Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Mol Biol Rep ; 51(1): 408, 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38460043

ABSTRACT

BACKGROUND: To describe an oncolytic adenovirus (OAd) encoding SP-SA-E7-4-1BBL that is capable of inducing tumor regression in therapeutic assays. Herein, we tested whether the antitumor effect is given by the induction of a tumor-specific immune response, as well as the minimum dose needed to elicit antitumor protection and monitor the OAd biodistribution over time. METHODS AND RESULTS: C57BL/6 mice (n = 5) per group were immunized twice with OAds encoding SP-SA-E7-4-1BBL, SA-E7-4-1BBL, or SP-SA-4-1BBL and challenged with TC-1 cancer cells. The DNA construct SP-SA-E7-4-1BBL was employed as a control via biolistic or PBS injection. Groups without tumor development at 47 days were rechallenged with TC-1 cells, and follow-up lasted until day 90. The minimum dose of OAd to induce the antitumor effect was established by immunization using serial dilution doses. The cytometry bead assay and the ELISpot assay were used to evaluate cytokine release in response to ex vivo antigenic stimulation. The distribution profile of the OAd vaccine was evaluated in the different organs by histological, immunohistochemical and qPCR analyses. The OAd SP-SA-E7-4-1BBL-immunized mice did not develop tumors even in a rechallenge. A protective antitumor effect was observed from a dose that is one hundredth of most reports of adenoviral vaccines. Immunization with OAd increases Interferon-gamma-producing cells in response to antigen stimulation. OAd was detected in tumors over time, with significant morphological changes, contrary to nontumor tissues. CONCLUSIONS: The OAd SP-SA-E7-4-1BBL vaccine confers a prophylactic, safe, long-lasting, and antigen-dependent antitumor effect mediated by a Th1 antitumor immune response.


Subject(s)
Cancer Vaccines , Neoplasms , Animals , Mice , Human papillomavirus 16 , 4-1BB Ligand/genetics , 4-1BB Ligand/pharmacology , Tissue Distribution , Mice, Inbred C57BL , Adenoviridae/genetics , Immunity , Neoplasms/therapy
2.
Cancer Immunol Immunother ; 72(9): 3029-3043, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37310433

ABSTRACT

Targeting co-stimulatory receptors promotes the activation and effector functions of anti-tumor lymphocytes. 4-1BB (CD137/TNFSF9), a member of the tumor necrosis factor receptor superfamily (TNFR-SF), is a potent co-stimulatory receptor that plays a prominent role in augmenting effector functions of CD8+ T cells, but also CD4+ T cells and NK cells. Agonistic antibodies against 4-1BB have entered clinical trials and shown signs of therapeutic efficacy. Here, we have used a T cell reporter system to evaluate various formats of 4-1BBL regarding their capacity to functionally engage its receptor. We found that a secreted 4-1BBL ectodomain harboring a trimerization domain derived from human collagen (s4-1BBL-TriXVIII) is a strong inducer of 4-1BB co-stimulation. Similar to the 4-1BB agonistic antibody urelumab, s4-1BBL-TriXVIII is very potent in inducing CD8+ and CD4+ T cell proliferation. We provide first evidence that s4-1BBL-TriXVIII can be used as an effective immunomodulatory payload in therapeutic viral vectors. Oncolytic measles viruses encoding s4-1BBL-TriXVIII significantly reduced tumor burden in a CD34+ humanized mouse model, whereas measles viruses lacking s4-1BBL-TriXVIII were not effective. Natural soluble 4-1BB ligand harboring a trimerization domain might have utility in tumor therapy especially when delivered to tumor tissue as systemic administration might induce liver toxicity.


Subject(s)
4-1BB Ligand , CD8-Positive T-Lymphocytes , Mice , Animals , Humans , 4-1BB Ligand/genetics , Immunomodulating Agents , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Receptors, Tumor Necrosis Factor , Killer Cells, Natural
3.
Cancer Immunol Immunother ; 71(6): 1531-1543, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34661709

