Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Cancer Res ; 82(24): 4571-4585, 2022 12 16.
Article En | MEDLINE | ID: mdl-36353752

Myofibroblastic cancer-associated fibroblast (myoCAF)-rich tumors generally contain few T cells and respond poorly to immune-checkpoint blockade. Although myoCAFs are associated with poor outcome in most solid tumors, the molecular mechanisms regulating myoCAF accumulation remain unclear, limiting the potential for therapeutic intervention. Here, we identify ataxia-telangiectasia mutated (ATM) as a central regulator of the myoCAF phenotype. Differentiating myofibroblasts in vitro and myoCAFs cultured ex vivo display activated ATM signaling, and targeting ATM genetically or pharmacologically could suppress and reverse differentiation. ATM activation was regulated by the reactive oxygen species-producing enzyme NOX4, both through DNA damage and increased oxidative stress. Targeting fibroblast ATM in vivo suppressed myoCAF-rich tumor growth, promoted intratumoral CD8 T-cell infiltration, and potentiated the response to anti-PD-1 blockade and antitumor vaccination. This work identifies a novel pathway regulating myoCAF differentiation and provides a rationale for using ATM inhibitors to overcome CAF-mediated immunotherapy resistance. SIGNIFICANCE: ATM signaling supports the differentiation of myoCAFs to suppress T-cell infiltration and antitumor immunity, supporting the potential clinical use of ATM inhibitors in combination with checkpoint inhibition in myoCAF-rich, immune-cold tumors.


Ataxia Telangiectasia Mutated Proteins , Cancer-Associated Fibroblasts , Immunotherapy , Neoplasms , Humans , Ataxia Telangiectasia Mutated Proteins/metabolism , Cancer-Associated Fibroblasts/metabolism , Cell Differentiation , Myofibroblasts/metabolism , Drug Resistance, Neoplasm
2.
Front Immunol ; 13: 954567, 2022.
Article En | MEDLINE | ID: mdl-36119104

Oral potentially malignant disorders (OPMD) are precursors of oral squamous cell carcinoma (OSCC), and the presence of oral epithelial dysplasia (OED) in OPMD confers an increased risk of malignant transformation. Emerging evidence has indicated a role for the immune system in OPMD disease progression; however, the underlying immune mechanisms remain elusive. In this study, we used immune signatures established from cancer to delineate the immune profiles of moderate and severe OED, which are considered high-risk OPMD. We demonstrated that moderate and severe OEDs exhibit high lymphocyte infiltration and upregulation of genes involved in both immune surveillance (major histocompatibility complex-I, T cells, B cells and cytolytic activity) and immune suppression (immune checkpoints, T regulatory cells, and tumor-associated macrophages). Notably, we identified three distinct subtypes of moderate and severe OED: immune cytotoxic, non-cytotoxic and non-immune reactive. Active immune surveillance is present in the immune cytotoxic subtype, whereas the non-cytotoxic subtype lacks CD8 immune cytotoxic response. The non-immune reactive subtype showed upregulation of genes involved in the stromal microenvironment and cell cycle. The lack of T cell infiltration and activation in the non-immune reactive subtype is due to the dysregulation of CTNNB1, PTEN and JAK2. This work suggests that moderate and severe OED that harbor the non-cytotoxic or non-immune reactive subtype are likely to progress to cancer. Overall, we showed that distinct immune responses are present in high-risk OPMD, and revealed targetable pathways that could lead to potential new approaches for non-surgical management of OED.


Carcinoma, Squamous Cell , Mouth Neoplasms , Precancerous Conditions , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Transformation, Neoplastic/genetics , Humans , Hyperplasia , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Precancerous Conditions/genetics , Tumor Microenvironment/genetics
3.
Cancers (Basel) ; 14(14)2022 Jul 06.
Article En | MEDLINE | ID: mdl-35884359

