Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.196
1.
J Immunother Cancer ; 12(5)2024 May 23.
Article En | MEDLINE | ID: mdl-38782540

BACKGROUND: Approximately half of the neuroblastoma patients develop high-risk neuroblastoma. Current treatment involves a multimodal strategy, including immunotherapy with dinutuximab (IgG ch14.18) targeting GD2. Despite achieving promising results, the recurrence rate remains high and poor survival persists. The therapeutic efficacy of dinutuximab is compromised by suboptimal activation of neutrophils and severe neuropathic pain, partially induced by complement activation. METHODS: To enhance neutrophil cytotoxicity, IgG ch14.18 was converted to the IgA isotype, resulting in potent neutrophil-mediated antibody-dependent cell-mediated cytotoxicity (ADCC), without complement activation. However, myeloid checkpoint molecules hamper neutrophil cytotoxicity, for example through CD47 that is overexpressed on neuroblastomas and orchestrates an immunosuppressive environment upon ligation to signal regulatory protein alpha (SIRPα) expressed on neutrophils. In this study, we combined IgA therapy with CD47 blockade. RESULTS: In vitro killing assays showed enhanced IgA-mediated ADCC by neutrophils targeting neuroblastoma cell lines and organoids in comparison to IgG. Notably, when combined with CD47 blockade, both IgG and IgA therapy were enhanced, though the combination with IgA resulted in the greatest improvement of ADCC. Furthermore, in a neuroblastoma xenograft model, we systemically blocked CD47 with a SIRPα fusion protein containing an ablated IgG1 Fc, and compared IgA therapy to IgG therapy. Only IgA therapy combined with CD47 blockade increased neutrophil influx to the tumor microenvironment. Moreover, the IgA combination strategy hampered tumor outgrowth most effectively and prolonged tumor-specific survival. CONCLUSION: These promising results highlight the potential to enhance immunotherapy efficacy against high-risk neuroblastoma through improved neutrophil cytotoxicity by combining IgA therapy with CD47 blockade.


CD47 Antigen , Immunoglobulin A , Neuroblastoma , Neutrophils , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/metabolism , CD47 Antigen/immunology , Humans , Neuroblastoma/immunology , Neuroblastoma/drug therapy , Neutrophils/immunology , Neutrophils/metabolism , Animals , Mice , Immunoglobulin A/immunology , Immunoglobulin A/pharmacology , Immunoglobulin A/metabolism , Cell Line, Tumor , Antibody-Dependent Cell Cytotoxicity , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Xenograft Model Antitumor Assays , Immunotherapy/methods , Female , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use
2.
Int Immunopharmacol ; 133: 112145, 2024 May 30.
Article En | MEDLINE | ID: mdl-38691920

Treatment strategies for paediatric neuroblastoma as well as many other cancers are limited by the unfavourable tumour microenvironment (TME). In this study, the TMEs of neuroblastoma were grouped by their genetic signatures into four distinct subtypes: immune enriched, immune desert, non-proliferative and fibrotic. An Immune Score and a Proliferation Score were constructed based on the molecular features of the subtypes to quantify the immune microenvironment or malignancy degree of cancer cells in neuroblastoma, respectively. The Immune Score correlated with a patient's response to immunotherapy; the Proliferation Score was an independent prognostic biomarker for neuroblastoma and proved to be more accurate than the existing clinical predictors. This double scoring system was further validated and the conserved molecular pattern associated with immune landscape and malignancy degree was confirmed. Axitinib and BI-2536 were confirmed as candidate drugs for neuroblastoma by the double scoring system. Both in vivo and in vitro experiments demonstrated that axitinib-induced pyroptosis of neuroblastoma cells activated anti-tumour immunity and inhibited tumour growth; BI-2536 induced cell cycle arrest at the S phase in neuroblastoma cells. The comprehensive double scoring system of neuroblastoma may predict prognosis and screen for therapeutic strategies which could provide personalized treatments.


Axitinib , Immunotherapy , Neuroblastoma , Tumor Microenvironment , Neuroblastoma/immunology , Neuroblastoma/therapy , Neuroblastoma/pathology , Neuroblastoma/drug therapy , Humans , Tumor Microenvironment/immunology , Prognosis , Animals , Immunotherapy/methods , Cell Line, Tumor , Axitinib/therapeutic use , Child , Male , Female , Child, Preschool , Mice , Infant , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects
3.
PLoS One ; 19(5): e0303136, 2024.
Article En | MEDLINE | ID: mdl-38743689

Superoxide dismutase (SOD) is an antioxidant enzyme that protects the body from free radicals. It has both antioxidant and immunomodulatory properties, inducing macrophage polarization from M1 to M2. Macrophages, key mediators of the innate immune response, are divided into the M1 (pro-inflammatory) and M2 (anti-inflammatory) subtypes. In this study, we aimed to assess the antioxidant and neuroprotective effects of SOD on nerve cells and its immunomodulatory effects on macrophages. We observed that SOD inhibited the accumulation of reactive oxygen species and enhanced the viability of H2O2-treated nerve cells. Furthermore, SOD reduced the degree of necrosis in nerve cells treated with the conditioned medium from macrophages, which induced inflammation. In addition, SOD promoted the M1 to M2 transition of macrophages. Our findings suggest that SOD protects nerve cells and regulates immune responses.


