scholarly journals Immune pressures drive the promoter hypermethylation of neoantigen genes

Author(s):  
Ming Yi ◽  
Bing Dong ◽  
Qian Chu ◽  
Kongming Wu

AbstractCancer cells with strong immunogenicity are susceptible for elimination by cancer immunoediting, while the subpopulations with weak immunogenicity survive. As a result, a subset of cancer cells evade the immune attack and evolve into overt clinical lesions. During cancer evolution, it has been well established that multiple alterations such as the dysfunction of antigen presentation machinery and the upregulation of immunosuppressive signals (e.g. PD-L1) play important roles in immune escape. Recently, promoter hypermethylation of neoantigen genes has been proposed to be a vital mechanism of immunoediting. This epigenetically mediated immune evasion enriches the mechanisms of carcinogenesis.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 603-603
Author(s):  
Simona Pagliuca ◽  
Carmelo Gurnari ◽  
Colin Hercus ◽  
Niroshan Nadarajah ◽  
Adam Wahida ◽  
...  

Abstract The pathogenesis of idiopathic aplastic anemia (IAA) involves a human leukocyte antigen (HLA)-restricted T-cell autoreactivity against unknown antigens preferentially distributed on early hematopoietic stem and progenitor cells (HSPCs). Genetically acquired GPI-anchor and HLA deficiency have been both linked to clonal immune evasion from T-cell pressure. We hypothesized that, in analogy to anti-tumor adaptive immune evasion, pathophysiology of immune escape in IAA originates together with a broader dysfunction of antigen presentation/processing machinery and immune regulatory proteins, beyond HLA molecules, as an effect of immune pressure under T-cell attack. This initial immune reaction would produce up-modulation of these pathways, ultimately promoting the acquisition of mutations and expansion of immune resistant clones. To test this hypothesis, we first performed single-cell RNAseq analysis in HSPCs in IAA patients at disease manifestation, 1 which showed signatures of dysfunction of antigen presentation machinery, with up-regulation of most of the HLA molecules, proteasome subunits and endoplasmic reticulum related organelle transporters. Strikingly, DRB1 was among the top 3 genes upregulated in IAA patients compared to controls (q-values 1.23E-35; Fig.1A), underscoring the etiological impact that antigen presentation via this locus has in the initiation of autoimmune process. Mild upregulation was also seen in DQB1 and B loci (q-values 4.7E-07 and 2.1E-10, respectively). We then studied molecular escape mechanisms by genotyping 204 IAA and PNH patients, with either a targeted or whole genome sequencing (WGS) platform. By application of a newly in-house developed bioinformatic pipeline, we detected somatic aberrations in HLA region involving both class I and II alleles in 36% of IAA patients including point mutations, frameshift insertions or deletions and copy number variations inducing allelic loss. B*14:02 and A*02:01 emerged as the most commonly mutated class I alleles with a few hotspot mutations identified, particularly in exon 1 (c.19C>T, p.R7X, Fig.1B,C), confirming previous reports. 2,3 In class II, DQB1 and DPA1 loci were frequently targeted by fine mutational events, while more complex allelic loss phenomena interested prevalently DRB1 and DQB1 loci. Those aberrations were identified at diagnosis (35%), during disease follow-up (33%) or at the time of malignant evolution (27%), with higher clonal size in specimens collected during the course of the disease (median VAF 3% [2-27%] at diagnosis, 8% [2-98%] at follow-up, and 2.2% [2.0-6.1%] at evolution). Of 41 patients with at least one HLA aberration and characterized with an extended genotypic study, only 6 harbored also >1 somatic myeloid mutation (14%), versus 30/90 (33%) not affected by somatic hits in HLA (p=.026; Fig.1D). HLA aberrant cases also showed lower number of somatic myeloid mutations (OR=0.44; p=.0262) with driver hits rarely present (Fig.1E). In terms of PIGA mutations, an increased PIGA mosaicism was observed in the HLA mutant group, underlying that both processes have similar pathophysiologic origin as a product of the immune selection pressure (OR: 1.55 [95%CI 1.1-4.2], p=.0201). We then investigated, through WGS of 53 patients, the presence of somatic mutations in other immune genes which could be triggered by immune pressure. Hence, in 47% of the cases we were able to find pathogenic or likely pathogenic hits in genes encompassing proteasome complex, vesicle trafficking, transactivators and interferon regulatory factors, including CREBBP, TAP1, CIITA, PSMC5, PSMB4 and IRF9 (Fig.1F), whose pathogenicity was computationally assessed through recently implemented somatic classifiers. 4 Those hits were not mutually exclusive neither with HLA nor with PIGA mutations, however their VAF was significantly lower compared to concurrent HLA and PIGA lesions, underscoring their lower driver potential within the immune escape environment compared to PNH and HLA-lacking clones. Altogether our results describe the diversity of molecular and immune events taking place in IAA and PNH. Our study suggests that following initial immune insult, clonal architecture of residual hematopoiesis can be dominated by multiple modes of immune escape, agonistically participating to a mechanism of "adaptive" clonal recovery, likely in opposition to the "maladaptive" malignant progression. Figure 1 Figure 1. Disclosures Maciejewski: Alexion: Consultancy; Regeneron: Consultancy; Novartis: Consultancy; Bristol Myers Squibb/Celgene: Consultancy.