We report, in brief, the results of a phase I, non-randomized study of idiotypic DNA vaccination in patients with B-cell non-Hodgkin's lymphoma (ISRCTN31090206). The DNA sequence of lymphoma-derived immunoglobulin variable regions was used as a tumor-specific antigen fused to the potato virus X coat protein. A conjugate of plasmid DNA with polyethylenimine was used for the intramuscular injections, followed by a boost with an oral live-attenuated Salmonella vaccine carrying the same plasmid. The patients with a complete or partial response to previous chemotherapy received one or two courses of vaccination, including four injections at monthly intervals. The vaccine was well tolerated, with low-grade adverse events. The T-cell immune responses were assessed by ELISpot, at last vaccine, one week and one month post-vaccination, and were detected in 11/14 (78.6%) of the patients. In cases of progression requiring chemotherapy, or the presence of a positive MRD after the first course of vaccination, the patients underwent a second course of vaccination. At the end point, 6/19 vaccinated patients had disease stabilization, while 13/19 were in complete remission. The overall survival was 100% at follow-up, of a median of 2.3 years.

4.
J Immunother Cancer ; 10(3)2022 03.
Article En | MEDLINE | ID: mdl-35361728

BACKGROUND: Cancer is characterized by an accumulation of somatic mutations, of which a significant subset can generate cancer-specific neoepitopes that are recognized by autologous T cells. Such neoepitopes are emerging as important targets for cancer immunotherapy, including personalized cancer vaccination strategies. METHODS: We used whole-exome and RNA sequencing analysis to identify potential neoantigens for a patient with non-small cell lung cancer. Thereafter, we assessed the autologous T-cell reactivity to the candidate neoantigens using a long peptide approach in a cultured interferon gamma ELISpot and tracked the neoantigen-specific T-cells in the tumor by T-cell receptor (TCR) sequencing. In parallel, identified gene variants were incorporated into a Modified Vaccinia Ankara-based vaccine, which was evaluated in the human leucocyte antigen A*0201 transgenic mouse model (HHD). RESULTS: Sequencing revealed a tumor with a low mutational burden: 2219 sequence variants were identified from the primary tumor, of which 23 were expressed in the transcriptome, involving 18 gene products. We could demonstrate spontaneous T-cell responses to 5/18 (28%) mutated gene variants, and further analysis of the TCR repertoire of neoantigen-specific CD4+ and CD8+ T cells revealed TCR clonotypes that were expanded in both blood and tumor tissue. Following vaccination of HHD mice, de novo T-cell responses were generated to 4/18 (22%) mutated gene variants; T cells reactive against two variants were also evident in the autologous setting. Subsequently, we determined the major histocompatibility complex restriction of the T-cell responses and used in silico prediction tools to determine the likely neoepitopes. CONCLUSIONS: Our study demonstrates the feasibility of efficiently identifying tumor-specific neoantigens that can be targeted by vaccination in tumors with a low mutational burden, promising successful clinical exploitation, with trials currently underway.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Humans , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Mice , Vaccination
5.
Front Immunol ; 12: 763086, 2021.
Article En | MEDLINE | ID: mdl-34733290

HPV-independent head and neck squamous cell carcinoma (HNSCC) is a common cancer globally. The overall response rate to anti-PD1 checkpoint inhibitors (CPIs) in HNSCC is ~16%. One major factor influencing the effectiveness of CPI is the level of tumor infiltrating T cells (TILs). Converting TILlow tumors to TILhigh tumors is thus critical to improve clinical outcome. Here we describe a novel DNA vaccines to facilitate the T-cell infiltration and control tumor growth. We evaluated the expression of target antigens and their respective immunogenicity in HNSCC patients. The efficacy of DNA vaccines targeting two novel antigens were evaluated with or without CPI using a syngeneic model. Most HNSCC patients (43/44) co-expressed MAGED4B and FJX1 and their respective tetramer-specific T cells were in the range of 0.06-0.12%. In a preclinical model, antigen-specific T cells were induced by DNA vaccines and increased T cell infiltration into the tumor, but not MDSC or regulatory T cells. The vaccines inhibited tumor growth and improved the outcome alone and upon combination with anti-PD1 and resulted in tumor clearance in approximately 75% of mice. Pre-existence of MAGED4B and FJX1-reactive T cells in HNSCC patients suggests that these widely expressed antigens are highly immunogenic and could be further expanded by vaccination. The DNA vaccines targeting these antigens induced robust T cell responses and with the anti-PD1 antibody conferring excellent tumor control. This opens up an opportunity for combination immunotherapy that might benefit a wider population of HNSCC patients in an antigen-specific manner.


Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Immune Checkpoint Inhibitors/therapeutic use , Squamous Cell Carcinoma of Head and Neck/therapy , Vaccines, DNA/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Combined Modality Therapy , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology , T-Lymphocytes/immunology , Young Adult
7.
Cells ; 9(9)2020 09 15.
Article En | MEDLINE | ID: mdl-32942747

Head and neck squamous cell carcinomas (HNSCC) are a heterogeneous group of malignant tumours typically caused by alcohol and tobacco consumption, although an increasing number of HNSCC arise due to persistent infection with high-risk human papilloma virus (HPV). The treatment of HNSCC remains challenging, and the first-line setting is focused on surgery and chemoradiotherapy. A substantial proportion of HNSCC patients die from their disease, especially those with recurrent and metastatic disease. Among factors linked with good outcome, immune cell infiltration appears to have a major role. HPV-driven HNSCC are often T-cell rich, reflecting the presence of HPV antigens that are immunogenic. Tumour-associated antigens that are shared between patients or that are unique to an individual person may also induce varying degrees of immune response; studying these is important for the understanding of the interaction between the host immune system and the cancer. The resulting knowledge is critical for the design of better immunotherapies. Key questions are: Which antigens lead to an adaptive immune response in the tumour? Which of these are exploitable for immunotherapy? Here, we review the current thinking regarding tumour antigens in HNSCC and what has been learned from early phase clinical trials.


Antigens, Viral, Tumor/immunology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Immunotherapy/methods , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/therapy , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Papillomaviridae/immunology , Papillomavirus Infections/complications , Papillomavirus Infections/virology , T-Lymphocytes/immunology
8.
Cancer Res ; 80(9): 1846-1860, 2020 05 01.
Article En | MEDLINE | ID: mdl-32122909

Determining mechanisms of resistance to αPD-1/PD-L1 immune-checkpoint immunotherapy is key to developing new treatment strategies. Cancer-associated fibroblasts (CAF) have many tumor-promoting functions and promote immune evasion through multiple mechanisms, but as yet, no CAF-specific inhibitors are clinically available. Here we generated CAF-rich murine tumor models (TC1, MC38, and 4T1) to investigate how CAFs influence the immune microenvironment and affect response to different immunotherapy modalities [anticancer vaccination, TC1 (HPV E7 DNA vaccine), αPD-1, and MC38] and found that CAFs broadly suppressed response by specifically excluding CD8+ T cells from tumors (not CD4+ T cells or macrophages); CD8+ T-cell exclusion was similarly present in CAF-rich human tumors. RNA sequencing of CD8+ T cells from CAF-rich murine tumors and immunochemistry analysis of human tumors identified significant upregulation of CTLA-4 in the absence of other exhaustion markers; inhibiting CTLA-4 with a nondepleting antibody overcame the CD8+ T-cell exclusion effect without affecting Tregs. We then examined the potential for CAF targeting, focusing on the ROS-producing enzyme NOX4, which is upregulated by CAF in many human cancers, and compared this with TGFß1 inhibition, a key regulator of the CAF phenotype. siRNA knockdown or pharmacologic inhibition [GKT137831 (Setanaxib)] of NOX4 "normalized" CAF to a quiescent phenotype and promoted intratumoral CD8+ T-cell infiltration, overcoming the exclusion effect; TGFß1 inhibition could prevent, but not reverse, CAF differentiation. Finally, NOX4 inhibition restored immunotherapy response in CAF-rich tumors. These findings demonstrate that CAF-mediated immunotherapy resistance can be effectively overcome through NOX4 inhibition and could improve outcome in a broad range of cancers. SIGNIFICANCE: NOX4 is critical for maintaining the immune-suppressive CAF phenotype in tumors. Pharmacologic inhibition of NOX4 potentiates immunotherapy by overcoming CAF-mediated CD8+ T-cell exclusion. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/9/1846/F1.large.jpg.See related commentary by Hayward, p. 1799.