ABSTRACT

INTRODUCTION: Epstein-Barr virus (EBV) is associated with nasopharyngeal carcinoma (NPC), and provides a target for a dendritic cell (DC) vaccine. CD137 ligand (CD137L) expressed on antigen presenting cells, costimulates CD137-expressing T cells, and reverse CD137L signaling differentiates monocytes to CD137L-DC, a type of DC, which is more potent than classical DC in stimulating T cells. METHODS: In this phase I study, patients with locally recurrent or metastatic NPC were administered CD137L-DC pulsed with EBV antigens (CD137L-DC-EBV-VAX). RESULTS: Of the 12 patients treated, 9 received full 7 vaccine doses with a mean administered cell count of 23.9 × 106 per dose. Treatment was well tolerated with only 4 cases of grade 1 related adverse events. A partial response was obtained in 1 patient, and 4 patients are still benefitting from a progression free survival (PFS) of currently 2-3 years. The mean pre-treatment neutrophil: lymphocyte ratio was 3.4 and a value of less than 3 was associated with prolonged median PFS. Progressors were characterized by a high frequency of naïve T cells but a low frequency of CD8+ effector T cells while patients with a clinical benefit (CB) had a high frequency of memory T cells. Patients with CB had lower plasma EBV DNA levels, and a reduction after vaccination. CONCLUSION: CD137L-DC-EBV-VAX was well tolerated. The use of CD137L-DC-EBV-VAX is demonstrated to be safe. Consistent results were obtained from all 12 patients, indicating that CD137L-DC-EBV-VAX induces an anti-EBV and anti-NPC immune response, and warranting further studies in patients post effective chemotherapy. PRECIS: The first clinical testing of CD137L-DC, a new type of monocyte-derived DC, finds that CD137L-DC are safe, and that they can induce an immune response against Epstein-Barr virus-associated nasopharyngeal carcinoma that leads to tumor regression or prevents tumor progression.


Subject(s)
Epstein-Barr Virus Infections , Nasopharyngeal Neoplasms , 4-1BB Ligand/genetics , Dendritic Cells , Herpesvirus 4, Human , Humans , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/therapy
4.
Pharmacol Res ; 175: 106034, 2022 01.
Article in English | MEDLINE | ID: mdl-34915126

ABSTRACT

Renal carcinoma progresses aggressively in patients with metastatic disease while curative strategies are limited. Here, we constructed a recombinant non-replicating adenovirus (Ad) vaccine encoding an immune activator, CD137L, and a tumor antigen, CAIX, for treating renal carcinoma. In a subcutaneous tumor model, tumor growth was significantly suppressed in the Ad-CD137L/CAIX vaccine group compared with the single vaccine group. The induction and maturity of CD11C+ and CD8+CD11C+ dendritic cell (DC) subsets were promoted in Ad-CD137L/CAIX co-immunized mice. Furthermore, the Ad-CD137L/CAIX vaccine elicited stronger tumor-specific multifunctional CD8+ T cell immune responses as demonstrated by increased proliferation and cytolytic function of CD8+ T cells. Notably, depletion of CD8+ T cells greatly compromised the effective protection provided by Ad-CD137L/CAIX vaccine, suggesting an irreplaceable role of CD8+ T cells for the immunopotency of the vaccine. In both lung metastatic and orthotopic models, Ad-CD137L/CAIX vaccine treatment significantly decreased tumor metastasis and progression and increased the induction of tumor-specific multifunctional CD8+ T cells, in contrast to treatment with the Ad-CAIX vaccine alone. The Ad-CD137L/CAIX vaccine also augmented the tumor-specific multifunctional CD8+ T cell immune response in both orthotopic and metastatic models. These results indicated that Ad-CD137L/CAIX vaccine elicited a potent anti-tumor activity by inducing CD8+DC-mediated multifunctional CD8+ T cell immune responses. The potential strategy of CD137L-based vaccine might be served as a novel treatment for renal carcinoma or other malignant tumors.


Subject(s)
4-1BB Ligand/genetics , Adenovirus Vaccines/therapeutic use , Carbonic Anhydrase IX/genetics , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/therapy , Lung Neoplasms/therapy , 4-1BB Ligand/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Carbonic Anhydrase IX/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Dendritic Cells/immunology , Female , HEK293 Cells , Humans , Interleukin-6/blood , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mice, Inbred BALB C
5.
Aging (Albany NY) ; 13(17): 21571-21586, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34517345

ABSTRACT

Early metastasis of pancreatic cancer (PC) leads to high mortality, and the underlying mechanism of metastasis remains unclear. Tumor necrosis factor superfamily member 9 (TNFSF9) is associated with poor prognosis in PC. Here, we investigated the effect of TNFSF9 on PC proliferation and apoptosis, and focused on the effect of TNFSF9 on PC metastasis and its potential mechanism. We found that TNFSF9 promotes PC metastasis in vivo and in vitro, and may be partially dependent on the Wnt/Snail signaling pathway. In addition, TNFSF9 also regulates the release of cytokines IL-10 and transforming growth factor-ß (TGF-ß) in pancreatic cancer cells through Wnt signaling to induce the M2 polarization of macrophages and promote the migration of PC cells. Overall, our study found that TNFSF9 may directly promote PC metastasis or indirectly promote PC metastasis through macrophage M2 polarization. Our study provides a new costimulatory target for the treatment of PC.


