
3)
defective formation of the immunologic synapse
-
4)
neutralization of granzyme released from T cells
-
5)
resistance to death receptor-induced extrinsic apoptosis
-
6)
caspase mutations
-
7)
tumor cell intrinsic cell cycle alterations in response to cytotoxic insults
Discussion
1). Alterations in antigen processing and presentation
For CTLs to recognize an antigen on MHC I, the cell must first generate peptide to serve as antigen. Figure 1 demonstrates critical steps in this process and common genomic alterations that disrupt antigen processing and presentation. Broadly, antigen processing and presentation requires successful cleavage of proteins to appropriate size, loading of the antigen onto the MHC I molecule and trafficking of the complex to the cell membrane. Defects in this process have been documented as a common cellular feature allowing for tumor evasion of CTL recognition and subsequent killing22. Antigens are most commonly peptides of 8–10 amino acids generated via ubiquitination and degradation by the multi-catalytic proteasome complex23. Critical subunits of the multi-catalytic proteasome complex are IFNγ inducible24 and loss of response to IFNγ may downregulate this process15.
Next, antigens are assembled with MHC I and β2-microglobulin (B2M) in the endoplasmic reticulum. The MHC I peptide loading complex (PLC) is comprised of MHC I, the peptide transporter (TAP), the bridging factor tapasin, endoplasmic reticulum aminopeptidase and chaperones calreticulin, calnexin and ERp5725,26. Loss or down regulation of TAP1 results in a global decrease in MHC I cell surface expression17,24,27. Indeed, alterations in TAP1 expression occur in HNSCC at a rate of about 34%28 with downregulation of TAP1 and tapasin occurring in both primary and metastatic lesions29. Beyond the processing and loading machinery, issues may arise within the MHC I:B2M complex27. Two copies of the B2M gene are present in each cell and only one copy is needed for MHC I antigen presentation30; thus, loss of both functional B2M genes is required for loss of MHC I antigen presentation. Loss of both genes for this support protein renders tumor cells completed unresponsive to IFNγ stimulation of MHC I expression and unable to present antigens31,32.
Similarly, the absence of structurally sound MHC Class I molecules may arise due to total or partial loss of one copy of chromosome 6, which encodes for MHC I heavy chains or loss of MHC allospecificity33. Transcription of MHC I may be suppressed due to hypermethylation of DNA CpG islands, specifically of HLA-A, B and C promoter regions34, and histone hypoacetylation33. Transcription can be restored by DNA demethylating agents (5-azacytidine and decitabine) and histone deacetylases22. Thus, mutations or downregulation of either B2M or MHC I allows tumor cells to evade CTL recognition and elimination18. Clinically, loss of heterozygosity on chromosome 6p21, has been documented in HNSCC at a frequency of 36%24 and MHC class I downregulation is associated with clinical reduction in disease free interval and overall survival28. Finally, 80% of HNSCC are EGFR positive and EGFR-induced MHC I downregulation may contribute to resistance to T cell killing35.
2). Absent or defective IFNγ receptor expression or downstream signaling
Cytotoxic T cells express interferon-γ which induces upregulation of MHC I expression in tumor cells36. Binding of IFNγ to IFNγ receptor 1 and receptor 2 induces heterodimerization of the two components. Next, JAK2 phosphorylates the receptor allowing for binding of STAT1 proteins. STAT1 is phosphorylated, forms a homodimer and traffics to the nucleus25. Once in the nucleus, STAT1 interacts with interferon regulatory factor 1 (IRF-1) to activate a wide variety of cellular responses including specific and nonspecific mechanisms of defense against viral infection and malignant transformation37. Specifically, IFNγ is a major cytokine responsible for upregulation of MHC I, including induction of proteins required for MHC I:Antigen loading and processing such as TAP1, tapasin, calreticulin, calnexin and ERp5725. Disruption of any of these components can result in defective IFNγ signaling and a decrease in or absence of MHC I presentation on the tumor cell surface15–17.
Both HPV-positive and -negative HNSCC are enriched for somatic mutations in the IFNγ receptor38. Mutations in the JAK proteins have been widely documented in various cancer types, including melanoma39, and can produce a variety of phenotypes. Homozygous loss of function mutation in JAK1 leads to resistance to IFNγ signaling in addition to loss of IFNα and IFNβ response. In contrast, JAK2 mutations result in cells that are unresponsive to IFNγ only. Both mutations were documented in human melanoma lines and were correlated with initial tumor regression on immune checkpoint blockade with later disease progression40. In tumor lines with functional JAK proteins, IFNγ signaling can be constitutively inhibited by suppressor of cytokine signaling-3 (SOCS3). SOCS3 inhibits JAK-STAT signaling through prolonged and extensive phosphorylation of STAT1 at the S727 site thus inhibiting homodimerization translocation to the nucleus41. In contrast, Src homology 2 domain-containing phosphatase (SHP)-2 overexpression dephosphorylates pSTAH, also resulting in inhibited homodimerization and decreased expression of MHC I APM components42,43. When taken together, phosphorylation of STAT1 must be tightly regulated for proper functioning of the IFNγ induced JAK-STAT pathway and dysregulation results in abrogation of pathway function. Finally, within the nucleus the STAT1 complex interacts with interferon regulatory factor 1 (IRF-1) to activate IFN inducible genes. Methylation of IRF-1 prevents binding by STAT1 and transactivation of IFN inducible genes30. Encouragingly, demethylating agents have been shown to restore IFNγ induced MHC I expression44.