Macrophages , Neuroprotective Agents , Reactive Oxygen Species , Superoxide Dismutase , Animals , Superoxide Dismutase/metabolism , Mice , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Humans , Neuroprotective Agents/pharmacology , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Neuroblastoma/immunology , Neuroblastoma/pathology , Cell Line, Tumor , Hydrogen Peroxide/pharmacology , Cell Survival/drug effects , Antioxidants/pharmacology
4.
Front Immunol ; 15: 1382931, 2024.
Article En | MEDLINE | ID: mdl-38736882

Background: Neuroblastoma (NB) is characterized by both adrenergic (ADRN) and undifferentiated mesenchymal (MES) subsets. The ganglioside sialic acid-containing glycosphingolipid (GD2) is widely overexpressed on tumors of neuroectodermal origin promoting malignant phenotypes. MES cells are greatly enriched in post-therapy and relapsing tumors and are characterized by decreased expression of GD2. This event may cause failure of GD2-based immunotherapy. NK cells represent a key innate cell subset able to efficiently kill tumors. However, the tumor microenvironment (TME) that includes tumor cells and tumor-associated (TA) cells could inhibit their effector function. Methods: We studied eight NB primary cultures that, in comparison with commercial cell lines, more faithfully reflect the tumor cell characteristics. We studied four primary NB-MES cell cultures and two pairs of MES/ADRN (691 and 717) primary cultures, derived from the same patient. In particular, in the six human NB primary cultures, we assessed their phenotype, the expression of GD2, and the enzymes that control its expression, as well as their interactions with NK cells, using flow cytometry, RT-qPCR, and cytotoxicity assays. Results: We identified mature (CD105+/CD133-) and undifferentiated (CD133+/CD105-) NB subsets that express high levels of the MES transcripts WWTR1 and SIX4. In addition, undifferentiated MES cells display a strong resistance to NK-mediated killing. On the contrary, mature NB-MES cells display an intermediate resistance to NK-mediated killing and exhibit some immunomodulatory capacities on NK cells but do not inhibit their cytolytic activity. Notably, independent from their undifferentiated or mature phenotype, NB-MES cells express GD2 that can be further upregulated in undifferentiated NB-MES cells upon co-culture with NK cells, leading to the generation of mature mesenchymal GD2bright neuroblasts. Concerning 691 and 717, they show high levels of GD2 and resistance to NK cell-mediated killing that can be overcome by the administration of dinutuximab beta, the anti-GD2 monoclonal antibody applied in the clinic. Conclusions: NB is a heterogeneous tumor representing a further hurdle in NB immunotherapy. However, different from what was reported with NB commercial cells and independent of their MES/ADRN phenotype, the expression of GD2 and its displayed sensitivity to anti-GD2 mAb ADCC indicated the possible effectiveness of anti-GD2 immunotherapy.


Gangliosides , Killer Cells, Natural , Neuroblastoma , Tumor Escape , Tumor Microenvironment , Humans , Neuroblastoma/immunology , Neuroblastoma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Gangliosides/immunology , Gangliosides/metabolism , Tumor Microenvironment/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Tumor Cells, Cultured , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism
5.
FASEB J ; 38(10): e23644, 2024 May 31.
Article En | MEDLINE | ID: mdl-38738472

Tumors typically lack canonical danger signals required to activate adaptive immunity and also frequently employ substantial immunomodulatory mechanisms that downregulate adaptive responses and contribute to escape from immune surveillance. Given the variety of mechanisms involved in shielding tumors from immune recognition, it is not surprising that single-agent immunomodulatory approaches have been largely unsuccessful in generating durable antitumor responses. Here we report a unique combination of immunomodulatory and cytostatic agents that recondition the tumor microenvironment and eliminate complex and/or poor-prognosis tumor types including the non-immunogenic 4T-1 model of TNBC, the aggressive MOC-2 model of HNSCC, and the high-risk MYCN-amplified model of neuroblastoma. A course of therapy optimized for TNBC cured a majority of tumors in both ectopic and orthotopic settings and eliminated metastatic spread in all animals tested at the highest doses. Immune responses were transferable between therapeutic donor and naïve recipient through adoptive transfer, and a sizeable abscopal effect on distant, untreated lesions could be demonstrated experimentally. Similar results were observed in HNSCC and neuroblastoma models, with characteristic remodeling of the tumor microenvironment documented in all model systems. scRNA-seq analysis implicated upregulation of innate immune responses and antigen presentation in tumor cells and the myeloid cell compartment as critical early events. This analysis also highlighted the potential importance of the autonomic nervous system in the governance of inflammatory processes. The data indicate that the targeting of multiple pathways and mechanisms of action can result in substantial synergistic antitumor effects and suggest follow-up in the neoadjuvant setting may be warranted.