Subject(s)
4-1BB Ligand/metabolism , Cell Proliferation , Macrophages/physiology , Pancreatic Neoplasms/pathology , 4-1BB Ligand/genetics , Animals , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , Interleukin-10/metabolism , Lymphatic Metastasis , Macrophage Activation , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/metabolism , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Xenograft Model Antitumor Assays
6.
Cancer Immunol Immunother ; 70(9): 2701-2719, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34244816

ABSTRACT

Recombinant agonists that activate co-stimulatory and cytokine receptors have shown limited clinical anticancer utility, potentially due to narrow therapeutic windows, the need for coordinated activation of co-stimulatory and cytokine pathways and the failure of agonistic antibodies to recapitulate signaling by endogenous ligands. RTX-240 is a genetically engineered red blood cell expressing 4-1BBL and IL-15/IL-15Rα fusion (IL-15TP). RTX-240 is designed to potently and simultaneously stimulate the 4-1BB and IL-15 pathways, thereby activating and expanding T cells and NK cells, while potentially offering an improved safety profile through restricted biodistribution. We assessed the ability of RTX-240 to expand and activate T cells and NK cells and evaluated the in vivo efficacy, pharmacodynamics and tolerability using murine models. Treatment of PBMCs with RTX-240 induced T cell and NK cell activation and proliferation. In vivo studies using mRBC-240, a mouse surrogate for RTX-240, revealed biodistribution predominantly to the red pulp of the spleen, leading to CD8 + T cell and NK cell expansion. mRBC-240 was efficacious in a B16-F10 melanoma model and led to increased NK cell infiltration into the lungs. mRBC-240 significantly inhibited CT26 tumor growth, in association with an increase in tumor-infiltrating proliferating and cytotoxic CD8 + T cells. mRBC-240 was tolerated and showed no evidence of hepatic injury at the highest feasible dose, compared with a 4-1BB agonistic antibody. RTX-240 promotes T cell and NK cell activity in preclinical models and shows efficacy and an improved safety profile. Based on these data, RTX-240 is now being evaluated in a clinical trial.


Subject(s)
4-1BB Ligand/genetics , Cell- and Tissue-Based Therapy , Erythrocytes/metabolism , Gene Expression , Genetic Therapy , Interleukin-15/genetics , 4-1BB Ligand/metabolism , Animals , Cell- and Tissue-Based Therapy/methods , Erythroid Precursor Cells/metabolism , Female , Flow Cytometry , Genes, Reporter , Genetic Engineering , Genetic Therapy/methods , Humans , Interleukin-15/metabolism , Mice , Models, Animal , Protein Binding , Treatment Outcome , Xenograft Model Antitumor Assays
7.
Nat Commun ; 12(1): 2637, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976146

ABSTRACT

Checkpoint inhibitors and T-cell therapies have highlighted the critical role of T cells in anti-cancer immunity. However, limitations associated with these treatments drive the need for alternative approaches. Here, we engineer red blood cells into artificial antigen-presenting cells (aAPCs) presenting a peptide bound to the major histocompatibility complex I, the costimulatory ligand 4-1BBL, and interleukin (IL)-12. This leads to robust, antigen-specific T-cell expansion, memory formation, additional immune activation, tumor control, and antigen spreading in tumor models in vivo. The presence of 4-1BBL and IL-12 induces minimal toxicities due to restriction to the vasculature and spleen. The allogeneic aAPC, RTX-321, comprised of human leukocyte antigen-A*02:01 presenting the human papilloma virus (HPV) peptide HPV16 E711-19, 4-1BBL, and IL-12 on the surface, activates HPV-specific T cells and promotes effector function in vitro. Thus, RTX-321 is a potential 'off-the-shelf' in vivo cellular immunotherapy for treating HPV + cancers, including cervical and head/neck cancers.


Subject(s)
Antigen-Presenting Cells/transplantation , Cell Engineering/methods , Erythrocytes/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , 4-1BB Ligand/genetics , 4-1BB Ligand/immunology , 4-1BB Ligand/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Line, Tumor , Coculture Techniques , Disease Models, Animal , Erythrocytes/metabolism , Female , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-12/metabolism , Lymphocyte Activation , Neoplasms/immunology , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus E7 Proteins/metabolism , Primary Cell Culture , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Transplantation, Homologous/methods
8.
Oncoimmunology ; 10(1): 1877459, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33643694

