ArticlePublisher preview available

A vaccine targeting resistant tumours by dual T cell plus NK cell attack

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Most cancer vaccines target peptide antigens, necessitating personalization owing to the vast inter-individual diversity in major histocompatibility complex (MHC) molecules that present peptides to T cells. Furthermore, tumours frequently escape T cell-mediated immunity through mechanisms that interfere with peptide presentation1. Here we report a cancer vaccine that induces a coordinated attack by diverse T cell and natural killer (NK) cell populations. The vaccine targets the MICA and MICB (MICA/B) stress proteins expressed by many human cancers as a result of DNA damage2. MICA/B serve as ligands for the activating NKG2D receptor on T cells and NK cells, but tumours evade immune recognition by proteolytic MICA/B cleavage3,4. Vaccine-induced antibodies increase the density of MICA/B proteins on the surface of tumour cells by inhibiting proteolytic shedding, enhance presentation of tumour antigens by dendritic cells to T cells and augment the cytotoxic function of NK cells. Notably, this vaccine maintains efficacy against MHC class I-deficient tumours resistant to cytotoxic T cells through the coordinated action of NK cells and CD4+ T cells. The vaccine is also efficacious in a clinically important setting: immunization following surgical removal of primary, highly metastatic tumours inhibits the later outgrowth of metastases. This vaccine design enables protective immunity even against tumours with common escape mutations. A vaccine targeting stress proteins expressed by many cancers blocks a tumour escape mechanism, enabling protective immunity mediated by diverse T cell and NK cell populations.
Efficacy of the MICA/B α3 domain cancer vaccine a, Design of the MICA/B vaccine. pAbs, polyclonal serum IgG. b, MICB-specific serum antibody (Ab) titres quantified by flow cytometry (n = 4 mice per group) in MICB-transgenic mice immunized with Ctrl-vax (blue) or MICB-vax (red). MFI, median fluorescence intensity. c, MICB-specific CD4⁺ T cell responses following immunization with MICB-vax or Ctrl-vax; CFSE dilution of splenocytes stimulated with MICB or control protein (ovalbumin (OVA)); shown are representative flow cytometry plots (left) and quantification for three mice per group (right). d, Cell-surface levels of MICB on B16F10 (MICB) tumours from mice immunized with MICB-vax or Ctrl-vax (n = 4 mice per group); staining of tumour cells with isotype-control monoclonal antibody (grey) or anti-MICA/B monoclonal antibody (specific for the α1–α2 domains, monoclonal antibody not blocked by vaccine-induced antibodies). e, Therapeutic efficacy of MICB-vax (red) or Ctrl-vax (blue) in mice with established B16F10 (MICB) tumours immunized at the indicated time points (n = 7 mice per group). Vax, vaccination. *P = 0.0137, ****P < 0.0001; NS (not significant), P > 0.999. f, g, Vaccine efficacy in two models of spontaneous metastasis. Mice were immunized with Ctrl-vax (blue) or MICB-vax (red) following surgical removal of primary tumours using the B16-B6 melanoma (f; 10 mice per group) or 4T1 breast cancer (g; 13 mice per group) models. Shown are the size of primary tumours at the time of surgery (left), representative images of lung metastases (middle) and quantification of the total number of lung surface metastases (right). D, day; s.c., subcutaneous. h, i, Immunogenicity of the rhesus MICA/B α3 domain vaccine in the rhesus macaque model. h, Timeline of vaccination; blood was drawn 24 h before indicated immunization or boost. i, Serum titres of antibody