Cancer-Associated Fibroblasts , Neoplasms , Animals , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Humans , Immunotherapy , Mice , NADPH Oxidase 4 , Reactive Oxygen Species
9.
Front Immunol ; 9: 830, 2018.
Article En | MEDLINE | ID: mdl-29740440

Head and neck cancer (HNC) is a heterogeneous group of squamous cell cancers that affect the oral cavity, pharynx, and larynx. Worldwide, it is the sixth most common cancer but in parts of Southern and South-East Asia, HNC is one of the most common cancers. A significant proportion of HNC is driven by human papillomavirus (HPV) infection, whereas HPV-independent HNC is associated with alcohol, smoking, and smokeless tobacco consumption. Here, we review the past and present experience of targeting HNC with vaccination focusing on HPV-derived antigens as well as non-viral antigens for HPV-negative HNC. Novel therapeutic approaches for HNC will focus not only on effective vaccine platforms but will also target the stroma-rich immunosuppressive microenvironment found in those tumours.


Antigens, Neoplasm/immunology , Antigens, Viral/immunology , Cancer Vaccines/therapeutic use , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Animals , Antigens, Viral/administration & dosage , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Mice , Papillomavirus Infections/complications , Risk Factors , Smoking , Tobacco Use , Tumor Microenvironment
10.
Cancer Immunol Immunother ; 67(4): 627-638, 2018 Apr.
Article En | MEDLINE | ID: mdl-29330557

Vaccination with DNA that encodes cancer antigens is a simple and convenient way to raise immunity against cancer and has already shown promise in the clinical setting. Conventional plasmid DNA is commonly used which together with the encoded antigen also includes bacterial immunostimulatory CpG motifs to target the DNA sensor Toll-like receptor 9. Recently DNA vaccines using doggybone DNA (dbDNA™), have been developed without the use of bacteria. The cell-free process relies on the use of Phi29 DNA polymerase to amplify the template followed by protelomerase TelN to complete individual closed linear DNA. The resulting DNA contains the required antigenic sequence, a promoter and a poly A tail but lacks bacterial sequences such as an antibiotic resistance gene, prompting the question of immunogenicity. Here we compared the ability of doggybone DNA vaccine with plasmid DNA vaccine to induce adaptive immunity using clinically relevant oncotargets E6 and E7 from HPV. We demonstrate that despite the inability to trigger TLR9, doggybone DNA was able to induce similar levels of cellular and humoral immunity as plasmid DNA, with suppression of established TC-1 tumours.


CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunity, Cellular/immunology , Lung Neoplasms/immunology , Plasmids/immunology , Toll-Like Receptor 9/immunology , Vaccines, DNA/immunology , Animals , Disease Models, Animal , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Mice , Mice, Inbred C57BL , Plasmids/administration & dosage , Plasmids/genetics , Tumor Cells, Cultured , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
11.
Eur J Dermatol ; 27(5): 511-518, 2017 Oct 01.
Article En | MEDLINE | ID: mdl-29084638

BACKGROUND: Human papilloma virus (HPV) infects keratinocytes of the skin and mucous membranes, and is associated with the induction of cutaneous warts and malignancy. Warts can induce significant morbidity and disability but most therapies, including cryotherapy, laser, and radiofrequency devices show low efficacy and induce discomfort through tissue destruction. Microwaves are readily capable of passing through highly keratinised skin to deliver energy and induce heating of the tissue in a highly controllable, uniform manner. OBJECTIVES: To determine the effects of microwave on cutaneous HPV infection. MATERIALS & METHODS: We undertook a pilot study of microwave therapy to the skin in 32 consecutive individuals with 52 recalcitrant long-lived viral cutaneous warts. Additionally, we undertook a molecular characterisation of the effects of microwaves on the skin. RESULTS: Tissue inflammation was minimal, but 75.9% of lesions cleared which compares favourably with previous studies showing a clearance rate of 23-33% for cryotherapy or salicylic acid. We show that microwaves specifically induce dendritic cell cross-presentation of HPV antigen to CD8+ T cells and suggest that IL-6 may be important for DC IRF1 and IRF4 modulation to enhance this process. CONCLUSION: Keratinocyte-skin dendritic cell cross-talk is integral to host defence against HPV infections, and this pilot study supports the concept of microwave induction of anti-HPV immunity which offers a promising approach for treatment of HPV-induced viral warts and potentially HPV-related cancers.