ABSTRACT

Rhabdomyosarcoma (RMS) is a heterogeneous soft tissue neoplasm most frequently found in children and adolescents. As the prognosis for recurrent and metastatic RMS remains poor, immunotherapies are hoped to improve quality of life and survival. CD137 is a member of tumor necrosis factor receptor family and a T cell costimulatory molecule which induces potent cellular immune responses that are able to eliminate malignant cells. Therefore, it was puzzling to find expression of CD137 on an RMS tissue microarray by multiplex staining. CD137 is not only expressed by infiltrating T cells but also by malignant RMS cells. Functional in vitro experiments demonstrate that CD137 on RMS cells is being transferred to adjacent antigen-presenting cells by trogocytosis, where it downregulates CD137 ligand, and thereby reduces T cell costimulation which results in reduced killing of RMS cells. The transfer of CD137 and the subsequent downregulation of CD137 ligand is a physiological negative feedback mechanism that is likely usurped by RMS, and may facilitate its escape from immune surveillance. In addition, CD137 signals into RMS cells and induces IL-6 and IL-8 secretion, which are linked to RMS metastasis and poor prognosis. However, the ectopic expression of CD137 on RMS cells is an Achilles' heel that may be utilized for immunotherapy. Natural killer cells expressing an anti-CD137 chimeric antigen receptor specifically kill CD137-expressing RMS cells. Our study implicates ectopic CD137 expression as a pathogenesis mechanism in RMS, and it demonstrates that CD137 may be a novel target for immunotherapy of RMS.


Subject(s)
Quality of Life , Rhabdomyosarcoma , 4-1BB Ligand/genetics , Adolescent , Child , Humans , Immunotherapy , Rhabdomyosarcoma/genetics , T-Lymphocytes , Tumor Necrosis Factor Receptor Superfamily, Member 9
9.
Am J Hematol ; 96(6): 671-679, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33725422

ABSTRACT

The prognosis of relapsed acute lymphoblastic leukemia (ALL) after allogeneic transplantation is dismal when treated with conventional approaches. While single-target CD19 or CD22 chimeric antigen receptor (CAR) T-cell therapy has achieved high complete remission (CR) rates in refractory/relapsed B-ALL, it could not maintain a durable remission in most patients. To prolong relapse-free survival, we sequentially combined CD19 and CD22 CAR-T cells to treat post-transplant relapsed B-ALL patients with both CD19/CD22 antigen expression on lymphoblasts. Patient-derived donor cells were collected to produce CAR-T cells that were transfected by lentiviral vectors encoding second generation CARs composed of CD3ζ and 4-1BB. The second T-cell infusion was scheduled at least 1 month, and usually within 6 months after the first CAR-T treatment. Twenty-seven adult and pediatric patients, including 11 (41%) with extramedullary diseases (EMD), received the first CD19 CAR-T and 23 (85%) achieved CR. Subsequently, 21 out of 27 patients received the second CD22 CAR-T and were followed-up for a median of 19.7 (range, 5.6-27.3) months; 14 cases remained in CR, seven relapsed and two of them died from disease progression; Kaplan-Meier survival analysis showed overall survival and event-free survival rates of 88.5% and 67.5%, respectively, at both 12 months and 18 months. CAR-T associated graft-versus-host disease (GVHD) occurred in 23% of patients, with 8% new-onset acute GVHD and 15% persistent or worsened pre-existing cGVHD before CAR-T. This combination strategy of sequential CD19 and CD22 CAR-T therapy significantly improved the long-term survival in B-ALL patients who relapsed after transplantation.


Subject(s)
Antigens, CD19/immunology , Antigens, Neoplasm/immunology , Immunotherapy, Adoptive/methods , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Salvage Therapy , Sialic Acid Binding Ig-like Lectin 2/immunology , 4-1BB Ligand/genetics , Adolescent , Adult , Allografts , CD3 Complex/genetics , Child , Child, Preschool , Combined Modality Therapy , Cord Blood Stem Cell Transplantation , Female , Follow-Up Studies , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation , Humans , Infant , Kaplan-Meier Estimate , Male , Middle Aged , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Progression-Free Survival , Recurrence , Treatment Outcome , Young Adult
10.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33722906

ABSTRACT

BACKGROUND: Adoptive cell therapy based on the infusion of chimeric antigen receptor (CAR) T cells has shown remarkable efficacy for the treatment of hematologic malignancies. The primary mechanism of action of these infused T cells is the direct killing of tumor cells expressing the cognate antigen. However, understanding why only some T cells are capable of killing, and identifying mechanisms that can improve killing has remained elusive. METHODS: To identify molecular and cellular mechanisms that can improve T-cell killing, we utilized integrated high-throughput single-cell functional profiling by microscopy, followed by robotic retrieval and transcriptional profiling. RESULTS: With the aid of mathematical modeling we demonstrate that non-killer CAR T cells comprise a heterogeneous population that arise from failure in each of the discrete steps leading to the killing. Differential transcriptional single-cell profiling of killers and non-killers identified CD137 as an inducible costimulatory molecule upregulated on killer T cells. Our single-cell profiling results directly demonstrate that inducible CD137 is feature of killer (and serial killer) T cells and this marks a different subset compared with the CD107apos (degranulating) subset of CAR T cells. Ligation of the induced CD137 with CD137 ligand (CD137L) leads to younger CD19 CAR T cells with sustained killing and lower exhaustion. We genetically modified CAR T cells to co-express CD137L, in trans, and this lead to a profound improvement in anti-tumor efficacy in leukemia and refractory ovarian cancer models in mice. CONCLUSIONS: Broadly, our results illustrate that while non-killer T cells are reflective of population heterogeneity, integrated single-cell profiling can enable identification of mechanisms that can enhance the function/proliferation of killer T cells leading to direct anti-tumor benefit.