to rhesus MICA/B for animal ID 9312. Representative data are shown from at least three (b) or two (c–g) independent experiments. Data from a single experiment with technical replicates for each time point are shown in i. Statistical significance was assessed by two-tailed unpaired Student’s t test (b), two-way ANOVA with Sidak’s multiple-comparison test (c), one-way ANOVA with Tukey’s multiple-comparison test (d), two-way ANOVA with Bonferroni’s post hoc test (left) and the log-rank (Mantel–Cox) test (right) (e), two-tailed Mann–Whitney test (f, g) and two-way ANOVA with Tukey’s multiple-comparison test (i). Data are depicted as the mean ± s.e.m. (b–g) or mean ± s.d. (i). Source data
… 
Vaccine induces T cell and NK cell recruitment into tumours a, Strategy for characterization of tumour-infiltrating immune cells by flow cytometry (FACS) and scRNA-seq. Mice received two immunizations (days 0 and 14), B16F10 (MICB-dox) tumour cells were implanted (day 21), MICB expression was induced on tumour cells by doxycycline treatment (day 28) and tumour-infiltrating immune cells were analysed 7 d later. b–d, Tumour-infiltrating T cell populations following immunization with Ctrl-vax (blue) or MICB-vax (red) (Ctrl-vax, n = 9 mice per group; MICB-vax, n = 10 mice per group; n = 7 mice per group for the γδ T cell panel). Treg cells, regulatory T cells. e, Tumour-infiltrating NK cells (n = 8 mice per group). f, g, Quantification of IFNγ-positive CD4⁺ and CD8⁺ T cells (Ctrl-vax, n = 9 mice per group; MICB-vax, n = 10 mice per group). h, i, UMAP representation of all T cell clusters from scRNA-seq data (h) and the fraction of each T cell subpopulation among total CD45⁺ cells from the experimental group (MICB-vax + doxycycline; red) and three combined control groups (Ctrl-vax ± doxycycline and MICB-vax without doxycycline; blue) (i). DN, double negative. j, k, UMAP representation of NK and ILC1 cells from all experimental groups (j) and the fraction of these subpopulations among total CD45⁺ cells from experimental (red) and combined control (blue) groups (k). l, Fraction of T cells representing expanded clones based on TCR sequence analysis for the experimental group (red) and combined control groups (blue). m, Contribution of CD4⁺ T cells, NK cells and NKG2D receptor to vaccine efficacy. Mice were first immunized with MICB-vax (M) or Ctrl-vax (C) (days 0 and 14) and treated with isotype-control monoclonal antibody (iso), depleting monoclonal antibody (targeting CD4⁺ T cells or NK cells (αCD4 and αNK1.1, respectively)) or NKG2D receptor-blocking monoclonal antibody (αNKG2D) starting on day 21, followed by implantation of B16F10 (MICB) tumour cells (n = 7 mice per group). Representative data from three independent experiments are shown in b–g. scRNA-seq data from a single experiment with sorted CD45⁺ cells pooled from five mice per group are shown in h–l. Representative data from two independent experiments are shown in m. Statistical significance was assessed by two-tailed Mann–Whitney test (b–g) or log-rank (Mantel–Cox) test (m). Data are depicted as the mean ± s.e.m. Source data
… 