Microwaves/therapeutic use , Papillomavirus Infections/radiotherapy , Warts/radiotherapy , Warts/virology , Adult , Aged , Antigens, Viral/immunology , CD8 Antigens/immunology , Dendritic Cells/immunology , Dendritic Cells/radiation effects , Female , Humans , Interferon Regulatory Factor-1/immunology , Interferon Regulatory Factors/immunology , Interleukin-6/immunology , Keratinocytes/immunology , Keratinocytes/radiation effects , Male , Middle Aged , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Pilot Projects , Receptor Cross-Talk , T-Lymphocytes/immunology , Warts/immunology , Young Adult
12.
Curr Top Microbiol Immunol ; 405: 123-143, 2017.
Article En | MEDLINE | ID: mdl-27704269

In the last decade, immunotherapy with monoclonal antibodies targeting immunological check points has become a breakthrough therapeutic modality for solid cancers. However, only up to 50 % of patients benefit from this powerful approach. For others vaccination might provide a plausible addition or alternative. For induction of effective anticancer immunity CD4+ T cell help is required, which is often difficult to induce to self cancer targets because of tolerogenic mechanisms. Our approach for cancer vaccines has been to incorporate into the vaccine design sequences able to activate foreign T cell help, through genetically linking cancer targets to microbial sequences (King et al. in Nat Med 4(11):1281-1286, 1998; Savelyeva et al. in Nat Biotechnol 19(8):760-764, 2001). This harnesses the non-tolerized CD4 T cell repertoire available in patients to help induction of effective immunity against fused cancer antigens. Multiple immune effector mechanisms including antibody, CD8+ T cells as well as CD4 effector T cells can be activated using this strategy. Delivery via DNA vaccines has already indicated clinical efficacy. The same principle of linked T cell help has now been transferred to other novel vaccine modalities to further potentiate immunity against cancer targets.


CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Vaccination , CD8-Positive T-Lymphocytes/immunology , Humans
13.
Oncotarget ; 7(35): 56781-56797, 2016 Aug 30.
Article En | MEDLINE | ID: mdl-27462861

Human papilloma virus (HPV)-associated head and neck squamous cell carcinoma (HNSCC) has a better prognosis than it's HPV negative (HPV(-)) counterpart. This may be due to the higher numbers of tumor-infiltrating lymphocytes (TILs) in HPV positive (HPV(+)) tumors. RNA-Sequencing (RNA-Seq) was used to evaluate whether the differences in clinical behaviour simply reflect a numerical difference in TILs or whether there is a fundamental behavioural difference between TILs in these two settings. Thirty-nine HNSCC tumors were scored for TIL density by immunohistochemistry. After the removal of 16 TILlow tumors, RNA-Seq analysis was performed on 23 TILhigh/med tumors (HPV(+) n=10 and HPV(-) n=13). Using EdgeR, differentially expressed genes (DEG) were identified. Immune subset analysis was performed using Functional Analysis of Individual RNA-Seq/ Microarray Expression (FAIME) and immune gene RNA transcript count analysis. In total, 1,634 DEGs were identified, with a dominant immune signature observed in HPV(+) tumors. After normalizing the expression profiles to account for differences in B- and T-cell number, 437 significantly DEGs remained. A B-cell associated signature distinguished HPV(+) from HPV(-) tumors, and included the DEGs CD200, GGA2, ADAM28, STAG3, SPIB, VCAM1, BCL2 and ICOSLG; the immune signal relative to T-cells was qualitatively similar between TILs of both tumor cohorts. Our findings were validated and confirmed in two independent cohorts using TCGA data and tumor-infiltrating B-cells from additional HPV(+) HNSCC patients. A B-cell associated signal segregated tumors relative to HPV status. Our data suggests that the role of B-cells in the adaptive immune response to HPV(+) HNSCC requires re-assessment.