Tumor Microenvironment , Animals , Mice , Tumor Microenvironment/immunology , Cell Line, Tumor , Neuroblastoma/immunology , Neuroblastoma/therapy , Neuroblastoma/pathology , Female , Humans , Immunomodulation , Mice, Inbred C57BL
6.
Nature ; 623(7988): 820-827, 2023 Nov.
Article En | MEDLINE | ID: mdl-37938771

The majority of oncogenic drivers are intracellular proteins, constraining their immunotherapeutic targeting to mutated peptides (neoantigens) presented by individual human leukocyte antigen (HLA) allotypes1. However, most cancers have a modest mutational burden that is insufficient for generating responses using neoantigen-based therapies2,3. Neuroblastoma is a paediatric cancer that harbours few mutations and is instead driven by epigenetically deregulated transcriptional networks4. Here we show that the neuroblastoma immunopeptidome is enriched with peptides derived from proteins essential for tumorigenesis. We focused on targeting the unmutated peptide QYNPIRTTF discovered on HLA-A*24:02, which is derived from the neuroblastoma-dependency gene and master transcriptional regulator PHOX2B. To target QYNPIRTTF, we developed peptide-centric chimeric antigen receptors (PC-CARs) through a counter panning strategy using predicted potentially cross-reactive peptides. We further proposed that PC-CARs can recognize peptides on additional HLA allotypes when presenting a similar overall molecular surface. Informed by our computational modelling results, we show that PHOX2B PC-CARs also recognize QYNPIRTTF presented by HLA-A*23:01, the most common non-A2 allele in people with African ancestry. Finally, we demonstrate potent and specific killing of neuroblastoma cells expressing these HLAs in vitro and complete tumour regression in mice. These data suggest that PC-CARs have the potential to expand the pool of immunotherapeutic targets to include non-immunogenic intracellular oncoproteins and allow targeting through additional HLA allotypes in a clinical setting.


Antigens, Neoplasm , Neuroblastoma , Oncogene Proteins , Peptides , Receptors, Chimeric Antigen , Animals , Humans , Mice , Africa/ethnology , Alleles , Amino Acid Sequence , Carcinogenesis , Cross Reactions , HLA-A Antigens/chemistry , HLA-A Antigens/immunology , Neuroblastoma/genetics , Neuroblastoma/immunology , Neuroblastoma/therapy , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/immunology , Peptides/antagonists & inhibitors , Peptides/chemistry , Peptides/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use
8.
Oncoimmunology ; 11(1): 2146860, 2022.
Article En | MEDLINE | ID: mdl-36479153

Tumor-associated macrophages (TAM) and cancer-associated fibroblasts (CAF) and their precursor mesenchymal stromal cells (MSC) are often detected together in tumors, but how they cooperate is not well understood. Here, we show that TAM and CAF are the most abundant nonmalignant cells and are present together in untreated human neuroblastoma (NB) tumors that are also poorly infiltrated with T and natural killer (NK) cells. We then show that MSC and CAF-MSC harvested from NB tumors protected human monocytes (MN) from spontaneous apoptosis in an interleukin (IL)-6 dependent mechanism. The interactions of MN and MSC with NB cells resulted in a significant induction or increase in the expression of several pro-tumorigenic cytokines/chemokines (TGF-ß1, MCP-1, IL-6, IL-8, and IL-4) but not of anti-tumorigenic cytokines (TNF-α, IL-12) by MN or MSC, while also inducing cytokine expression in quiescent NB cells. We then identified a TGF-ß1/IL-6 pathway where TGF-ß1 stimulated the expression of IL-6 in NB cells and MSC, promoting TAM survival. Evidence for the contribution of TAM and MSC to the activation of this pathway was then provided in xenotransplanted NB tumors and patients with primary tumors by demonstrating a direct correlation between the presence of CAF and p-SMAD2 and p-STAT3. The data highlight a new mechanism of interaction between TAM and CAF supporting their pro-tumorigenic function in cancer.