Vaccine retains efficacy against MHC-I-deficient tumours a, b, Comparison of vaccine efficacy against B16F10 (MICB) WT tumours and tumours with resistance mutations in the B2m (a) or Ifngr1 (b) gene. Mice received MICB-vax or Ctrl-vax and were then challenged with tumours of the indicated genotype (n = 7 mice per group). KO, knockout. c, Effect of CD4⁺ T cell and NK cell depletion on immunity to B2m-knockout tumours. Mice were immunized with MICB-vax or Ctrl-vax; treatment with depleting or isotype-control monoclonal antibody was started 2 d before injection of B2m-knockout B16F10 (MICB) tumour cells (n = 7 mice per group). d, Contribution of vaccine-induced anti-MICB antibodies to NK cell-mediated cytotoxicity against B2m-knockout B16F10 (MICB) tumour cells. CFSE-labelled B2m-knockout B16F10 (MICB) tumour cells were pre-incubated with 10 µg per well of purified serum IgG from mice immunized with MICB-vax or Ctrl-vax before the addition of NK cells at different effector to target (E:T) ratios as indicated. The percentage of dead target cells was assessed by flow cytometry. e, Effect of CD4⁺ T cell depletion on vaccine-induced NK cell infiltration into tumours. Flow cytometry quantification of total NK cell numbers is shown in WT (left) and B2m-knockout (right) tumours for the following treatment groups: Ctrl-vax + isotype-control monoclonal antibody (blue), Ctrl-vax + anti-CD4 (orange), MICB-vax + isotype-control monoclonal antibody (red) and MICB-vax + anti-CD4 (green) (n = 7 mice per group). Representative data from two independent experiments are shown in a–e. Statistical significance was assessed by two-way ANOVA with Bonferroni’s post hoc test (left) and log-rank (Mantel–Cox) test (right) (a–c), two-way ANOVA with Sidak’s multiple-comparison test (d) and one-way ANOVA with Tukey’s multiple-comparison test (e). Data are depicted as the mean ± s.e.m. Source data
… 
Role of CD4⁺ T cells and cDC1 cells in NK cell recruitment to tumours a, Effect of CD4⁺ T cells on migratory DC populations in the tdLNs of mice immunized with MICB-vax versus Ctrl-vax. Total migratory DCs as well as cDC1 and cDC2 cells were quantified 2 d after induction of MICB expression in tumour cells by doxycycline treatment (n = 7 mice per group, except n = 6 for Ctrl-vax without anti-CD4). b, Migratory DC subsets within the tdLN of MICB-vax-immunized mice treated following immunization (days 28 + 30) with isotype-control, CD4-depleting or CD40L-blocking monoclonal antibody (n = 7 mice per group). c, Quantification of DC populations within the tumours of mice immunized with Ctrl-vax (blue) or MICB-vax (red) on day 7 following induction of MICB expression with doxycycline (n = 7 mice per group). d, Effect of cDC1 depletion on MICB vaccine-induced T cell and NK cell accumulation within tumours in Xcr1DTR mice. Mice were treated with DT or left untreated starting on day 26 following immunization with Ctrl-vax or MICB-vax (days 0 + 14) and B16F10 (MICB-dox) tumour implantation (day 21). Immune cells were analysed in tumours 7 d after induction of MICB expression on tumours with doxycycline (day 37) (n = 7 mice per group). e, Contribution of vaccine-induced anti-MICB antibodies to DC-mediated cross-presentation of tumour antigens to CD8⁺ T cells. Bone marrow-derived DCs (BMDCs) were pre-incubated with B2m-knockout B16F10 (MICB-OVA) tumour cells in the presence of affinity-purified serum IgG from mice immunized with Ctrl-vax or MICB-vax at the indicated concentrations. DCs were co-cultured with CFSE-labelled OT-1 CD8⁺ T cells with T cell proliferation as the readout. The role of activating Fc receptor (FcR) was assessed using BMDCs from Fcer1g–/– mice (orange) or pre-incubation of BMDCs with FcR-blocking antibody (yellow) before tumour cell addition. Representative data from two independent experiments are shown in a–e. Statistical significance was assessed by one-way ANOVA with Tukey’s multiple-comparison test (a, b, d), two-tailed Mann–Whitney test (c) and two-way ANOVA with Tukey’s multiple-comparison test (e). Data are depicted as the mean ± s.e.m. (a–d) or mean ± s.d. (e). Source data
… 
This content is subject to copyright. Terms and conditions apply.