3). Defective formation of immunologic synapse
Immune cells communicate directly with each other via cell-cell contacts also known as the immune synapse. Cytotoxic cells of the immune system also use this synapse during direct attacks on infected and tumor cells45. Formation of the functional synapse requires the assembly of various structural components clustered around the central T-cell receptor, including the critical leukocyte function associated antigen-1 (LFA-1) which is activated when bound to intercellular adhesion molecule 1 (ICAM-1) on the antigen presenting cell46. This interaction is especially critical in antitumor immune function (8666900) and can activate CD8+ T-cells in the absence of classical costimulatory molecules such as CD80 and CD8647,48. It would then logically follow that down regulation of ICAM-1 would confer resistance to cytotoxic lymphocyte killing. Indeed, discovery of this key interaction was first described in melanoma cell lines and multiple groups have shown that ICAM-1 expression enhances in vivo lymphocyte adhesion in murine fibrosarcoma models49,50 and in breast, gastrointestinal and hematologic malignancies51–53. However, establishing patterns of ICAM-1 expression in head and neck cancer has proved difficult with varying expression levels among patient samples and between cancer types54,55. Patients with nasopharyngeal carcinoma and adenoid cystic carcinoma of the head and neck were demonstrated to have elevated soluble ICAM-1 in their sera as compared with normal controls54,56. In the case of nasopharyngeal carcinoma, increased levels of soluble ICAM-1 were correlated with recurrence and poor prognosis57 but in adenoid cystic carcinoma ICAM-1 downregulation was found to coexist with increased distant metastasis and poor prognosis56. Thus, from a mechanistic stance down regulation of ICAM could confer resistance to CTL killing but in vitro evidence is lacking, and clinical correlates have yet to be fully elucidated.
4). Neutralization of granzyme release from T cells
After successful recognition and binding of the TCR to MHC I, T-cells can induce apoptosis of cancer cells via the granule exocytosis pathway, important in early onset target cell apoptosis20,21,58. Activated lymphocytes secrete perforin, a membrane permeability protein, and granzymes, proteolytic enzymes that activate caspases59, from cytotoxic granules toward the target cells. It is not fully understood whether release of perforin into the immunosynapse forms pores directly on the tumor plasma membrane to allow the entry of granzymes into the tumor cell or if the granules enter target tumors cells through receptor-mediated endocytosis followed by the subsequent perforin-dependent release of granzyme content from the endosome into tumor cytosol60. It is known, however, that calcium-dependent polymerization of perforin leads to loss of plasma osmotic stability and leads to the channel-like pore formation61. After pore formation, granzyme B is the main effector enzyme. Granzyme B causes target cell apoptosis directly through cleavage of BID, a member of the pro-apoptotic BCL-2 family62,63 resulting in cytochrome c release. Thus, granzyme B induced apoptosis acts in a caspase independent manner64.
Tumor cells may be resistant to granzyme mediated apoptosis due to expression of the serine protease inhibitor (serpin) PI-9, or the murine counterpart SPI-6, which inactivates the effects of Granzyme B59. Normally, PI-9 is expressed in immune privileged sites, such as in lymphoid tissue, on T lymphocytes, and on NK cells, possibly as a mechanism to inactivate immune cell self-destruction by Granzyme B65,66. PI-6 is expressed in human melanoma, breast, cervical, and colon carcinomas and, notably, is not detectable in normal melanocytes, breast or cervical epithelium, or colon tissue66. PI-9 expression on tumor cells is clinically associated with poorer outcomes in melanoma patients with metastatic disease67.
5). Resistance to death receptor-induced extrinsic apoptosis
Late onset apoptosis mediated by T cells is largely induced through “death receptors” activated by tumor necrosis factor (TNF) receptor superfamily ligands such as Fas receptor (CD95/Apo-1) and death receptor (DR) 4 or 5, the receptors for tumor necrosis factor related apoptosis inducing ligand (TRAIL)21,68,69. Specifically, T cells express surface FAS ligand (FasL or CD95L) which binds to Fas on cancer cells. Ligand binding to Fas leads to the recruitment of Fas-associated death domain (FADD) and procaspase-8 to form the death inducing signaling complex (DISC), which in turn recruits and cleaves procaspases to form active caspases resulting in cell apoptosis21,70.
Fas is expressed in normal squamous epithelia, including gingival and basal cell layers. Malignant cells of head and neck cancer have variable expression of both Fas and FasL70. In tobacco related intraoral carcinoma, for example, tumor cells downregulate Fas receptor and upregulate FasL, which confers not only resistance to CTL killing but provides a mechanism of tumor induced killing of anti-tumor immune cells that also express the Fas receptor. FasL expression is high in poorly differentiated oral squamous cell carcinoma and negatively correlated with the degree of apoptosis in these tumors70.