B-Lymphocytes/cytology , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/virology , Papillomavirus Infections/complications , Aged , B-Lymphocytes/virology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/virology , Cluster Analysis , Female , Gene Expression Profiling , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/virology , Human papillomavirus 16 , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Prognosis , Sequence Analysis, RNA , Squamous Cell Carcinoma of Head and Neck , Transcriptome
14.
Oncoimmunology ; 5(6): e1166323, 2016 Jun.
Article En | MEDLINE | ID: mdl-27471642

Passive antibody therapy for cancer is an effective but costly treatment modality. Induction of therapeutically potent anticancer antibodies by active vaccination is an attractive alternative but has proven challenging in cancer due to tolerogenic pressure in patients. Here, we used the clinically relevant cancer target Her2, known to be susceptible to targeting by antibody therapy, to demonstrate how potent antibody can be induced by vaccination. A novel 44kD Her2 protein fragment was generated and found to be highly effective at inducing anti-Her2 antibody including trastuzumab-like reactivities. In the tolerant and spontaneous BALB-neuT mouse model of metastatic breast cancer this Her2-targeting vaccine was only effective if the fragment was conjugated to a foreign immunogenic carrier; Fragment C of tetanus toxin. Only the conjugate vaccine induced high affinity anti-Her2 antibody of multiple isotypes and suppressed tumor development. The magnitude of CD4(+) T-cell help and breadth of cytokines secreted by the CD4(+) T helper (Th) cells induced to the foreign antigen was critical. We used a highly efficient plant-based bio-manufacturing process for protein antigens, magnICON, for vaccine expression, to underpin feasibility of future clinical testing. Hence, our novel Her2-targeting conjugate vaccine combines preclinical efficacy with clinical deliverability, thus setting the scene for therapeutic testing.

15.
Blood ; 127(24): 3015-25, 2016 06 16.
Article En | MEDLINE | ID: mdl-27002119

Kinase inhibitors targeting the B-cell receptor (BCR) are now prominent in the treatment of chronic lymphocytic leukemia (CLL). We have focused here on interleukin 4 (IL-4), a cytokine that protects normal and malignant B cells from apoptosis and increases surface immunoglobulin M (sIgM) expression on murine splenic B cells. First, we have demonstrated that IL-4 treatment increased sIgM expression in vitro on peripheral blood B cells obtained from healthy individuals. In CLL, IL-4 target genes are overexpressed in cells purified from the lymph nodes of patients compared with cells derived from matched blood and bone marrow samples. As for normal B cells, IL-4 increased sIgM expression on CLL cells in vitro, especially in samples expressing unmutated V-genes. IL-4-induced sIgM expression was associated with increased receptor signalling activity, measured by anti-IgM-induced calcium mobilization, and with increased expression of CD79B messenger RNA and protein, and the "mature" glycoform of sIgM. Importantly, the ability of the BCR-associated kinase inhibitors idelalisib and ibrutinib, approved for treatment of CLL and other B-cell malignancies, to inhibit anti-IgM-induced signalling was reduced following IL-4 pretreatment in samples from the majority of patients. In contrast to stimulatory effects on sIgM, IL-4 decreased CXCR4 and CXCR5 expression; therefore, CLL cells, particularly within the progressive unmutated V-gene subset, may harness the ability of IL-4 to promote BCR signalling and B-cell retention within lymph nodes. Effects of IL-4 were mediated via JAK3/STAT6 and we propose a potential role for JAK inhibitors in combination with BCR kinase inhibitors for the treatment of CLL.


Cell Membrane/metabolism , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Interleukin-4/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Drug Interactions , Gene Expression Regulation, Leukemic/drug effects , Humans , Janus Kinase 3/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Neutrophils/drug effects , Neutrophils/metabolism , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , STAT6 Transcription Factor/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
16.
PLoS One ; 10(2): e0118096, 2015.
Article En | MEDLINE | ID: mdl-25692288