Subject(s)
4-1BB Ligand/genetics , Gene Expression Profiling , Immunotherapy, Adoptive , Leukemia/therapy , Ovarian Neoplasms/therapy , Receptors, Chimeric Antigen/genetics , Single-Cell Analysis , T-Lymphocytes/transplantation , Transcriptome , 4-1BB Ligand/metabolism , Animals , Cytotoxicity, Immunologic/genetics , Female , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Immunophenotyping , K562 Cells , Leukemia/genetics , Leukemia/immunology , Leukemia/metabolism , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Phenotype , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
11.
J Immunother Cancer ; 9(2)2021 02.
Article in English | MEDLINE | ID: mdl-33579736

ABSTRACT

Background Human cancers are extraordinarily heterogeneous in terms of tumor antigen expression, immune infiltration and composition. A common feature, however, is the host's inability to mount potent immune responses that prevent tumor growth effectively. Often, naturally primed CD8+ T cells against solid tumors lack adequate stimulation and efficient tumor tissue penetration due to an immune hostile tumor microenvironment.Methods To address these shortcomings, we cloned tumor-associated antigens (TAA) and the immune-stimulatory ligand 4-1BBL into the genome of modified vaccinia Ankara (MVA) for intratumoral virotherapy.Results Local treatment with MVA-TAA-4-1BBL resulted in control of established tumors. Intratumoral injection of MVA localized mainly to the tumor with minimal leakage to the tumor-draining lymph node. In situ infection by MVA-TAA-4-1BBL triggered profound changes in the tumor microenvironment, including the induction of multiple proinflammatory molecules and immunogenic cell death. These changes led to the reactivation and expansion of antigen-experienced, tumor-specific cytotoxic CD8+ T cells that were essential for the therapeutic antitumor effect. Strikingly, we report the induction of a systemic antitumor immune response including tumor antigen spread by local MVA-TAA-4-1BBL treatment which controlled tumor growth at distant, untreated lesions and protected against local and systemic tumor rechallenge. In all cases, 4-1BBL adjuvanted MVA was superior to MVA.Conclusion Intratumoral 4-1BBL-armed MVA immunotherapy induced a profound reactivation and expansion of potent tumor-specific CD8+ T cells as well as favorable proinflammatory changes in the tumor microenvironment, leading to elimination of tumors and protective immunological memory.


Subject(s)
4-1BB Ligand/genetics , Antigens, Neoplasm/genetics , Melanoma, Experimental/therapy , Oncolytic Virotherapy/methods , Vaccinia virus/physiology , 4-1BB Ligand/metabolism , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cloning, Molecular , Combined Modality Therapy , Drug Synergism , Female , Immunologic Memory , Melanoma, Experimental/immunology , Mice , Treatment Outcome , Tumor Microenvironment , Vaccinia virus/genetics
12.
Methods Mol Biol ; 2248: 221-229, 2021.
Article in English | MEDLINE | ID: mdl-33185879

ABSTRACT

The tumor microenvironment (TME) contains noncancerous cells such as various types of immune cells and fibroblasts. Cancer cells direct these stromal cells to create a microenvironment favorable for tumor growth and intercellular interactions have a critical role in this process. In established tumors, interactions between CD137 and its ligand (CD137L) contribute to tumor immune evasion and tumor growth. Therefore, it is important to identify cells expressing CD137 and CD137L within tumors. In this chapter, we will introduce a simple, powerful method of analyzing CD137- and CD137L-expressing tumor cells using Fluorescence-activated cell sorting.


Subject(s)
4-1BB Ligand/metabolism , Neoplasms/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , 4-1BB Ligand/genetics , Biomarkers , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression , Humans , Immunophenotyping/methods , Macrophages/immunology , Macrophages/metabolism , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
13.
J Exp Med ; 218(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-32991668

ABSTRACT

Immunotherapy has revolutionized the treatment of many tumors. However, most glioblastoma (GBM) patients have not, so far, benefited from such successes. With the goal of exploring ways to boost anti-GBM immunity, we developed a B cell-based vaccine (BVax) that consists of 4-1BBL+ B cells activated with CD40 agonism and IFNγ stimulation. BVax migrates to key secondary lymphoid organs and is proficient at antigen cross-presentation, which promotes both the survival and the functionality of CD8+ T cells. A combination of radiation, BVax, and PD-L1 blockade conferred tumor eradication in 80% of treated tumor-bearing animals. This treatment elicited immunological memory that prevented the growth of new tumors upon subsequent reinjection in cured mice. GBM patient-derived BVax was successful in activating autologous CD8+ T cells; these T cells showed a strong ability to kill autologous glioma cells. Our study provides an efficient alternative to current immunotherapeutic approaches that can be readily translated to the clinic.