Author(s):  
И.Ю. Малышев ◽  
О.П. Буданова ◽  
Л.Ю. Бахтина

Обзор посвящен анализу нарушений иммунитета при раке предстательной железы и возможности иммунотерапии его восстанавливать для уничтожения опухоли. В первой части обзора анализируется противораковый иммунный цикл и его регуляторы CTLA-4 и PD-1. Этот цикл состоит из семи этапов: 1 - высвобождение антигенов опухоли; 2 - захват антигенов; 3 - антигенпрезентация и активация CTL; 4 - перемещение CTL с кровотоком к месту локализации опухоли; 5 - инфильтрация опухоли CTL; 6 - распознавание раковых клеток CTL; и 7 - уничтожение раковых клеток. Далее рассматривается способность опухоли иммуноредактировать противораковый цикл с помощью: 1 - выставления не-иммунногенных антигенов или апоптотической гибели без высвобождения антигенов; 2 - увеличения продукции IDO, антивоспалительных цитокинов, CTLA-4, PD-1 и PD-L1/PD-L2, привлечения MDSC, Tregs и М2 макрофагов для нарушения процесса антиген-презентации и снижения активности и выживаемости СТL; 3 - разрушения хемокинов CX3CL1, CXCL9, CXCL10 и CCL5 и выделения VEGF для ослабления миграции CTL в опухоль; и 4 - уменьшения количества MHCI для снижения распознавания раковых клеток СТL. Эти механизмы обеспечивают выживание опухоли при иммунной атаке. Проведён анализ технологий иммунотерапии рака: 1 - технологии прямого стимулирования антиопухолевого иммунитета, такие как вакцины и CTL/TIL; 2 - технологии имитации ключевых этапов цикла против рака, такие как лимфоциты с модифицированным TCR и CAR-T клетки; 3 - технологии ингибирования иммуносупрессии, такие как ингибиторы PD-1 и CTLA-4; и 4 - комбинированные технологии из двух и более технологий. Уже сегодня иммунотерапия демонстрирует потенциал одного из самых эффективных способов лечения рака, в первую очередь, благодаря использованию иммунитета самого больного. Это порождает надежду на то, что иммунотерапия, по мере своего совершенствования, сможет полностью и безопасно уничтожить рак. The review focused on analysis of immunity disorders in prostate cancer and capabilities of immunotherapy for recovering the immunity to destroy the tumor. The first part of the review analyzed the anticancer immune cycle and its regulators, CTLA-4 and PD-1. This cycle consists of seven stages: 1) releasing tumor antigens; 2) capturing antigens; 3) antigen presentation and CTL activation; 4) CTL transportation with blood flow to the tumor site; 5) CTL infiltration of the tumor; 6) CTL recognition of cancer cells; and 7) destruction of cancer cells. Then the authors addressed the tumor capability for immunoediting the anticancer cycle by 1) presentation of non-immunogenic antigens or apoptotic death without releasing antigens; 2) increasing production of IDO, the anti-inflammatory cytokines, CTLA-4, PD-1, and PD-L1/PD-L2, and engaging MDSC, Tregs, and M2 macrophages to disrupt the antigen presentation process and reduce the CTL activity and survival; 3) destruction of the chemokines, CX3CL1, CXCL9, CXCL10, and CCL5 and release of VEGF to reduce CTL migration to the tumor; and 4) reducing the amount of MHCI to restrict the CTL recognition of cancer cells. These mechanisms ensure the tumor survival during the immune attack. The following technologies of anti-cancer immunotherapy were analyzed: 1) direct stimulation of anti-tumor immunity, for instance with vaccines and CTL/TIL; 2) mimicking key stages of the anti-cancer cycle, for instance, using lymphocytes with modified TCR and CAR-T cells; 3) inhibition of immunosuppression, for instance, with PD-1 and CTLA-4 inhibitors; and 4) combinations of two or more of these technologies. Already now immunotherapy demonstrates its potentiality as a most effective anti-cancer treatment largely due to using the patient’s own immunity. This gives rise to the hope that as immunotherapy is enhanced it will be able to completely and safely destroy cancer.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2546-2546
Author(s):  
Joseph Kim ◽  
Jung-Hyun Kim ◽  
Leslie Cope ◽  
Liping Li ◽  
Raajit Rampal ◽  
...  