Induction of potent antibody is the goal of many vaccines targeted against infections or cancer. Modern vaccine designs that use virus-like particles (VLP) have shown efficacy for prophylactic vaccination against virus-associated cancer in the clinic. Here we used plant viral particles (PVP), which are structurally analogous to VLP, coupled to a weak idiotypic (Id) tumour antigen, as a conjugate vaccine to induce antibody against a murine B-cell malignancy. The Id-PVP vaccine incorporates a natural adjuvant, the viral ssRNA, which acts via TLR7. It induced potent protective anti-Id antibody responses in an in vivo mouse model, superior to the "gold standard" Id vaccine, with prevalence of the IgG2a isotype. Combination with alum further increased antibody levels and maintained the IgG2a bias. Engagement of TLR7 in vivo was followed by secretion of IFN-α by plasmacytoid dendritic cells and by activation of splenic CD11chi conventional dendritic cells. The latter was apparent from up-regulation of co-stimulatory molecules and from secretion of a wide range of inflammatory cytokines and chemokines including the Th1-governing cytokine IL-12, in keeping with the IgG2a antibody isotype distribution. PVP conjugates are a novel cancer vaccine design, offering an attractive molecular form, similar to VLP, and providing T-cell help. In contrast to VLP, they also incorporate a safe "in-built" ssRNA adjuvant.


Antibodies, Viral/immunology , Plant Viruses/immunology , Toll-Like Receptor 7/metabolism , Animals , Dendritic Cells , Female , Immunoglobulin G/immunology , Interleukin-12/metabolism , Lymphoma , Male , Mice , Mice, Inbred C57BL , Nicotiana/virology , Toll-Like Receptor 7/genetics , Virion/immunology
17.
Haematologica ; 99(7): 1138-48, 2014 Jul.
Article En | MEDLINE | ID: mdl-24986876

Biologists and clinicians agree that the B-cell receptor influences the behavior of chronic lymphocytic leukemia, and promising new drugs are aimed at receptor-associated kinases. Engagement of surface immunoglobulin by antigen is a key driver of malignant cells with outcome influenced by the nature of the cell, the level of stimulation and the microenvironment. Analysis of surface immunoglobulin-mediated signaling in the two major disease subsets defined by IGHV mutational status reveals bifurcation of responses toward proliferation or anergy. Mutated chronic lymphocytic leukemia, generally of relatively good prognosis, is mainly, but not exclusively, driven towards anergy in vivo. In contrast, unmutated chronic lymphocytic leukemia shows less evidence for anergy in vivo retaining more responsiveness to surface immunoglobulin M-mediated signaling, possibly explaining increased tumor progression. Expression and function of surface immunoglobulin M in unmutated chronic lymphocytic leukemia appear rather homogeneous, but mutated chronic lymphocytic leukemia exhibits a highly heterogeneous profile that may relate to further variable clinical behavior within this subset. Anergy should increase susceptibility to apoptosis but, in leukemic cells, this may be countered by overexpression of the B-cell lymphoma-2 survival protein. Maintained anergy spreads to chemokines and adhesion molecules, restraining homing and migration. However, anergy is not necessarily completely benign, being able to reverse and regenerate surface immunoglobulin M-mediated responses. A two-pronged attack on proliferative and anti-apoptotic pathways may succeed. Increased understanding of how chronic lymphocytic leukemia cells are driven to anergy or proliferation should reveal predictive biomarkers of progression and of likely response to kinase inhibitors, which could assist therapeutic decisions.


Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Animals , Antigens/immunology , Antigens/metabolism , Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Proliferation , Clonal Anergy/immunology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/immunology
18.
Int J Cancer ; 133(6): 1400-7, 2013 Sep 15.
Article En | MEDLINE | ID: mdl-23494538

The cancer/testis antigen NY-ESO-1 contains an immunodominant HLA-A2-binding peptide (SLLMWITQC), designated S9C, an attractive target for vaccination against several human cancers. As cysteine contains a reactive -SH, the oxidation status of exogenous synthetic peptide is uncertain. We have designed tolerance-breaking DNA fusion vaccines incorporating a domain of tetanus toxin fused to tumor-derived peptide sequences (p.DOM-peptide), placed at the C-terminus for optimal immunogenicity. In a "humanized" HLA-A2 preclinical model, p.DOM-S9C primed S9C-specific CD8+ T cells more effectively than adjuvanted synthetic peptide. A DNA vaccine encoding the full NY-ESO-1 sequence alone induced only weak S9C-specific responses, amplified by addition of DOM sequence. The analog peptide (SLLMWITQL) also primed peptide-specific CD8+ T cells, again increased by DNA delivery. Importantly, T cells induced by S9C-encoding DNA vaccines killed tumor cells expressing endogenous NY-ESO-1. Only a fraction of T cells induced by the S9L-encoding DNA vaccines was able to recognize S9C and kill tumor cells. These data indicate that DNA vaccines mimic posttranslational modifications of -SH-containing peptides expressed by tumor cells. Instability of synthetic peptides and the potential dangers of analog peptides contrast with the ability of DNA vaccines to induce high levels of tumor-lytic peptide-specific CD8+ T cells. These findings encourage clinical exploration of this vaccine strategy to target NY-ESO-1.


Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cytotoxicity, Immunologic , Epitopes/immunology , Immunodominant Epitopes , Membrane Proteins/immunology , Vaccines, DNA/immunology , Amino Acid Sequence , Animals , Cysteine/immunology , Humans , Male , Mice , Molecular Sequence Data
19.
Blood ; 118(3): 650-9, 2011 Jul 21.
Article En | MEDLINE | ID: mdl-21613251

Induction of antibody-mediated immunity against hematologic malignancies requires CD4(+) T-cell help, but weak tumor antigens generally fail to induce adequate T-cell responses, or to overcome tolerance. Conjugate vaccines can harness alternative help to activate responses, but memory B cells may then be exposed to leaking tumor-derived antigen without CD4(+) T-cell support. We showed previously using lymphoma-derived idiotypic antigen that exposure to "helpless" antigen silences the majority of memory IgG(+) B cells. Transfer experiments now indicate that silencing is permanent. In marked contrast to IgG, most coexisting IgM(+) memory B cells exposed to "helpless" antigen survive. Confirmation in a hapten (NP) model allowed measurement of affinity, revealing this, rather than isotype, as the determinant of survival. IgM(+) B cells had Ig variable region gene usage similar to IgG but with fewer somatic mutations. Survival of memory B cells appears variably controlled by affinity for antigen, allowing a minority of low affinity IgG(+), but most IgM(+), memory B cells to escape deletion in the absence of T-cell help. The latter remain, but the majority fail to undergo isotype switch. These findings could apply to other tumor antigens and are relevant for vaccination strategies aimed to induce long-term antibody.


Antigens, Neoplasm/immunology , B-Lymphocytes/immunology , Cancer Vaccines/immunology , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Immunologic Memory/immunology , Adoptive Transfer , Animals , B-Lymphocytes/cytology , Cell Survival/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis/immunology , Vaccines, DNA/immunology
20.
Eur J Immunol ; 40(4): 976-85, 2010 Apr.
Article En | MEDLINE | ID: mdl-20104490

Antigen-specific CD4(+) T cells are central to natural and vaccine-induced immunity. An ongoing antigen-specific T-cell response can, however, influence surrounding T cells with unrelated antigen specificities. We previously observed this bystander effect in healthy human subjects following recall vaccination with tetanus toxoid (TT). Since this interplay could be important for maintenance of memory, we have moved to a mouse model for further analysis. We investigated whether boosting memory CD4(+) T cells against TT in vivo would influence injected CD4(+) TCR transgenic T cells (OT-II) specific for an unrelated OVA peptide. If OT-II cells were pre-activated with OVA peptide in vitro, these cells showed a bystander proliferative response during the ongoing parallel TT-specific response. Bystander proliferation was dependent on boosting of the TT-specific memory response in the recipients, with no effect in naive mice. Bystander stimulation was also proportional to the strength of the TT-specific memory T-cell response. T cells activated in vitro displayed functional receptors for IL-2 and IL-7, suggesting these as potential mediators. This crosstalk between a stimulated CD4(+) memory T-cell response and CD4(+) T cells activated by an unrelated antigen could be important in human subjects continually buffeted by environmental antigens.


Bystander Effect/immunology , CD4-Positive T-Lymphocytes/immunology , Immunization, Secondary , Immunologic Memory/immunology , Lymphocyte Activation , Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/drug effects , Cytokines/pharmacology , Humans , Interleukin Receptor Common gamma Subunit/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor beta Subunit/immunology , Interleukins/biosynthesis , Interleukins/genetics , Interleukins/pharmacology , Interleukins/physiology , Lymphocyte Subsets/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Peptide Fragments/immunology , Receptors, Interleukin/immunology , Recombinant Proteins/pharmacology , T-Cell Antigen Receptor Specificity , Tetanus Toxoid/immunology
...