Fibroblasts , Interleukin-6 , Macrophages , Neuroblastoma , Transforming Growth Factor beta1 , Humans , Neuroblastoma/immunology , Fibroblasts/immunology , Macrophages/immunology , Animals
9.
Genes Immun ; 23(3-4): 129-140, 2022 06.
Article En | MEDLINE | ID: mdl-35525858

Neuroblastoma is the most common extracranial childhood solid tumor. The majority of high-risk neuroblastoma is resistant/refractory to the current high intensity therapy. Neuroblastoma lacks classical HLA Class I expression and exhibits low mutation burden, allowing neuroblastoma cells to evade CD8+ T cell-mediated immunity. Neuroblastoma cells do not express PD-L1, and tumor-associated macrophages are the predominant PD-L1+ cells in the tumor. In this study, we performed gene expression profiling and survival analyses on large neuroblastoma datasets to address the prognostic effect of PD-L1 gene expression and the possible involvement of the SLAMF7 pathway in the anti-neuroblastoma immunity. High-level expression of PD-L1 was found significantly associated with better outcome of high-risk neuroblastoma patients; two populations of PD-1+ PD-L1+ macrophages could be present in high-risk tumors with PD-1/PD-L1 ratios, ≈1 and >1. Patients with the PD-1/PD-L1 ratio >1 tumor showed inferior survival. High-level co-expression of SLAMF7 and SH2D1B was significantly associated with better survival of the high-risk neuroblastoma patients. Together, this study supports the hypothesis that macrophages are important effector cells in the anti-high-risk neuroblastoma immunity, that PD-1 blockade therapy can be beneficial to the high-risk neuroblastoma subset with the PD-1/PD-L1 expression ratio >1, and that SLAMF7 is a new therapeutic target of high-risk neuroblastoma.


B7-H1 Antigen , Macrophages , Neuroblastoma , B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes , Humans , Macrophages/immunology , Neuroblastoma/genetics , Neuroblastoma/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Signaling Lymphocytic Activation Molecule Family/genetics , Tumor Escape
10.
Cancer Immunol Immunother ; 71(1): 153-164, 2022 Jan.
Article En | MEDLINE | ID: mdl-34043024

Neuroblastoma (NBL) accounts for a disproportionate number of deaths among childhood malignancies despite intensive multimodal therapy that includes antibody targeting disialoganglioside GD2, a NBL antigen. Unfortunately, resistance to anti-GD2 immunotherapy is frequent and we aimed to investigate mechanisms of resistance in NBL. GD2 expression was quantified by flow cytometry and anti-GD2 antibody internalization was measured using real-time microscopy in 20 human NBL cell lines. Neutrophil-mediated antibody-dependent cellular cytotoxicity (ADCC) assays were performed on a subset of the cell lines (n = 12), and results were correlated with GD2 expression and antibody internalization. GD2 was expressed on 19 of 20 NBL cell lines at variable levels, and neutrophil-mediated ADCC was observed only in GD2-expressing cell lines. We found no correlation between level of GD2 expression and sensitivity to neutrophil-mediated ADCC, suggesting that GD2 expression of many cell lines was above a threshold required for maximal ADCC, such that expression level could not be used to predict subsequent cytotoxicity. Instead, anti-GD2 antibody internalization, a process that occurred universally but differentially across GD2-expressing NBL cell lines, was inversely correlated with ADCC. Treatment with endocytosis inhibitors EIPA, chlorpromazine, MBCD, and cytochalasin-D showed potential to inhibit antibody internalization; however, only MBCD resulted in significantly increased sensitivity to neutrophil-mediated ADCC in 4 of 4 cell lines in vitro. Our data suggest that antibody internalization may represent a novel mechanism of immunotherapy escape by NBL and provide proof-of-principle that targeting pathways involved in antibody internalization may improve the efficacy of anti-GD2 immunotherapies.


Antibodies/chemistry , Drug Resistance , Gangliosides/chemistry , Immunotherapy/methods , Neuroblastoma/immunology , Neuroblastoma/therapy , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Endocytosis , Flow Cytometry , Gangliosides/immunology , Humans , Immunologic Factors , Killer Cells, Natural/immunology , Neutrophils/metabolism
11.
Cancer Immunol Immunother ; 71(1): 71-83, 2022 Jan.
Article En | MEDLINE | ID: mdl-34023958

Immunotherapy with anti-GD2 monoclonal antibodies (mAbs) provides some benefits for patients with neuroblastoma (NB). However, the therapeutic efficacy remains limited, and treatment is associated with significant neuropathic pain. Targeting O-acetylated GD2 (OAcGD2) by 8B6 mAb has been proposed to avoid pain by more selective tumor cell targeting. Thorough understanding of its mode of action is necessary to optimize this treatment strategy. Here, we found that 8B6-mediated antibody-dependent cellular phagocytosis (ADCP) performed by macrophages is a key effector mechanism. But efficacy is limited by upregulation of CD47 expression on neuroblastoma cells in response to OAcGD2 mAb targeting, inhibiting 8B6-mediated ADCP. Antibody specific for the CD47 receptor SIRPα on macrophages restored 8B6-induced ADCP of CD47-expressing NB cells and improved the antitumor activity of 8B6 mAb therapy. These results identify ADCP as a critical mechanism for tumor cytolysis by anti-disialoganglioside mAb and support a combination with SIRPα blocking agents for effective neuroblastoma therapy.