The main mechanisms of tumor-intrinsic resistance to Fas -mediated apoptosis are overexpression of c-FLIP and expression of decoy receptors. c-FLIP, or cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein, is a relatively proximal anti-apoptotic protein that interacts with the Fas induced apoptosis pathway71. c-FLIP resembles procaspase-8 but lacks the catalytic site required for downstream death-inducing signaling complex formation and completion of apoptosis. An increased ratio of c-FLIP to caspase-8 expression is associated with increased resistance to Fas-mediated apoptosis72. Clinically, c-FLIP overexpression associated with higher clinical stage and lymph node metastases specifically in HNSCC73. Cells can be re-sensitized to Fas mediated apoptosis by decreasing or silencing c-FLIP providing and possible therapeutic target. Silencing of c-FLIP utilizing the small molecule inhibitor MLN4924 has been pre-clinically studied in HNSCC74 and melanoma72,75, and clinical trials across a variety of cancer types are in progress. In oral squamous cell carcinoma, the PI3/AKT signaling cascade can enhance Fas-mediated apoptosis through downregulation of cFLIP76; however therapeutic benefits of this interaction have yet to be explored.
Notably, c-FLIP overexpression does not inhibit granzyme B-mediated apoptosis77, but in vivo c-FLIP overexpression does result in tumor immune escape, suggesting that evasion Fas-dependent apoptosis may be a more influential mechanism for in vivo tumor immune escape than Granzyme B-mediated apoptosis78,79. In a mouse model of viral FLIP (v-FLIP), tumor immune escape only occurred in immune competent mice and the effect of v-FLIP was not seen in mice lacking B and T cells further supporting the hypothesis that FLIPs effects on Fas-mediated apoptosis is directly related to cytotoxic lymphocyte killing80. Finally, the possibility exists that c-FLIP’s overall impact on cancer severity is related to c-FLIP’s activating effects on the NFκB, MAPK, and ERK pathways71,81,82 in addition to increased resistance to CTL killing; however, these effects require further study.
Decoy receptors are members of the TNF receptor family that morphologically mimic true death receptors but lack a functional death domain79,83. As a result, they competitively bind to FasL, and prevent Fas death receptor-induced apoptosis. Soluble Fas (sCD95) and decoy receptor 3 (DcR3) are two decoy receptors that prevent FasL binding83,84. sCD95 is expressed in many cancer types, including melanoma85 and is specifically associated with a worse prognosis in melanoma86. DcR3 expression and function has been described in several cancer types87. Tumor necrosis factor related apoptosis inducing ligand (TRAIL) is another member of the TNF superfamily expressed by cytotoxic lymphocytes that can initiate apoptosis by binding to its associated death receptors DR4 and DR569. These receptors induce apoptosis via the same mechanisms as the Fas mediated pathway; however, TRAIL mediated signaling can also induce cleavage of the proapoptotic BH3 protein Bid creating the active, truncated Bid (tBid), which in turn interacts with Bax and Bak at the mitochondrial membrane to release apoptotic factors, including cytochrome c69,88.
HNSCC cells demonstrate resistance to TRAIL-death receptor mediated apoptosis via a variety of mechanisms. Beginning at the receptor site, resistance may be conferred by polymorphisms in DR4 resulting in loss of receptor function due to dominant negative expression of the mutated protein. Down regulation of DR5 receptor expression via histone deacetylation also confers resistance to CTL mediated killing89. Malignant cells may also express TRAIL receptors DcR-1 and DcR-2, decoy receptors that do not possess an intracellular death domain and cannot form signaling complexes to induce apoptosis. These receptors have been documented in several cancer types including HNSCC90.
6). Caspase Mutations
Caspase 8 is involved death receptor mediated mechanisms of cytotoxic T-lymphocyte killing and, to a lesser extent, granzyme B mediated apoptosis91. Caspase 8 mutations are associated with tumor resistance in many cancer types, including head and neck cancers92,93. Caspase 8’s critical role in the extrinsic apoptosis pathway likely means that mutated Caspase 8 results in a global decrease in extrinsic apoptosis pathway activation. In HNSCC, mutated procaspase-8 can be constitutively bound to FADD and can impair recruitment of DISC components. Finally, tumors bearing caspase 8 mutations are further associated with enhanced tumor migration, invasion, and growth93.
7). Intrinsic cell cycle alterations in response to cytotoxic insults
Many tumors including HNSCC harbor mutations in proteins such as p53, p21 and Rb that affect the G1/S cell cycle checkpoint94,95. As a result, cancer cells become more dependent upon the G2/M cell cycle checkpoint for cellular and DNA repair after exposure to cytotoxic or genotoxic insults. This gives cells time to repair injury before segregation of damaged DNA during mitosis. Cell cycle pause after insult has been a recognized phenomenon following chemotherapy and ionizing radiation for some time96. More recent work has demonstrated that granzyme B and TNF-α similarly activate the G2/M cell cycle DNA damage checkpoint during early and late phases of T cell killing97–99. This intrinsic mechanism of resisting damage and induction of apoptosis reduces tumor cell susceptibility to T cell killing. Reversal of G2/M cell cycle arrest via Wee1 kinase inhibition makes them more susceptible to in vitro and in vivo T-cell killing in multiple pre-clinical models including models of HNSCC99,100.