Abstract Introduction: Myeloproliferative Neoplasms (MPN) are blood diseases caused by mutations in hematopoietic stem cells (HSCs) which lead to clonal expansion and overproduction of myeloid lineages. Individuals with MPN are at increased risk for transformation to bone marrow fibrosis (myelofibrosis, MF) and acute myeloid leukemia (AML), both of which are associated with poor clinical outcomes. However, targetable mechanisms underlying progression remain elusive. The High Mobility Group A1 (HMGA1) gene encodes chromatin regulators which are enriched in stem cells and aberrantly overexpressed in aggressive tumors (Xian et al Nature Commun 2017;8:15008, Resar et al Cancer Res 2018;78:1890). Transgenic mice misexpressing Hmga1 in lymphoid cells develop lethal leukemia by dysregulating gene networks associated with aberrant proliferation and inflammation (Hillion et al Cancer Res 2008;68:10121, Schuldenfrei et al BMC Genomics, 2011;12:549). We discovered that HMGA1 is overexpressed in MPN with progression and required for leukemic transformation in preclinical models (Resar et al Blood 2018;132 Suppl 1:102). We therefore sought to: 1) test the hypothesis that HMGA1 drives MPN progression by dysregulating gene networks involved in immune evasion, and, 2) identify mediators of immune escape that could be disrupted in therapy. Methods: To elucidate transcriptional networks regulated by HMGA1 during MPN progression to AML, we integrated multi-omics sequencing (seq) analyses, including RNAseq, chromatin immunoprecipitation seq (ChIPseq), and ATACseq in AML cell lines from JAK2 V617Fmutant MPN after leukemic transformation (DAMI, SET-2) + HMGA1 depletion. HMGA1 gene expression was inactivated using CRISPR/Cas9 or short hairpin RNA (shRNA)-mediated gene silencing. Gene set enrichment analysis was used to dissect molecular mechanisms underlying immune invasion by HMGA1. To validate results in human MPN, RNAseq was performed in peripheral blood mononuclear cells (PBMCs) from matched MF patients who transformed to AML. To reconstruct the immune cell composition of primary MPN samples, we applied xCell, a robust computational method that converts gene expression profiles to immune cell types. Transcriptional networks were validated at the level of mRNA and protein via quantitative RT-PCR and flow cytometry. To identify drugs to disrupt HMGA1 immune evasion networks, we applied the Broad Institute Connectivity Map (CMAP) and cytotoxicity assays. Results: Integration of RNAseq, ChIPseq, and ATACseq in MPN AML cells (DAMI, SET-2) revealed that HMGA1 represses genes involved in immune activation (inflammatory response, TNFa signaling, NF-κB networks) and antigen presentation [Interferon gamma (IFNγ) response networks], including genes encoding the major histocompatibility complex (MHC) class I and II antigens. Inhibiting HMGA1 results in up-regulation of MHC class I and II antigen genes, with greatest induction of HLA-DRA (the alpha paralog for HLA Class II antigens). Similarly, HMGA1 depletion increases cell surface expression of HLA-DRA antigens. Strikingly, RNAseq from MPN patients with MF after transformation to AML reveal that HMGA1 is up-regulated in MPN AML concurrent with repression in gene networks of immune activation and antigen presentation. Immune cell transcriptomes are also depleted in MPN cells after leukemic transformation. To determine if immune evasion by HMGA1 could be modulated in therapy, we applied CMAP which identified histone deacetylase inhibitors (HDACi) as drugs targeting the HMGA1 transcriptome. The HDACi, entinostat, is cytotoxic and synergizes with the JAK inhibitor, ruxolitinib, in MPN AML cell lines. Further, entinostat induces expression of MHC class II genes and antigens. Moreover, HMGA1 depletion enhances sensitivity of MPN AML cells to entinostat. Conclusions: We discovered an epigenetic program whereby HMGA1 drives immune evasion during MPN progression by binding to chromatin and enhancing chromatin accessibility to activate transcriptional networks that repress antigen presentation and immune attack. Most importantly, HMGA1 immune evasion networks are dysregulated in human MPN and can be targeted by HDACi therapy. Together, our studies reveal a new paradigm whereby HMGA1 down-regulates MHC antigens during MPN progression, suggesting that targeting HMGA1 networks could activate an immune attack and prevent MPN progression. Figure 1 Figure 1. Disclosures Rampal: Novartis: Consultancy; Pharmaessentia: Consultancy; Kartos: Consultancy; Blueprint: Consultancy; Disc Medicine: Consultancy; BMS/Celgene: Consultancy; Stemline: Consultancy, Research Funding; Constellation: Research Funding; Jazz Pharmaceuticals: Consultancy; Sierra Oncology: Consultancy; Abbvie: Consultancy; CTI: Consultancy; Incyte: Consultancy, Research Funding; Memorial Sloan Kettering: Current Employment.