992 | Nature | Vol 606 | 30 June 2022
Article
A vaccine targeting resistant tumours by
dual Tcell plus NK cell attack
Soumya Badrinath1,2, Maxence O. Dellacherie3,4,13, Aileen Li3 ,4,1 0,13 , Shiwei Zheng5,11 ,13,
Xixi Zhang1,2, Miguel Sobral3,4, Jason W. Pyrdol1, Kathryn L. Smith1, Yuheng Lu5,
Sabrina Haag1,2, Hamza Ijaz4, Fawn Connor-Stroud6, Tsuneyasu Kaisho7, Glenn Dranoff8,1 2,
Guo-Cheng Yuan5,11, David J. Mooney3,4 & Kai W. Wucherpfennig1,2,9 ✉
Most cancer vaccines target peptide antigens, necessitating personalization owing
to the vast inter-individual diversity in major histocompatibility complex (MHC)
molecules that present peptides to Tcells. Furthermore, tumours frequently
escape Tcell-mediated immunity through mechanisms that interfere with peptide
presentation1. Here we report a cancer vaccine that induces a coordinated attack
by diverse Tcell and natural killer (NK) cell populations. The vaccine targets the
MICA and MICB (MICA/B) stress proteins expressed by many human cancers as a
result of DNA damage2. MICA/B serve as ligands for the activating NKG2D receptor
on Tcells and NK cells, but tumours evade immune recognition by proteolytic
MICA/B cleavage3,4. Vaccine-induced antibodies increase the density of MICA/B
proteins on the surface of tumour cells by inhibiting proteolytic shedding, enhance
presentation of tumour antigens by dendritic cells to Tcells and augment the
cytotoxic function of NK cells. Notably, this vaccine maintains ecacy against
MHC class I-decient tumours resistant to cytotoxic Tcells through the
coordinated action of NK cells and CD4+ Tcells. The vaccine is also ecacious in a
clinically important setting: immunization following surgical removal of primary,
highly metastatic tumours inhibits the later outgrowth of metastases. This vaccine
design enables protective immunity even against tumours with common escape
mutations.
We developed a conceptually new cancer vaccine that targets a
tumour immune escape mechanism. The vaccine targets the MICA
and MICB (MICA/B) stress proteins that are upregulated in response
to DNA damage in many types of human cancers but are expressed
at low or undetectable levels by healthy cells
2,3,5,6
. Engagement of
the activating NKG2D receptor by membrane-bound MICA/B trig-
gers the cytotoxicity programme in natural killer (NK) cells and
co-stimulatory signalling in CD8+ Tcells7–9. However, many human
tumours evade this important immune recognition pathway by
proteolytic shedding of MICA/B from the cell surface
4,10–13
. Shed-
ding substantially reduces the surface density on tumour cells of
these immunostimulatory ligands for the NKG2D receptor12,14. Shed
MICA/B proteins have also been reported to induce NKG2D recep-
tor internalization and inhibit NK cell function12,1416. Patients with
melanoma responding to an autologous cell-based cancer vaccine
(GVAX) plus anti-CTLA-4 were found to develop anti-MICA antibod-
ies, and the presence of these antibodies correlated with reduced
serum levels of shed MICA and augmented CD8
+
Tcell and NK cell
responses17,18.
Design of the MICA and MICB α3 domain vaccine
Our vaccine targeted the highly conserved α3 domain in MICA/B, the site
of proteolytic shedding, and was designed to induce tumour immunity
by Tcells and NK cells (Fig.1a)19. We intentionally omitted the α1–α2
domains to avoid induction of antibodies that could block NKG2D
receptor binding
20
. Multivalent display of vaccine antigens greatly
enhances immunogenicity
21
, and we therefore fused the α3 domain
of MICB or MICA to the N terminus of ferritin from Heliobacter pylori,
which forms particles composed of 24 subunits22 (Extended Data
Fig.1a–c). Ferritin was used as a control antigen in all experiments.