Clinical repercussions of altered tumor cell immunogenicity
In tumors composed of cells that harbor intact IFN responses and T cell recognition pathways, lack of development of T cell responses to immunorecessive antigens may be overcome with immunotherapies such as immune checkpoint blockade101. However, the mechanisms of altered tumor cell immunogenicity described above suggest that a tumor cell could evade T cell recognition and destruction despite harboring one or more T cell antigens. Accordingly, recent genomic studies exploring predictors of response to immune checkpoint inhibitors have demonstrated tumors harboring alterations in IFNγ response genes and genes encoding for JAK1/2 and β2M do not respond to treatment39,40,102,103 If defects in immunogenicity are detected within a tumor, what other immunotherapy approaches could be effective? It likely depends upon which pathways are altered. If a tumor harbors defects in T cell antigen processing and presentation, defective immune synapse formation or response to IFN, these primarily alter TCR recognition of intracellular antigen presented via MHC I. Thus, immunotherapy that does not rely on TCR signaling such as NK cellular therapies (which are MHC and antigen independent) or chimeric antigen receptor (CAR) engineered T cellular therapies (which recognize a cell surface epitope similar to an antibody) may be effective. Conversely, if tumors harbor defects in responses to granzymes or TNFR superfamily ligands (FasL, TRAIL) or caspase mutations, then any form of immunotherapy is likely to be ineffective since both T and NK cellular therapeutics ultimately use the same mechanisms to destroy a tumor cell. In these situations, standard anti-cancer approaches such as surgical resection or localized ionizing radiation may be the only rational approach. Ultimately, integration of genomic characterization of tumors with different T and NK cell-based immunotherapy approaches is needed to determine if identification of such alterations predicts responses. If they do, this would be a major advance toward the ability to personalize treatment approaches for patients with HNSCC.
Conclusions
Recent advances in cancer immunotherapies have indeed provided exciting therapeutic options for HNSCC patients; however, current FDA-approved therapies stimulate endogenous immunity being blocked by immune checkpoints only. This can certainly benefit som e patients. Yet, many tumors harbor cells with defects in antigen processing and presentation, responses to IFN, and direct susceptibility to T cell killing through granzymes and TNFR superfamily ligands. As a result, some tumors may harbor a population of cells that simply cannot be detected and/or destroyed by T cells. Only through complete genomic characterization of tumors and a thorough understanding of how immune cells detect and eliminate tumor cells can we hope to prospectively assign specific immunotherapy or standard anti-cancer therapy approaches to individual patients in a personalized fashion. As more immunotherapy tools are developed that are independent of T cells, such as NK cellular therapies, integration of genomic characterization into prospective trials to determine which tumor genomic alterations predict responses to which immunotherapy becomes critical. Such ability is a major goal of the immunotherapy research community in order to minimize treatment of cancer patients with treatment approaches that have little to no chance of patient benefit.
Highlights.
Tumor cells presenting immunodominant antigens may be eliminated by immunity
Tumor mutations may make other tumor cells invisible to T cells
Both of these processes lead to selection of tumor cells that escape immunity
Recognition of these tumor subpopulations may guide treatment in the future
Acknowledgments
Grant support
This work was supported by the Intramural Research Program of the NIH, NIDCD, project number ZIA-DC000087.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Meetings presented
None
Conflict of Interest Statement
The authors have no conflicts of interest to disclose
References
- 1.Nikolich-Zugich J, Slifka MK, Messaoudi I. The many important facets of T-cell repertoire diversity. Nat Rev Immunol. 2004;4(2):123–132. [DOI] [PubMed] [Google Scholar]
- 2.Lawrence MS, Stojanov P, Polak P, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499(7457):214–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69–74. [DOI] [PubMed] [Google Scholar]
- 4.Van Allen EM, Miao D, Schilling B, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350(6257):207–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Davis RJ, Van Waes C, Allen CT. Overcoming barriers to effective immunotherapy: MDSCs, TAMs, and Tregs as mediators of the immunosuppressive microenvironment in head and neck cancer. Oral Oncol. 2016;58:59–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Curry JM, Sprandio J, Cognetti D, et al. Tumor microenvironment in head and neck squamous cell carcinoma. Semin Oncol. 2014;41(2):217–234. [DOI] [PubMed] [Google Scholar]