2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A28-A29
Author(s):  
M Thelen ◽  
K Wennhold ◽  
J Lehmann ◽  
E Staib ◽  
MA Garcia Marquez ◽  
...  

BackgroundImmune-checkpoint inhibition (CKI) demonstrated remarkable therapeutic efficacy in several kinds of cancer. However, immune escape mechanisms lead to primary or secondary resistance in the majority of patients. Most predictive biomarkers failed, as the primary target of CKI is not the tumor cell itself, but the crosstalk between immune- and cancer cells. We aimed to characterize the immune evasion landscape in primary tumors across different entities.Materials and MethodsExpression of 32 immune-regulatory molecules on lymphocytes was analyzed in peripheral blood and tumor infiltrating lymphocytes (TILs) of 146 primary tumor patients across 10 different entities using flow cytometry. NanoString was applied to determine RNA expression of the respective ligands and 20 genes associated with antigen presentation. Expression of coinhibitory ligands on tumor cells was assessed by immunohistochemistry. To quantify the immune cell infiltration, digital pathology was used and the Immunoscore was generated for each patient.ResultsWhile an increase of regulatory T cells was a common feature across all entities, we found site-specific differences regarding other lymphocyte subsets and expression of immune-regulatory molecules by TILs and tumor cells. Expression of co-inhibitory molecules on tumor infiltrating T cells accumulated especially in advanced stage cancers whereas immune cell infiltration was mainly associated with enhanced antigen presentation. Co-expression of multiple immune-inhibitory ligands was most frequent in colorectal, lung and ovarian carcinoma. Genes related to antigen presentation were frequently dysregulated in seminoma, liver and lung cancer.ConclusionsImmune evasion is a common feature of cancer and frequently detected co-occurrence of multiple mechanisms probably contributes to resistance against immunotherapy. We describe substantial heterogeneity regarding immune escape mechanisms between patients with the same primary tumor. Individualized immunotherapeutic strategies based on pretherapeutic evaluation of the immune evasion landscape might help to improve response to CKI.Disclosure InformationM. Thelen: None. K. Wennhold: None. J. Lehmann: None. E. Staib: None. M.A. Garcia Marquez: None. P. Lohneis: None. A. Lechner: None. S. Wagener-Ryczek: None. P.S. Plum: None. D. Pfister: None. F. Dörr: None. D. Beutner: None. F. Thangarajah: None. D. Ratiu: None. W. Malter: None. S. Merkelbach-Bruse: None. C.J. Bruns: None. A. Quaas: None. M.S. von Bergwelt-Baildon: B. Research Grant (principal investigator, collaborator or consultant and pending grants as well as grants already received); Significant; Astellas. F. Consultant/Advisory Board; Modest; Bristol-Myers Squibb. H.A. Schlößer: B. Research Grant (principal investigator, collaborator or consultant and pending grants as well as grants already received); Significant; Astra Zeneca.