A recently developed mesoporous silica rod (MSR) biodegradable
scaffold formulated with granulocyte-macrophage colony-stimulating
factor (GM-CSF; for dendritic cell (DC) recruitment) and CpG ODN 1826
(adjuvant) was used for vaccine delivery23.
https://doi.org/10.1038/s41586-022-04772-4
Received: 30 July 2021
Accepted: 19 April 2022
Published online: 25 May 2022
Check for updates
1Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA. 2Department of Immunology, Harvard Medical School, Boston, MA, USA. 3John A. Paulson
School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA. 4Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA. 5Department
of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA. 6Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA. 7Department of Immunology, Institute of
Advanced Medicine, Wakayama Medical University, Wakayama, Japan. 8Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. 9Department of Neurology, Brigham
and Women’s Hospital, Boston, MA, USA. 10Present address: Lyell Immunopharma, South San Francisco, CA, USA. 11Present address: Department of Genetics and Genomic Sciences, Charles
Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA. 12Present address: Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
13These authors contributed equally: Maxence O. Dellacherie, Aileen Li, Shiwei Zheng. e-mail: Kai_Wucherpfennig@dfci.harvard.edu
Content courtesy of Springer Nature, terms of use apply. Rights reserved
... Due to the lack of innate immunity, it is difficult to be recognized and killed by immune cells, and the immune checkpoint inhibitor ICIs are also difficult to play a role [1]. The cause of its immune deficiency may be (1) lack of tumor-related antigens (2) deficiency of antigen-presenting cells APCs (3) deletion of T cell activation (4) damage to the transport of T cells to the tumor [2]. Therefore, how to convert cold tumors into hot tumors is also a research hotspot in tumor treatment. ...
... According to the classification of target sites, tumor vaccines can be divided into tumor-related antigen (TAAs) vaccines and new antigen (TSAs) vaccines. Both of these show significant effects in the activation of T cells and NK cells [4]. Among them, TAAs vaccines have strong immunogenicity, but they need to face autoimmune problems, which may produce some side effects. ...
... Among the above methods, tumor vaccine is a new therapy that is considered to have strong anticancer activity (Figure 2). It mainly realizes the conversion of cold tumors to hot tumors by activating the immune response of T cells and NK cells [4]. Tumor vaccine can expand the number of specific T cells and increase the transportation of T cells to the tumor area [19]. ...
Article
Full-text available
Compared with hot tumors, cold tumors are a more difficult problem at present. Because cold tumors lack in congenital immunity, it is difficult for ordinary immune examination inhibitors to work. Therefore, how to "ignite" cold tumors as hot tumors is the focus of the current research. At present, scholars have proposed different and corresponding methods for various causes of T cell immunodeficiency. Among them, tumor vaccines stand out from the principle of activating the immune response of T cells and NK cells, and is currently generally recognized as a new therapy and research hotspot with strong anti-cancer activity. By classifying the target sites of tumor vaccines, they can be divided into tumor-related antigen (TAAs) vaccines and new antigen (TSAs) vaccines. Both vaccines have shown significant effects in activating T cells and NK cells. In terms of the current development status of the two vaccines, more and more research teams have invested in the research of TSAs vaccines, and many teams still insist on the research of TAAs vaccines. Although these two vaccines have their own difficulties to overcome, they still have made great contributions to the development of tumor treatment research, and both have advantages and broad prospects that cannot be reached by other methods. It is believed that no more effective tumor vaccines have been put into clinical trials.
... A significant advancement in this field emerged with the development of a cancer vaccine targeting MICA and MICB (MICA/B). This innovative approach induces a coordinated attack by both T cell and NK cell populations, maintaining effectiveness against MHC class I-deficient tumors through the combined action of NK cells and CD4+ T cells (160). ...
Article
Full-text available
Neoantigen-based cancer vaccine therapy represents a promising precision oncology strategy that targets unique tumor-specific mutations to elicit a robust immune response. This therapeutic approach is designed to harness the host’s immune response against tumor-specific neoantigens to eliminate cancer cells. The efficacy of neoantigen vaccines dependents on the coordinated action of diverse immune cells, including T lymphocytes, dendritic cells, B lymphocytes, natural killer cells, and macrophages. Each cell type plays a distinct and crucial role in recognizing, targeting, and destroying malignant cells. Understanding the mechanisms governing both individual and collective immune cell dynamics is crucial for therapeutic success. This comprehensive review systematically explores the mechanisms of neoantigen-specific immune cells, their dynamic interactions, and clinical application progress, aiming to unveil their potential value and future development in cancer treatment.