- 7.Hadrup S, Donia M, Thor Straten P. Effector CD4 and CD8 T cells and their role in the tumor microenvironment. Cancer Microenviron. 2013;6(2):123–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Greaves M Evolutionary determinants of cancer. Cancer Discov. 2015;5(8):806–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mroz EA, Tward AD, Pickering CR, Myers JN, Ferris RL, Rocco JW. High intratumor genetic heterogeneity is related to worse outcome in patients with head and neck squamous cell carcinoma. Cancer. 2013;119(16):3034–3042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Urban JL, Kripke ML, Schreiber H. Stepwise immunologic selection of antigenic variants during tumor growth. J Immunol. 1986;137(9):3036–3041. [PubMed] [Google Scholar]
- 11.Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006;6(11):836–848. [DOI] [PubMed] [Google Scholar]
- 12.Sandberg JK, Grufman P, Wolpert EZ, Franksson L, Chambers BJ, Karre K. Superdominance among immunodominant H-2Kb-restricted epitopes and reversal by dendritic cell-mediated antigen delivery. J Immunol. 1998;160(7):3163–3169. [PubMed] [Google Scholar]
- 13.Van Waes C, Monach PA, Urban JL, Wortzel RD, Schreiber H. Immunodominance deters the response to other tumor antigens thereby favoring escape: prevention by vaccination with tumor variants selected with cloned cytolytic T cells in vitro. Tissue Antigens. 1996;47(5):399–407. [DOI] [PubMed] [Google Scholar]
- 14.Seung S, Urban JL, Schreiber H. A tumor escape variant that has lost one major histocompatibility complex class I restriction element induces specific CD8+ T cells to an antigen that no longer serves as a target. J Exp Med. 1993;178(3):933–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Katlinski KV, Gui J, Katlinskaya YV, et al. Inactivation of Interferon Receptor Promotes the Establishment of Immune Privileged Tumor Microenvironment. Cancer Cell. 2017;31(2):194–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Albacker LA, Wu J, Smith P, et al. Loss of function JAK1 mutations occur at high frequency in cancers with microsatellite instability and are suggestive of immune evasion. PLoS One. 2017;12(11):e0176181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Johnsen AK, Templeton DJ, Sy M, Harding CV. Deficiency of transporter for antigen presentation (TAP) in tumor cells allows evasion of immune surveillance and increases tumorigenesis. J Immunol. 1999;163(8):4224–4231. [PubMed] [Google Scholar]
- 18.McGranahan N, Rosenthal R, Hiley CT, et al. Allele-Specific HLA Loss and Immune Escape in Lung Cancer Evolution. Cell. 2017;171(6):1259–1271 e1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Raval A, Puri N, Rath PC, Saxena RK. Cytokine regulation of expression of class I MHC antigens. Exp Mol Med. 1998;30(1):1–13. [DOI] [PubMed] [Google Scholar]
- 20.Heusel JW, Wesselschmidt RL, Shresta S, Russell JH, Ley TJ. Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells. Cell. 1994;76(6):977–987. [DOI] [PubMed] [Google Scholar]
- 21.Shresta S, Pham CT, Thomas DA, Graubert TA, Ley TJ. How do cytotoxic lymphocytes kill their targets? Curr Opin Immunol. 1998;10(5):581–587. [DOI] [PubMed] [Google Scholar]
- 22.Lampen MH, van Hall T. Strategies to counteract MHC-I defects in tumors. Curr Opin Immunol. 2011;23(2):293–298. [DOI] [PubMed] [Google Scholar]
- 23.Madden DR. The three-dimensional structure of peptide-MHC complexes. Annu Rev Immunol. 1995;13:587–622. [DOI] [PubMed] [Google Scholar]
- 24.Seliger B Molecular mechanisms of MHC class I abnormalities and APM components in human tumors. Cancer Immunol Immunother. 2008;57(11):1719–1726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hansen TH, Bouvier M. MHC class I antigen presentation: learning from viral evasion strategies. Nat Rev Immunol. 2009;9(7):503–513. [DOI] [PubMed] [Google Scholar]
- 26.Kalu DN, Foster GV. Calcitonin. Its physiologic roles. N Y State J Med. 1976;76(2):230–233. [PubMed] [Google Scholar]
- 27.Campoli M, Chang CC, Ferrone S. HLA class I antigen loss, tumor immune escape and immune selection. Vaccine. 2002;20 Suppl 4:A40–45. [DOI] [PubMed] [Google Scholar]
- 28.Chang CC, Campoli M, Ferrone S. Classical and nonclassical HLA class I antigen and NK Cell-activating ligand changes in malignant cells: current challenges and future directions. Adv Cancer Res. 2005;93:189–234. [DOI] [PubMed] [Google Scholar]
- 29.Bandoh N, Ogino T, Katayama A, et al. HLA class I antigen and transporter associated with antigen processing downregulation in metastatic lesions of head and neck squamous cell carcinoma as a marker of poor prognosis. Oncol Rep. 2010;23(4):933–939. [DOI] [PubMed] [Google Scholar]
- 30.Seliger B, Ritz U, Abele R, et al. Immune escape of melanoma: first evidence of structural alterations in two distinct components of the MHC class I antigen processing pathway. Cancer Res. 2001;61(24):8647–8650. [PubMed] [Google Scholar]