Antibodies, Monoclonal/chemistry , Antigens, Differentiation/chemistry , Neuroblastoma/immunology , Phagocytosis , Receptors, Immunologic/chemistry , Animals , Antibodies/chemistry , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents, Immunological/pharmacology , CD47 Antigen/biosynthesis , Cell Line, Tumor , Flow Cytometry , Gangliosides/chemistry , Humans , Immunotherapy/methods , Macrophages/metabolism , Mice , Microscopy, Fluorescence , Neuroblastoma/metabolism , Up-Regulation
13.
J Clin Oncol ; 40(4): 335-344, 2022 02 01.
Article En | MEDLINE | ID: mdl-34871104

PURPOSE: We evaluated whether combining a humanized antidisialoganglioside monoclonal antibody (hu14.18K322A) throughout therapy improves early response and outcomes in children with newly diagnosed high-risk neuroblastoma. PATIENTS AND METHODS: We conducted a prospective, single-arm, three-stage, phase II clinical trial. Six cycles of induction chemotherapy were coadministered with hu14.18K322A, granulocyte-macrophage colony-stimulating factor (GM-CSF), and low-dose interleukin-2 (IL-2). The consolidation regimen included busulfan and melphalan. When available, an additional cycle of parent-derived natural killer cells with hu14.18K322A was administered during consolidation (n = 31). Radiation therapy was administered at the end of consolidation. Postconsolidation treatment included hu14.18K322A, GM-CSF, IL-2, and isotretinoin. Early response was assessed after the first two cycles of induction therapy. End-of-induction response, event-free survival (EFS), and overall survival (OS) were evaluated. RESULTS: Sixty-four patients received hu14.18K322A with induction chemotherapy. This regimen was well tolerated, with continuous infusion narcotics. Partial responses (PRs) or better after the first two chemoimmunotherapy cycles occurred in 42 of 63 evaluable patients (66.7%; 95% CI, 55.0 to 78.3). Primary tumor volume decreased by a median of 75% (range, 100% [complete disappearance]-5% growth). Median peak hu14.18K322A serum levels in cycle one correlated with early response to therapy (P = .0154, one-sided t-test). Sixty of 62 patients (97%) had an end-of-induction partial response or better. No patients experienced progressive disease during induction. The 3-year EFS was 73.7% (95% CI, 60.0 to 83.4), and the OS was 86.0% (95% CI, 73.8 to 92.8), respectively. CONCLUSION: Adding hu14.18K322A to induction chemotherapy improved early objective responses, significantly reduced tumor volumes in most patients, improved end-of-induction response rates, and yielded an encouraging 3-year EFS. These results, if validated in a larger study, may be practice changing.


Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Interleukin-2/therapeutic use , Neuroblastoma/drug therapy , Adolescent , Age Factors , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Female , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Induction Chemotherapy , Infant , Interleukin-2/adverse effects , Male , Neuroblastoma/immunology , Neuroblastoma/mortality , Neuroblastoma/pathology , Progression-Free Survival , Prospective Studies , Risk Assessment , Risk Factors , Time Factors , Tumor Burden/drug effects
14.
Front Immunol ; 12: 738958, 2021.
Article En | MEDLINE | ID: mdl-34721405

Immune checkpoint blockade (ICB) relieves CD8+ T-cell exhaustion in most mutated tumors, and TCF-1 is implicated in converting progenitor exhausted cells to functional effector cells. However, identifying mechanisms that can prevent functional senescence and potentiate CD8+ T-cell persistence for ICB non-responsive and resistant tumors remains elusive. We demonstrate that targeting Cbx3/HP1γ in CD8+ T cells augments transcription initiation and chromatin remodeling leading to increased transcriptional activity at Lef1 and Il21r. LEF-1 and IL-21R are necessary for Cbx3/HP1γ-deficient CD8+ effector T cells to persist and control ovarian cancer, melanoma, and neuroblastoma in preclinical models. The enhanced persistence of Cbx3/HP1γ-deficient CD8+ T cells facilitates remodeling of the tumor chemokine/receptor landscape ensuring their optimal invasion at the expense of CD4+ Tregs. Thus, CD8+ T cells heightened effector function consequent to Cbx3/HP1γ deficiency may be distinct from functional reactivation by ICB, implicating Cbx3/HP1γ as a viable cancer T-cell-based therapy target for ICB resistant, non-responsive solid tumors.