Subject(s)
4-1BB Ligand/immunology , B-Lymphocytes/immunology , CD40 Antigens/immunology , Cancer Vaccines/immunology , Glioblastoma/therapy , Interferon-gamma/immunology , Neoplasms, Experimental/therapy , 4-1BB Ligand/genetics , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD40 Antigens/genetics , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/pathology , Interferon-gamma/genetics , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology
14.
Proc Natl Acad Sci U S A ; 117(50): 31780-31788, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33239441

ABSTRACT

Engineered cytokines are gaining importance in cancer therapy, but these products are often limited by toxicity, especially at early time points after intravenous administration. 4-1BB is a member of the tumor necrosis factor receptor superfamily, which has been considered as a target for therapeutic strategies with agonistic antibodies or using its cognate cytokine ligand, 4-1BBL. Here we describe the engineering of an antibody fusion protein, termed F8-4-1BBL, that does not exhibit cytokine activity in solution but regains biological activity on antigen binding. F8-4-1BBL bound specifically to its cognate antigen, the alternatively spliced EDA domain of fibronectin, and selectively localized to tumors in vivo, as evidenced by quantitative biodistribution experiments. The product promoted a potent antitumor activity in various mouse models of cancer without apparent toxicity at the doses used. F8-4-1BBL represents a prototype for antibody-cytokine fusion proteins, which conditionally display "activity on demand" properties at the site of disease on antigen binding and reduce toxicity to normal tissues.


Subject(s)
4-1BB Ligand/administration & dosage , Antigens, Neoplasm/administration & dosage , Neoplasms/drug therapy , Recombinant Fusion Proteins/administration & dosage , 4-1BB Ligand/genetics , 4-1BB Ligand/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Fibronectins/genetics , Fibronectins/immunology , Humans , Mice , Neoplasms/immunology , Protein Domains/genetics , Protein Domains/immunology , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
15.
Sci Rep ; 10(1): 15160, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32939048

ABSTRACT

Genetically modified tumor cells harboring immunomodulators may be used as therapeutic vaccines to stimulate antitumor immunity. The therapeutic benefit of these tumor vaccines is extensively investigated and mechanisms by which they boost antitumor response may be further explored. Tumor cells are large secretors of extracellular vesicles (EVs). These EVs are able to vehiculate RNA and proteins to target cells, and engineered EVs also vehiculate recombinant proteins. In this study, we explore immunomodulatory properties of EVs derived from antitumor vaccines expressing the TNFSF ligands 4-1BBL and OX40L, modulating immune response mediated by immune cells and eliminating tumors. Our results suggest that the EVs secreted by genetically modified tumor cells harboring TNFSF ligands can induce T cell proliferation, inhibit the transcription factor FoxP3, associated with the maintenance of Treg phenotype, and enhance antitumor activity mediated by immune cells. The immunomodulatory extracellular vesicles have potential to be further engineered for developing new approaches for cancer therapy.


Subject(s)
4-1BB Ligand/immunology , Cancer Vaccines/therapeutic use , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , OX40 Ligand/immunology , 4-1BB Ligand/genetics , Animals , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Extracellular Vesicles/genetics , Extracellular Vesicles/immunology , Extracellular Vesicles/ultrastructure , Forkhead Transcription Factors/antagonists & inhibitors , Immunologic Factors/genetics , Immunologic Factors/immunology , Immunologic Factors/therapeutic use , In Vitro Techniques , Lymphocyte Activation , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , OX40 Ligand/genetics
16.
Immunotherapy ; 12(10): 681-696, 2020 07.
Article in English | MEDLINE | ID: mdl-32580597

ABSTRACT

Aim: CD19 chimeric antigen receptor (CAR) T cells have been approved by the US FDA for treatment of relapsed and refractory (R/R) B-cell malignancies. Patients & methods: This study investigated the safety and efficacy of autologous 4-1BB costimulatory domain-engineered CD19 CAR-T cells in R/R B-cell lymphoma. Results: After CD19 CAR-T-cell infusion, severe cytokine release syndrome occurred in 28.6% (4/14) of the patients. The overall response rate was 77% with complete remission observed in 6/14 patients at 3 months. A higher tumor burden and grade 3-4 of myelosuppression after chemotherapy were associated with severe cytokine-release syndrome. Notably, combining CD19 CAR-T cells and PD-1 blockade, but not CD19 CAR-T cells alone, reduced intracranial tumor burden in a patient with central invasion of lymphoma. Conclusion: CD19 CAR-T cells could effectively induce tumor remission and PD-1 blockade might improve the efficacy in Chinese patients with R/R B-cell lymphoma.