- 31.D’Urso CM, Wang ZG, Cao Y, Tatake R, Zeff RA, Ferrone S. Lack of HLA class I antigen expression by cultured melanoma cells FO-1 due to a defect in B2m gene expression. J Clin Invest. 1991;87(1):284–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sucker A, Zhao F, Real B, et al. Genetic evolution of T-cell resistance in the course of melanoma progression. Clin Cancer Res. 2014;20(24):6593–6604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Campoli M, Ferrone S. HLA antigen changes in malignant cells: epigenetic mechanisms and biologic significance. Oncogene. 2008;27(45):5869–5885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Nie Y, Yang G, Song Y, et al. DNA hypermethylation is a mechanism for loss of expression of the HLA class I genes in human esophageal squamous cell carcinomas. Carcinogenesis. 2001;22(10):1615–1623. [DOI] [PubMed] [Google Scholar]
- 35.Pollack BP, Sapkota B, Cartee TV. Epidermal growth factor receptor inhibition augments the expression of MHC class I and II genes. Clin Cancer Res. 2011;17(13):4400–4413. [DOI] [PubMed] [Google Scholar]
- 36.Tau GZ, Cowan SN, Weisburg J, Braunstein NS, Rothman PB. Regulation of IFN-gamma signaling is essential for the cytotoxic activity of CD8(+) T cells. J Immunol. 2001;167(10):5574–5582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bach EA, Aguet M, Schreiber RD. The IFN gamma receptor: a paradigm for cytokine receptor signaling. Annu Rev Immunol. 1997;15:563–591. [DOI] [PubMed] [Google Scholar]
- 38.Gillison ML, Akagi K, Xiao W, et al. Human papillomavirus and the landscape of secondary genetic alterations in oral cancers. Genome Res. 2019;29(1):1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Shin DS, Zaretsky JM, Escuin-Ordinas H, et al. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov. 2017;7(2):188–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med. 2016;375(9):819–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kovarik A, Fojtova M, Boudny V, Adamkova L, Lauerova L, Kovarik J. Interferon-gamma, but not interferon-alpha, induces SOCS 3 expression in human melanoma cell lines. Melanoma Res. 2005;15(6):481–488. [DOI] [PubMed] [Google Scholar]
- 42.Leibowitz MS, Srivastava RM, Andrade Filho PA, et al. SHP2 is overexpressed and inhibits pSTAT1-mediated APM component expression, T-cell attracting chemokine secretion, and CTL recognition in head and neck cancer cells. Clin Cancer Res. 2013;19(4):798–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Agazie YM, Hayman MJ. Molecular mechanism for a role of SHP2 in epidermal growth factor receptor signaling. Mol Cell Biol. 2003;23(21):7875–7886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Rodriguez T, Mendez R, Del Campo A, et al. Distinct mechanisms of loss of IFN-gamma mediated HLA class I inducibility in two melanoma cell lines. BMC Cancer. 2007;7:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Dieckmann NM, Frazer GL, Asano Y, Stinchcombe JC, Griffiths GM. The cytotoxic T lymphocyte immune synapse at a glance. J Cell Sci. 2016;129(15):2881–2886. [DOI] [PubMed] [Google Scholar]
- 46.Braakman E, Goedegebuure PS, Vreugdenhil RJ, Segal DM, Shaw S, Bolhuis RL. ICAM-melanoma cells are relatively resistant to CD3-mediated T-cell lysis. Int J Cancer. 1990;46(3):475–480. [DOI] [PubMed] [Google Scholar]
- 47.de Waal Malefyt R, Verma S, Bejarano MT, Ranes-Goldberg M, Hill M, Spits H. CD2/LFA-3 or LFA-1/ICAM-1 but not CD28/B7 interactions can augment cytotoxicity by virus-specific CD8+ cytotoxic T lymphocytes. Eur J Immunol. 1993;23(2):418–424. [DOI] [PubMed] [Google Scholar]
- 48.Jenkinson SR, Williams NA, Morgan DJ. The role of intercellular adhesion molecule-1/LFA-1 interactions in the generation of tumor-specific CD8+ T cell responses. J Immunol. 2005;174(6):3401–3407. [DOI] [PubMed] [Google Scholar]
- 49.Turner J, Rhee JG, Fabian DF, Lefor AT. Expression of ICAM-1 enhances in vivo lymphocyte adhesion in a murine fibrosarcoma. J Surg Oncol. 1997;66(1):39–44. [DOI] [PubMed] [Google Scholar]
- 50.Sartor WM, Kyprianou N, Fabian DF, Lefor AT. Enhanced expression of ICAM-1 in a murine fibrosarcoma reduces tumor growth rate. J Surg Res. 1995;59(1):66–74. [DOI] [PubMed] [Google Scholar]
- 51.Rosette C, Roth RB, Oeth P, et al. Role of ICAM1 in invasion of human breast cancer cells. Carcinogenesis. 2005;26(5):943–950. [DOI] [PubMed] [Google Scholar]
- 52.Mizoi T, Ohtani H, Suzuki Y, Shiiba K, Matsuno S, Nagura H. Intercellular adhesion molecule-1 expression by macrophages in human gastrointestinal carcinoma: possible roles as host immune/inflammatory reaction. Pathol Int. 1995;45(8):565–572. [DOI] [PubMed] [Google Scholar]
- 53.Terol MJ, Tormo M, Martinez-Climent JA, et al. Soluble intercellular adhesion molecule-1 (s-ICAM-1/s-CD54) in diffuse large B-cell lymphoma: association with clinical characteristics and outcome. Ann Oncol. 2003;14(3):467–474. [DOI] [PubMed] [Google Scholar]
- 54.Liu CM, Sheen TS, Ko JY, Shun CT. Circulating intercellular adhesion molecule 1 (ICAM-1), E-selectin and vascular cell adhesion molecule 1 (VCAM-1) in head and neck cancer. Br J Cancer. 1999;79(2):360–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Akcay F, Taysi S, Uslu C, Dogru Y, Gumustekin K. Levels of soluble intercellular adhesion molecule-1 and total sialic acid in serum of patients with laryngeal cancer. Jpn J Clin Oncol. 2001;31(12):584–588. [DOI] [PubMed] [Google Scholar]