CD8-Positive T-Lymphocytes/metabolism , Chromobox Protein Homolog 5/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Melanoma, Experimental/metabolism , Neuroblastoma/metabolism , Ovarian Neoplasms/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation , Cell Line, Tumor , Chromobox Protein Homolog 5/genetics , Chromosomal Proteins, Non-Histone/genetics , Coculture Techniques , Female , Gene Expression Regulation, Neoplastic , Immunotherapy, Adoptive , Interleukin-21 Receptor alpha Subunit/genetics , Interleukin-21 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoid Enhancer-Binding Factor 1/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuroblastoma/genetics , Neuroblastoma/immunology , Neuroblastoma/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Burden , Tumor Microenvironment
15.
J Hematol Oncol ; 14(1): 191, 2021 11 12.
Article En | MEDLINE | ID: mdl-34772439

The outcome of patients affected by high-risk or metastatic neuroblastoma (NB) remains grim, with ≥ 50% of the children experiencing relapse or progression of the disease despite multimodal, intensive treatment. In order to identify new strategies to improve the overall survival and the quality of life of these children, we recently developed and optimized a third-generation GD2-specific chimeric antigen receptor (CAR) construct, which is currently under evaluation in our Institution in a phase I/II clinical trial (NCT03373097) enrolling patients with relapsed/refractory NB. We observed that our CAR T-cells are able to induce marked tumor reduction and even achieve complete remission with a higher efficiency than that of other CAR T-cells reported in previous studies. However, often responses are not sustained and relapses occur. Here, we demonstrate for the first time a mechanism of resistance to GD2.CAR T-cell treatment, showing how polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) increase in the peripheral blood (PB) of NB patients after GD2.CAR T-cell treatment in case of relapse and loss of response. In vitro, isolated PMN-MDSC demonstrate to inhibit the anti-tumor cytotoxicity of different generations of GD2.CAR T-cells. Gene-expression profiling of GD2.CAR T-cells "conditioned" with PMN-MDSC shows downregulation of genes involved in cell activation, signal transduction, inflammation and cytokine/chemokine secretion. Analysis of NB gene-expression dataset confirms a correlation between expression of these genes and patient outcome. Moreover, in patients treated with GD2.CAR T-cells, the frequency of circulating PMN-MDSC inversely correlates with the levels of GD2.CAR T-cells, resulting more elevated in patients who did not respond or lost response to the treatment. The presence and the frequency of PMN-MDSC in PB of high-risk and metastatic NB represents a useful prognostic marker to predict the response to GD2.CAR T-cells and other adoptive immunotherapy. This study underlines the importance of further optimization of both CAR T-cells and clinical trial in order to target elements of the tumor microenvironment.


Immunotherapy, Adoptive , Myeloid-Derived Suppressor Cells/immunology , Neuroblastoma/therapy , Humans , Immunotherapy, Adoptive/methods , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neuroblastoma/immunology , Neuroblastoma/pathology , Treatment Outcome
16.
Immunol Lett ; 240: 31-40, 2021 12.
Article En | MEDLINE | ID: mdl-34600949

Neuroblastoma (NB) has high morality rates and is the most common malignant tumor found in children. High aggregation of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment results in immunosuppression and affects therapeutic effectiveness. At present, doxorubicin (DOX) and dopamine (DA) are the specific drugs used to selectively remove or mature MDSCs. The aim of the present study was to explore the feasibility and underlying mechanism of targeting elimination of MDSCs via DOX or DA administration to alleviate tumor immunosuppression in NB. In the present study, a BALB/c tumor-bearing mouse model was established, and mice were grouped into the control, DOX2.5, DOX5 and DA50 mg/kg groups. DOX or DA were injected intravenously on days 7 and 12 after inoculation, following which the parameters related to the signal transducer and activator of transcription (STAT) pathway in MDSCs, the proportion of MDSCs, T cell infiltration, programmed death-1 (PD-1) on the surface of T cells, the number of regulatory T cells (Tregs), polarization of tumor-related macrophages (TAMs) and tumor growth were compared between the groups on days 14, 17 and 23 after inoculation. The results demonstrated that following DOX or DA administration, STAT1/phosphorylated (p)-STAT1 decreased, whereas STAT3/p-STAT3, STAT5/p-STAT5 and STAT6/p-STAT6 increased, which was accompanied by a decrease in the MDSC proportion in each experimental group. Simultaneously, T cell infiltration in tumors was increased, whereas expression of PD-1, the number of Tregs, TAM polarization and tumor growth were inhibited. The most significant findings were observed in the DOX2.5 mg/kg group. To conclude, low dose DOX or DA administration could effectively regulate the STAT pathway to eliminate MDSCs, alleviate immunosuppression and improve the immune response against NB tumor cells.


Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/immunology , Neuroblastoma/immunology , STAT Transcription Factors/immunology , Signal Transduction/immunology , Tumor Microenvironment/immunology , Animals , Mice , Mice, Inbred BALB C
17.
Cancer Control ; 28: 10732748211033751, 2021.
Article En | MEDLINE | ID: mdl-34569303

BACKGROUND: Neuroblastoma (NBL) is the most common extracranial solid tumor in childhood, and patients with high-risk neuroblastoma had a relatively poor prognosis despite multimodal treatment. To improve immunotherapy efficacy in neuroblastoma, systematic profiling of the immune landscape in neuroblastoma is an urgent need. METHODS: RNA-seq and according clinical information of neuroblastoma were downloaded from the TARGET database and GEO database (GSE62564). With an immune-related-gene set obtained from the ImmPort database, Immune-related Prognostic Gene Pairs for Neuroblastoma (IPGPN) for overall survival (OS) were established with the TARGET-NBL cohort and then verified with the GEO-NBL cohort. Immune cell infiltration analysis was subsequently performed. The integrated model was established with IPGPN and clinicopathological parameters. Immune cell infiltration was analyzed with the XCELL algorithm. Functional enrichment analysis was performed with clusterProfiler package in R. RESULTS: Immune-related Prognostic Gene Pairs for Neuroblastoma was successfully established with seven immune-related gene pairs (IGPs) involving 13 unique genes in the training cohort. In the training cohort, IPGPN successfully stratified neuroblastoma patients into a high and low immune-risk groups with different OS (HR=3.92, P = 2 × 10-8) and event-free survival (HR=3.66, P=2 × 10-8). ROC curve analysis confirmed its predictive power. Consistently, high IPGPN also predicted worse OS (HR=1.84, P = .002) and EFS in validation cohort (HR=1.38, P = .06) Moreover, higher activated dendritic cells, M1 macrophage, Th1 CD4+, and Th2 CD4+ T cell enrichment were evident in low immune-risk group. Further integrating IPGPN with age and stage demonstrated improved predictive performance than IPGPN alone. CONCLUSION: Herein, we presented an immune landscape with IPGPN for prognosis prediction in neuroblastoma, which complements the present understanding of the immune signature in neuroblastoma.


Neuroblastoma/genetics , Neuroblastoma/pathology , Algorithms , Databases, Genetic , Female , Gene Expression Profiling , Humans , Infant , Kaplan-Meier Estimate , Male , Neoplasm Staging , Neuroblastoma/immunology , Neuroblastoma/mortality , Prognosis , Risk Factors , Tumor Microenvironment
18.
Front Immunol ; 12: 668307, 2021.
Article En | MEDLINE | ID: mdl-34489927

Management for high-risk neuroblastoma (NBL) has included autologous hematopoietic stem cell transplant (HSCT) and anti-GD2 immunotherapy, but survival remains around 50%. The aim of this study was to determine if allogeneic HSCT could serve as a platform for inducing a graft-versus-tumor (GVT) effect against NBL with combination immunocytokine and NK cells in a murine model. Lethally irradiated C57BL/6 (B6) x A/J recipients were transplanted with B6 bone marrow on Day +0. On day +10, allogeneic HSCT recipients were challenged with NXS2, a GD2+ NBL. On days +14-16, mice were treated with the anti-GD2 immunocytokine hu14.18-IL2. In select groups, hu14.18-IL2 was combined with infusions of B6 NK cells activated with IL-15/IL-15Rα and CD137L ex vivo. Allogeneic HSCT alone was insufficient to control NXS2 tumor growth, but the addition of hu14.18-IL2 controlled tumor growth and improved survival. Adoptive transfer of ex vivo CD137L/IL-15/IL-15Rα activated NK cells with or without hu14.18-IL2 exacerbated lethality. CD137L/IL-15/IL-15Rα activated NK cells showed enhanced cytotoxicity and produced high levels of TNF-α in vitro, but induced cytokine release syndrome (CRS) in vivo. Infusing Perforin-/- CD137L/IL-15/IL-15Rα activated NK cells had no impact on GVT, whereas TNF-α-/- CD137L/IL-15/IL-15Rα activated NK cells improved GVT by decreasing peripheral effector cell subsets while preserving tumor-infiltrating lymphocytes. Depletion of Ly49H+ NK cells also improved GVT. Using allogeneic HSCT for NBL is a viable platform for immunocytokines and ex vivo activated NK cell infusions, but must be balanced with induction of CRS. Regulation of TNFα or activating NK subsets may be needed to improve GVT effects.