Subject(s)
Cytokine Release Syndrome/epidemiology , Drug-Related Side Effects and Adverse Reactions/epidemiology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/therapy , 4-1BB Ligand/genetics , 4-1BB Ligand/metabolism , Adult , Antigens, CD19/genetics , Antigens, CD19/metabolism , Combined Modality Therapy , Cytokine Release Syndrome/etiology , Humans , Lymphoma, B-Cell/epidemiology , Lymphoma, B-Cell/immunology , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Recurrence , Remission Induction , Tumor Burden , Young Adult
17.
J Immunol ; 204(11): 2887-2899, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32295876

ABSTRACT

CD137 modulates type 1 diabetes (T1D) progression in NOD mice. We previously showed that CD137 expression in CD4 T cells inhibits T1D, but its expression in CD8 T cells promotes disease development by intrinsically enhancing the accumulation of ß-cell-autoreactive CD8 T cells. CD137 is expressed on a subset of FOXP3+ regulatory CD4 T cells (Tregs), and CD137+ Tregs are the main source of soluble CD137. Soluble CD137 suppresses T cells in vitro by binding to the CD137 ligand (CD137L) upregulated on activated T cells. To further study how the opposing functions of CD137 are regulated, we successfully targeted Tnfsf9 (encoding CD137L) in NOD mice using the CRISPR/Cas9 system (designated NOD.Tnfsf9 -/-). Relative to wild-type NOD mice, T1D development in the NOD.Tnfsf9 -/- strain was significantly delayed, and mice developed less insulitis and had reduced frequencies of ß-cell-autoreactive CD8 T cells. Bone marrow chimera experiments showed that CD137L-deficient hematopoietic cells were able to confer T1D resistance. Adoptive T cell transfer experiments showed that CD137L deficiency on myeloid APCs was associated with T1D suppression. Conversely, lack of CD137L on T cells enhanced their diabetogenic activity. Furthermore, neither CD137 nor CD137L was required for the development and homeostasis of FOXP3+ Tregs. However, CD137 was critical for the in vivo T1D-suppressive activity of FOXP3+ Tregs, suggesting that the interaction between CD137 and CD137L regulates their function. Collectively, our results provide new insights into the complex roles of CD137-CD137L interaction in T1D.


Subject(s)
4-1BB Ligand/metabolism , Diabetes Mellitus, Type 1/immunology , T-Lymphocytes, Regulatory/metabolism , 4-1BB Ligand/genetics , Animals , CD4 Antigens/metabolism , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats , Forkhead Transcription Factors/metabolism , Homeostasis , Humans , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, Knockout , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Transplantation Chimera , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
18.
Expert Rev Proteomics ; 17(2): 175-185, 2020 02.
Article in English | MEDLINE | ID: mdl-32125181

ABSTRACT

Background: The immune-inducing effect of the quorum sensing (QS) molecule autoinducer-2 (AI-2) on macrophages has not been previously comprehensively studied.Methods: We performed proteomic analysis on macrophages cocultured with purified Fusobacterium nucleatum (F. nucleatum) AI-2 and performed western blot analysis to verify the differential protein expression. We further used the Gene Expression Profiling Interactive Analysis and Tumor Immune Estimation Resource databases to analyze the expression of differentially expressed proteins in microbial-associated digestive tract tumors.Results: Based on proteomic analysis, we identified 46 upregulated proteins and 11 downregulated proteins. The upregulated proteins were mostly inflammatory factors such as tumor necrosis factor ligand superfamily member 9 (TNFSF9). These proteins have a range of biological functions associated with the regulation of inflammatory responses, apoptosis and tumorigenesis. TNFSF9 is highly expressed in pancreatic adenocarcinoma (PAAD) tissues and is associated with M1 polarization of macrophages.Conclusions: Our data indicated that F. nucleatum AI-2 induced inflammatory responses and activated multiple signaling pathways in macrophages. TNFSF9 is the most significantly differentially expressed protein induced by F. nucleatum AI-2 and is involved in regulating immune cell infiltration in PAAD. Thus, AI-2 may become a new focus for studying the relationship between bacteria and cancer.