Author(s):  
Stacey J. Scott ◽  
Xiaodun Li ◽  
Sriganesh Jammula ◽  
Ginny Devonshire ◽  
Catherine Lindon ◽  
...  

AbstractPolyploidy is present in many cancer types and is increasingly recognized as an important factor in promoting chromosomal instability, genome evolution, and heterogeneity in cancer cells. However, the mechanisms that trigger polyploidy in cancer cells are largely unknown. In this study, we investigated the origin of polyploidy in esophageal adenocarcinoma (EAC), a highly heterogenous cancer, using a combination of genomics and cell biology approaches in EAC cell lines, organoids, and tumors. We found the EAC cells and organoids present specific mitotic defects consistent with problems in the attachment of chromosomes to the microtubules of the mitotic spindle. Time-lapse analyses confirmed that EAC cells have problems in congressing and aligning their chromosomes, which can ultimately culminate in mitotic slippage and polyploidy. Furthermore, whole-genome sequencing, RNA-seq, and quantitative immunofluorescence analyses revealed alterations in the copy number, expression, and cellular distribution of several proteins known to be involved in the mechanics and regulation of chromosome dynamics during mitosis. Together, these results provide evidence that an imbalance in the amount of proteins implicated in the attachment of chromosomes to spindle microtubules is the molecular mechanism underlying mitotic slippage in EAC. Our findings that the likely origin of polyploidy in EAC is mitotic failure caused by problems in chromosomal attachments not only improves our understanding of cancer evolution and diversification, but may also aid in the classification and treatment of EAC and possibly other highly heterogeneous cancers.


Author(s):  
Anders Berglund ◽  
Ryan M. Putney ◽  
Imene Hamaidi ◽  
Sungjune Kim

AbstractCancer immune evasion is one of the hallmarks of carcinogenesis. Cancer cells employ multiple mechanisms to avoid immune recognition and suppress antitumor immune responses. Recently, accumulating evidence has indicated that immune-related pathways are epigenetically dysregulated in cancer. Most importantly, the epigenetic footprint of immune-related pathways is associated with the patient outcome, underscoring the crucial need to understand this process. In this review, we summarize the current evidence for epigenetic regulation of immune-related pathways in cancer and describe bioinformatics tools, informative visualization techniques, and resources to help decipher the cancer epigenome.


2021 ◽  
Vol 13 (1) ◽  
Author(s):  
Jonathan D. Licht ◽  
Richard L. Bennett

Abstract Background Epigenetic mechanisms regulate chromatin accessibility patterns that govern interaction of transcription machinery with genes and their cis-regulatory elements. Mutations that affect epigenetic mechanisms are common in cancer. Because epigenetic modifications are reversible many anticancer strategies targeting these mechanisms are currently under development and in clinical trials. Main body Here we review evidence suggesting that epigenetic therapeutics can deactivate immunosuppressive gene expression or reprogram tumor cells to activate antigen presentation mechanisms. In addition, the dysregulation of epigenetic mechanisms commonly observed in cancer may alter the immunogenicity of tumor cells and effectiveness of immunotherapies. Conclusions Therapeutics targeting epigenetic mechanisms may be helpful to counter immune evasion and improve the effectiveness of immunotherapies.


2021 ◽  
Vol 21 (5) ◽  
pp. 298-312
Author(s):  
Suchit Jhunjhunwala ◽  
Christian Hammer ◽  
Lélia Delamarre

Sign in / Sign up

Export Citation Format

Share Document