- 56.Shirai A, Furukawa M, Yoshizaki T. Expression of intercellular adhesion molecule (ICAM)-1 in adenoid cystic carcinoma of the head and neck. Laryngoscope. 2003;113(11):1955–1960. [DOI] [PubMed] [Google Scholar]
- 57.Yu Y, Dong W, Zhou X, Li S. The significance of serum soluble intercellular adhesion molecule 1 and transforming growth factor alpha in patients with nasopharyngeal carcinoma. Arch Otolaryngol Head Neck Surg. 2004;130(10):1205–1208. [DOI] [PubMed] [Google Scholar]
- 58.Kawaguchi Y, Kono K, Mizukami Y, Mimura K, Fujii H. Mechanisms of escape from trastuzumab-mediated ADCC in esophageal squamous cell carcinoma: relation to susceptibility to perforin-granzyme. Anticancer Res. 2009;29(6):2137–2146. [PubMed] [Google Scholar]
- 59.Medema JP, de Jong J, Peltenburg LT, et al. Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI-9/SPI-6 constitutes a mechanism for immune escape by tumors. Proc Natl Acad Sci U S A. 2001;98(20):11515–11520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Spicer BA, Conroy PJ, Law RHP, Voskoboinik I, Whisstock JC. Perforin-A key (shaped) weapon in the immunological arsenal. Semin Cell Dev Biol. 2017;72:117–123. [DOI] [PubMed] [Google Scholar]
- 61.Motyka B, Korbutt G, Pinkoski MJ, et al. Mannose 6-phosphate/insulin-like growth factor II receptor is a death receptor for granzyme B during cytotoxic T cell-induced apoptosis. Cell. 2000;103(3):491–500. [DOI] [PubMed] [Google Scholar]
- 62.Heibein JA, Goping IS, Barry M, et al. Granzyme B-mediated cytochrome c release is regulated by the Bcl-2 family members bid and Bax. J Exp Med. 2000;192(10):1391–1402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Pinkoski MJ, Waterhouse NJ, Heibein JA, et al. Granzyme B-mediated apoptosis proceeds predominantly through a Bcl-2-inhibitable mitochondrial pathway. J Biol Chem. 2001;276(15):12060–12067. [DOI] [PubMed] [Google Scholar]
- 64.Barry M, Heibein JA, Pinkoski MJ, et al. Granzyme B short-circuits the need for caspase 8 activity during granule-mediated cytotoxic T-lymphocyte killing by directly cleaving Bid. Mol Cell Biol. 2000;20(11):3781–3794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sun J, Bird CH, Sutton V, et al. A cytosolic granzyme B inhibitor related to the viral apoptotic regulator cytokine response modifier A is present in cytotoxic lymphocytes. J Biol Chem. 1996;271(44):27802–27809. [DOI] [PubMed] [Google Scholar]
- 66.Bladergroen BA, Strik MC, Bovenschen N, et al. The granzyme B inhibitor, protease inhibitor 9, is mainly expressed by dendritic cells and at immune-privileged sites. J Immunol. 2001;166(5):3218–3225. [DOI] [PubMed] [Google Scholar]
- 67.van Houdt IS, Oudejans JJ, van den Eertwegh AJ, et al. Expression of the apoptosis inhibitor protease inhibitor 9 predicts clinical outcome in vaccinated patients with stage III and IV melanoma. Clin Cancer Res. 2005;11(17):6400–6407. [DOI] [PubMed] [Google Scholar]
- 68.Krammer PH. CD95’s deadly mission in the immune system. Nature. 2000;407(6805):789–795. [DOI] [PubMed] [Google Scholar]
- 69.Yuan X, Gajan A, Chu Q, Xiong H, Wu K, Wu GS. Developing TRAIL/TRAIL death receptor-based cancer therapies. Cancer Metastasis Rev. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Loro LL, Vintermyr OK, Johannessen AC, Liavaag PG, Jonsson R. Suppression of Fas receptor and negative correlation of Fas ligand with differentiation and apoptosis in oral squamous cell carcinoma. J Oral Pathol Med. 1999;28(2):82–87. [DOI] [PubMed] [Google Scholar]
- 71.Safa AR. Roles of c-FLIP in Apoptosis, Necroptosis, and Autophagy. J Carcinog Mutagen. 2013;Suppl 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Griffith TS, Chin WA, Jackson GC, Lynch DH, Kubin MZ. Intracellular regulation of TRAIL-induced apoptosis in human melanoma cells. J Immunol. 1998;161(6):2833–2840. [PubMed] [Google Scholar]
- 73.Li X, Pan X, Zhang H, et al. Overexpression of cFLIP in head and neck squamous cell carcinoma and its clinicopathologic correlations. J Cancer Res Clin Oncol. 2008;134(5):609–615. [DOI] [PubMed] [Google Scholar]
- 74.Zhao L, Yue P, Lonial S, Khuri FR, Sun SY. The NEDD8-activating enzyme inhibitor, MLN4924, cooperates with TRAIL to augment apoptosis through facilitating c-FLIP degradation in head and neck cancer cells. Mol Cancer Ther. 2011;10(12):2415–2425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Irmler M, Thome M, Hahne M, et al. Inhibition of death receptor signals by cellular FLIP. Nature. 1997;388(6638):190–195. [DOI] [PubMed] [Google Scholar]
- 76.Kondo G, Iwase M, Watanabe H, et al. Enhancement of susceptibility to Fas-mediated apoptosis in oral squamous cell carcinoma cells by phosphatidylinositol 3-kinase inhibitor. Oral Oncol. 2006;42(7):745–752. [DOI] [PubMed] [Google Scholar]
- 77.Kataoka T, Schroter M, Hahne M, et al. FLIP prevents apoptosis induced by death receptors but not by perforin/granzyme B, chemotherapeutic drugs, and gamma irradiation. J Immunol. 1998;161(8):3936–3942. [PubMed] [Google Scholar]