... This innovative vaccine demonstrated potential for developing therapies that combine multiple immune effects to combat cancer. [71] Moreover, FTn is organized in octahedral symmetry with eight threefold axes on its surface, making it well-suited for presenting viral proteins. Leveraging this property, Nabel's group employed the structural knowledge of FTn to design selfassembled nanoparticles capable of eliciting more potent immune responses than traditional flu vaccines. ...
Article
Full-text available
Nanomedicines have demonstrated significant potential in disease diagnosis and therapy, revolutionizing traditional drug development patterns. Recently, inspired by both natural and engineering principles, synthetic biology integrates the complexity of biological systems with the precision of engineering to design and create novel biological components, devices, and systems. This convergence of synthetic biology and nanomedicine has led to the emergence of a new concept: synthetic biological nanomedicine. Unlike traditional or biomimetic nanomedicines, synthetic biological nanomedicines are designed using gene engineering‐based strategies. In this Perspective, the foundational concepts of synthetic biological nanomedicine are introduced and its relationship to, and differences from, traditional and biomimetic nanomedicine are explored. Drawing from synthetic biology, synthetic biological nanomedicine also incorporates two main approaches: top‐down and bottom‐up strategies. The latest advancements in the application of synthetic biology to nanomedicine are reviewed, these developments are categorized according to the aforementioned strategies, and a discussion of the potential advantages and challenges associated with utilizing synthetic biology in nanomedicine development is concluded.
... Among patients in high-risk group, reduced immune infiltration suggests potential benefits from combination immunotherapies, such as PD-1/PD-L1 inhibitors, targeted suppression of inhibitory cytokines, cancer vaccines, or autologous immune cell infusions, may enhance antitumor efficacy (50). Recent advancements have also led to the development of cancer vaccines capable of inducing dual antitumor responses from T cells and NK cells (51). Therefore, the CBRD risk score system may provide potential predictive value for combination immunotherapy strategies, particularly for high-risk breast cancer patients with diminished immune infiltration. ...
Article
Full-text available
Introduction Breast cancer is among the most prevalent malignant tumors globally, with carboplatin serving as a standard treatment option. However, resistance often compromises its efficacy. DNA damage repair (DDR) pathways are crucial in determining responses to treatment and are also associated with immune infiltration. This study aimed to identify the DDR genes involved in carboplatin resistance and to elucidate their effects on prognosis, immune infiltration, and drug sensitivity in breast cancer patients. Methods A 3D-culture model resistant to carboplatin was constructed and sequenced. Co-expressed DDR genes were analyzed to develop a predictive model. Immune infiltration analysis tools were employed to assess the immune microenvironment of patients with varying expression levels of these risk genes. Additionally, drug sensitivity predictions were made to evaluate the efficacy of other DNA damage-related drugs across different risk groups. Molecular assays were performed to investigate the role of the key gene TONSL in breast cancer. Results By integrating data from public database, we established a prognostic signature comprising thirteen DDR genes. Our analysis indicated that this model is associated with immune infiltration patterns in breast cancer patients, particularly concerning CD8+ T cells and NK cells. Additionally, it demonstrated a significant correlation with sensitivity to other DDR-related drugs, suggesting its potential as a biomarker for treatment efficacy. Compared to the control group, TONSL-knockdown cell lines exhibited a diminished response to DNA-damaging agents, marked by a notable increase in DNA damage levels and enhanced drug sensitivity. Furthermore, single-cell analysis revealed elevated TONSL expression in dendritic and epithelial cells, particularly in triple-negative breast cancers. Conclusions Carboplatin resistance-related DDR genes are associated with prognosis, immune infiltration, and drug sensitivity in breast cancer patients. TONSL may serve as a potential therapeutic target for breast cancer, particularly in triple-negative breast cancer, indicating new treatment strategies for these patients.