Antineoplastic Agents, Immunological/pharmacology , Cytokines/pharmacology , Gangliosides/antagonists & inhibitors , Graft vs Tumor Effect , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Killer Cells, Natural/drug effects , Killer Cells, Natural/transplantation , Lymphocyte Activation/drug effects , Neuroblastoma/therapy , Animals , Cell Line, Tumor , Combined Modality Therapy , Female , Gangliosides/immunology , Gangliosides/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/immunology , Neuroblastoma/metabolism , Neuroblastoma/pathology
19.
J Immunol Methods ; 498: 113135, 2021 11.
Article En | MEDLINE | ID: mdl-34478717

In recent years, there has been an expansion in the use of flow cytometry (FC) immunophenotyping in the diagnosis and monitoring of childhood solid neoplasms. Neuroblastoma (NB), in turn, is the most common extracranial solid tumor in childhood. In the present study, we sought to compare FC and anatomopathological examination (PA) / immunohistochemistry (IHC) of children diagnosed or suspected with NB. The median age was 59 months (minimum 0; maximum 325 months), of these 12 were male (57.1%, 12/21). Forty-eight samples (27 bone marrow (BM), 10 peripheral blood (PB), 8 primary tumors (PT) and 2 liver nodules (HN) and 1 rib fragment (RF)) from 21 patients were evaluated. Twenty-nine samples were from patients with clinical suspicion while 19 samples were from patients with previously confirmed diagnosis. Thirteen samples (7 BM, 5 PT and 1 HN) presented NB when analyzed in FC while 8 (3 BM and 5 PT) samples were positive for NB in the PA/IHC. They were concordant in 88.9% of the cases. No NB cells were identified in any PB. Considering the PA as the gold standard, the FC obtained a sensitivity of 100%, a specificity of 86%, a positive predictive value of 67% and a negative predictive value of 100%. This study demonstrates that FC can be used as a methodology for diagnosis and assessment of NB involvement. In addition, FC has the advantage of allowing a quick diagnosis and accurate classification of the disease, and can also assist in monitoring the treatment.


Biomarkers, Tumor/analysis , Flow Cytometry , Immunohistochemistry , Neuroblastoma/diagnosis , Biomarkers, Tumor/genetics , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Neuroblastoma/chemistry , Neuroblastoma/genetics , Neuroblastoma/immunology , Ploidies , Predictive Value of Tests , Reproducibility of Results , Time Factors , Workflow
20.
J Immunol Methods ; 498: 113124, 2021 11.
Article En | MEDLINE | ID: mdl-34425081

Commercially available antibodies that bind to the human muscle acetylcholine receptor (ACHR) have been validated previously for flow cytometric use (Keefe et al., 2009; Leite et al., 2008; Lozier et al., 2015). Despite a multitude of commercially available antibodies to other nicotinic ACHRs, validation in a wide variety of immunoassay formats is lacking; when studied, a large proportion of these antibodies have been deemed not fit for most research purposes (Garg and Loring, 2017). We have recently described a flow cytometric immunomodulation assay for the diagnosis of Autoimmune Autonomic Ganglionopathy (AAG) (Urriola et al., 2021) that utilises the monoclonal antibody mab35(Urriola et al., 2021) which is specific for ganglionic ACHR (gnACHR) that contain α3 subunits (Vernino et al., 1998). Other fluorescent ligands for α3-gnACHR have not been validated for flow cytometric use. We investigated 7 commercially sourced antibodies and 3 synthetic fluorescent novel conotoxins purported to specifically bind to the extracellular domains of the gnACHR, and compared the results to staining by mab35, using flow cytometry with the neuroblastoma cell line IMR-32. We also evaluated the degree of non-specific binding by depleting the cell membrane of the relevant acetylcholine receptor with a pre-incubation step involving the serum from a patient with Autoimmune Autonomic Ganglionopathy containing pathogenic antibodies to the ganglionic acetylcholine receptor. None of the assessed conotoxins, and only one antibody (mab35) was found to perform adequately in flow cytometric staining of the native ganglionic acetylcholine receptor.


Antibodies, Monoclonal/immunology , Autoimmune Diseases of the Nervous System/diagnosis , Autonomic Nervous System Diseases/diagnosis , Conotoxins/chemistry , Flow Cytometry , Fluorescent Dyes/chemistry , Ganglia, Autonomic/immunology , Neuroblastoma/immunology , Receptors, Cholinergic/analysis , Antibody Specificity , Autoantibodies/blood , Autoimmune Diseases of the Nervous System/immunology , Autonomic Nervous System Diseases/immunology , Cell Line, Tumor , Epitopes , Humans , Predictive Value of Tests , Receptors, Cholinergic/immunology
...