- 78.Medema JP, de Jong J, van Hall T, Melief CJ, Offringa R. Immune escape of tumors in vivo by expression of cellular FLICE-inhibitory protein. J Exp Med. 1999;190(7):1033–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Igney FH, Krammer PH. Death and anti-death: tumour resistance to apoptosis. Nat Rev Cancer. 2002;2(4):277–288. [DOI] [PubMed] [Google Scholar]
- 80.Djerbi M, Screpanti V, Catrina AI, Bogen B, Biberfeld P, Grandien A. The inhibitor of death receptor signaling, FLICE-inhibitory protein defines a new class of tumor progression factors. J Exp Med. 1999;190(7):1025–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kataoka T, Budd RC, Holler N, et al. The caspase-8 inhibitor FLIP promotes activation of NF-kappaB and Erk signaling pathways. Curr Biol. 2000;10(11):640–648. [DOI] [PubMed] [Google Scholar]
- 82.Hu WH, Johnson H, Shu HB. Activation of NF-kappaB by FADD, Casper, and caspase-8. J Biol Chem. 2000;275(15):10838–10844. [DOI] [PubMed] [Google Scholar]
- 83.Ashkenazi A Targeting death and decoy receptors of the tumour-necrosis factor superfamily. Nat Rev Cancer. 2002;2(6):420–430. [DOI] [PubMed] [Google Scholar]
- 84.Topfer K, Kempe S, Muller N, et al. Tumor evasion from T cell surveillance. J Biomed Biotechnol. 2011;2011:918471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Midis GP, Shen Y, Owen-Schaub LB. Elevated soluble Fas (sFas) levels in nonhematopoietic human malignancy. Cancer Res. 1996;56(17):3870–3874. [PubMed] [Google Scholar]
- 86.Ugurel S, Rappl G, Tilgen W, Reinhold U. Increased soluble CD95 (sFas/CD95) serum level correlates with poor prognosis in melanoma patients. Clin Cancer Res. 2001;7(5):1282–1286. [PubMed] [Google Scholar]
- 87.Roth W, Isenmann S, Nakamura M, et al. Soluble decoy receptor 3 is expressed by malignant gliomas and suppresses CD95 ligand-induced apoptosis and chemotaxis. Cancer Res. 2001;61(6):2759–2765. [PubMed] [Google Scholar]
- 88.Hussein MR, Haemel AK, Wood GS. Apoptosis and melanoma: molecular mechanisms. J Pathol. 2003;199(3):275–288. [DOI] [PubMed] [Google Scholar]
- 89.Jazirehi AR, Kurdistani SK, Economou JS. Histone deacetylase inhibitor sensitizes apoptosis-resistant melanomas to cytotoxic human T lymphocytes through regulation of TRAIL/DR5 pathway. J Immunol. 2014;192(8):3981–3989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Nguyen PT, Nguyen D, Chea C, Miyauchi M, Fujii M, Takata T. Interaction between N-cadherin and decoy receptor-2 regulates apoptosis in head and neck cancer. Oncotarget. 2018;9(59):31516–31530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Medema JP, Toes RE, Scaffidi C, et al. Cleavage of FLICE (caspase-8) by granzyme B during cytotoxic T lymphocyte-induced apoptosis. Eur J Immunol. 1997;27(12):3492–3498. [DOI] [PubMed] [Google Scholar]
- 92.Mandruzzato S, Brasseur F, Andry G, Boon T, van der Bruggen P. A CASP-8 mutation recognized by cytolytic T lymphocytes on a human head and neck carcinoma. J Exp Med. 1997;186(5):785–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Li C, Egloff AM, Sen M, Grandis JR, Johnson DE. Caspase-8 mutations in head and neck cancer confer resistance to death receptor-mediated apoptosis and enhance migration, invasion, and tumor growth. Mol Oncol. 2014;8(7):1220–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Shaw PH. The role of p53 in cell cycle regulation. Pathol Res Pract. 1996;192(7):669–675. [DOI] [PubMed] [Google Scholar]
- 95.Cancer Genome Atlas N Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015;517(7536):576–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Fu S, Wang Y, Keyomarsi K, Meric-Bernstein F. Strategic development of AZD1775, a Wee1 kinase inhibitor, for cancer therapy. Expert Opin InvestigDrugs. 2018;27(9):741–751. [DOI] [PubMed] [Google Scholar]
- 97.Chen G, Shi L, Litchfield DW, Greenberg AH. Rescue from granzyme B-induced apoptosis by Wee1 kinase. J Exp Med. 1995;181(6):2295–2300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Darzynkiewicz Z, Williamson B, Carswell EA, Old LJ. Cell cycle-specific effects of tumor necrosis factor. Cancer Res. 1984;44(1):83–90. [PubMed] [Google Scholar]
- 99.Sun L, Moore E, Berman R, et al. WEE1 kinase inhibition reverses G2/M cell cycle checkpoint activation to sensitize cancer cells to immunotherapy. Oncoimmunology. 2018;7(10):e1488359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Friedman J, Morisada M, Sun L, et al. Inhibition of WEE1 kinase and cell cycle checkpoint activation sensitizes head and neck cancers to natural killer cell therapies. J Immunother Cancer. 2018;6(1):59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Memarnejadian A, Meilleur CE, Shaler CR, et al. PD-1 Blockade Promotes Epitope Spreading in Anticancer CD8(+) T Cell Responses by Preventing Fratricidal Death of Subdominant Clones To Relieve Immunodomination. J Immunol. 2017;199(9):3348–3359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ayers M, Lunceford J, Nebozhyn M, et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017;127(8):2930–2940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Gao J, Shi LZ, Zhao H, et al. Loss of IFN-gamma Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell. 2016;167(2):397–404 e399. [DOI] [PMC free article] [PubMed] [Google Scholar]