Subject(s)
Homoserine/analogs & derivatives , Lactones/pharmacology , Macrophage Activation , Macrophages/metabolism , Proteome/metabolism , 4-1BB Ligand/genetics , 4-1BB Ligand/metabolism , Cell Line, Tumor , Fusobacterium nucleatum/chemistry , Homoserine/pharmacology , Humans , Macrophages/drug effects , Proteome/genetics , Proteomics/methods , Tandem Mass Spectrometry/methods , Up-Regulation
19.
Cell Cycle ; 19(5): 577-591, 2020 03.
Article in English | MEDLINE | ID: mdl-31992123

ABSTRACT

Anti-tumor immune response and the prognosis of tumor are the results of competition between stimulatory and inhibitory checkpoints. Except for upregulating inhibitory checkpoints, lowering some immune accelerating molecules to convert an immunostimulatory microenvironment into an immunodormant one through "decelerating the accelerator" might be another effective immune escape pattern. 4-1BBL is a classical transmembrane costimulatory molecule involving in antitumor immune responses. In contrast, we demonstrated that 4-1BBL is predominantly localized in the nuclei of cancer cells in colon cancer specimens and is positively correlated with tumor size, lymph node metastasis, and a lower survival ratio. Furthermore, the nuclear localization of 4-1BBL was also ascertained in vitro. 4-1BBL knockout (KO) arrests the proliferation and impaired the migration and invasion ability of colon cancer cells in vitro and retarded tumor growth in vivo. 4-1BBL KO increased the accumulation of Gsk3ß in the nuclei of colon cancer cells and consequently decreased the expression of Wnt pathway target genes and thus alter tumor biological behavior. We hypothesized that unlike membrane-expressed 4-1BBL, which stimulates the 4-1BB signaling of antitumor cytotoxic T cells, the nuclear-localized 4-1BBL could facilitate the malignant behavior of colon cancer cells by circumventing antitumor signaling and driving some key oncotropic signal pathway in the nucleus. Nuclear-localized 4-1BBL might be an indicator of colon cancer malignancy and serve as a promising target of immunotherapy.


Subject(s)
4-1BB Ligand/metabolism , Cell Movement , Cell Nucleus/metabolism , Colonic Neoplasms/diagnosis , Colonic Neoplasms/pathology , Glycogen Synthase Kinase 3 beta/metabolism , 4-1BB Ligand/biosynthesis , 4-1BB Ligand/deficiency , 4-1BB Ligand/genetics , Animals , Base Sequence , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Proliferation , Cohort Studies , Colonic Neoplasms/enzymology , Colonic Neoplasms/metabolism , Disease Models, Animal , Gene Knockout Techniques , HCT116 Cells , Humans , Mice , Neoplasm Transplantation , Prognosis , Protein Transport , Survival Analysis
20.
Front Immunol ; 10: 2216, 2019.
Article in English | MEDLINE | ID: mdl-31632390

ABSTRACT

Dendritic cell (DC)-based immunotherapies are being explored for over 20 years and found to be very safe. Most often, granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4)-induced monocyte-derived DCs (moDCs) are being used, which have demonstrated some life-prolonging benefit to patients of multiple tumors. However, the limited clinical response and efficacy call for the development of more potent DCs. CD137L-DC may meet this demand. CD137L-DCs are a novel type of monocyte-derived inflammatory DCs that are induced by CD137 ligand (CD137L) agonists. CD137L is expressed on the surface of antigen-presenting cells, including monocytes, and signaling of CD137L into monocytes induces their differentiation to CD137L-DCs. CD137L-DCs preferentially induce type 1 T helper (Th1) cell polarization and strong type 1 CD8+ T cell (Tc1) responses against tumor-associated viral antigens. The in vitro T cell-stimulatory capacity of CD137L-DCs is superior to that of conventional moDCs. The transcriptomic profile of CD137L-DC is highly similar to that of in vivo DCs at sites of inflammation. The strict activation dependence of CD137 expression and its restricted expression on activated T cells, NK cells, and vascular endothelial cells at inflammatory sites make CD137 an ideally suited signal for the induction of monocyte-derived inflammatory DCs in vivo. These findings and their potency encouraged a phase I clinical trial of CD137L-DCs against Epstein-Barr virus-associated nasopharyngeal carcinoma. In this review, we introduce and summarize the history, the characteristics, and the transcriptional profile of CD137L-DC, and discuss the potential development and applications of CD137L-DC.


Subject(s)
4-1BB Ligand/immunology , Cell Differentiation/immunology , Dendritic Cells , Epstein-Barr Virus Infections , Herpesvirus 4, Human/immunology , Immunization , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , 4-1BB Ligand/genetics , Clinical Trials, Phase I as Topic , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/transplantation , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/therapy , Humans , Monocytes/immunology , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/therapy , T-Lymphocytes/immunology , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...