... These antibodies attach to MICA/B at the point of proteolytic cleavage, hence preventing shedding. In preclinical research, immunisation produced strong anti-tumour responses that were reliant on both NK and T cells [62,63] . Since CLN-619 is the only MICA/B-targeted mAb available in the clinic, Cullinan Oncology is taking a promising approach to targeting suppression of MICA/B shedding. ...
Article
Full-text available
Background: Immunological oncology has transformed cancer therapy, thereby increasing patients' chances of survival and quality of life. Natural killer (NK) cells, specifically, have come to prominence as powerful engines of the inherent immune system response, making them a prospective immunotherapy tool. One of the most crucial NK cell receptors namely NK-group 2, member D (NKG2D) has a pivotal role in both innate and adaptive immunity for establishing the degree of activation of NK cells. It serves as a pertinent activating receptor in the immunological identification and extermination of aberrant cells by natural killer cells and T lymphocytes, recognising a wide variety of ligands to offer comprehensive target specificity. Objective: This article aimed to emphasise a better understanding of the NKG2D receptor's structure, its signalling mechanism, and its potential and prospective implementation in a variety of medical contexts. Method: A search for published material was carried out using some combinations of the terms "cancer", "immunotherapy", "natural killer" and “NKG2D” on PubMed, ScienceDirect, Scopus, and Google Scholar. All citations from the selected papers were examined. Conclusion: In light of the numerous studies conducted, it is deduced that NKG2D-mediated cancer chemotherapy offers an excellent prospect for usage as a type of chemotherapy soon; nevertheless, additional clinical trials are required before it can be employed in clinical settings. It is crucial to identify and comprehend the functions of several transcription factors that control the expression of NKG2D on the cell membrane by binding to its ligands. Furthermore, approaches centred on augmenting NKG2D expression in immune cells and elevating NKG2DL expression in cancer cells may efficiently trigger the antitumor immune response. Keywords: cancer, immunotherapy, natural killer cell, NK group-2 D receptor
Article
Full-text available
Ultrasound therapy has turned up as a noninvasive multifunctional tool for cancer immunotherapy. However, the insufficient co‐stimulating molecules and loss of peptide‐major histocompatibility complex I (MHC‐I) expression on tumor cells lead to poor therapy of sonoimmunotherapies. Herein, this work develops a sonosensitive system to augment MHC‐I unrestricted natural killer (NK) cell‐mediated innate immunity and T cell‐mediated adaptive immunity by leveraging antigen presentation cell (APC)‐like tumor cells. Genetically engineered tumor cells featuring sufficient co‐stimulating molecules are cryo‐shocked and conjugated with a sonosensitizer, hematoporphyrin monomethyl ether, using click chemistry. These cells (DPNLs) exhibit characteristics of tumor and draining lymph node homing. Under ultrasound, NK cell‐mediated innate immunity within the tumor microenvironment could be activated, and T cells in the tumor‐draining lymph nodes (TDLNs) are stimulated through co‐stimulatory molecules. In combination with programmed cell death ligand 1 (PD‐L1) antibody, DPNLs extend the survival time and inhibited lung metastasis in triple‐negative breast cancer (TNBC) models. This study provides an alternative approach for sonoimmunotherapy with precise sonosensitizer delivery and enhanced NK cell and T cell activation.
Article
Full-text available
Although drug delivery technology has promoted the clinical translation of small molecule drugs, there is an urgent need for advanced delivery systems to overcome complex physiological barriers and the increasing development of biological drugs. This review overviews the emerging applications of synthetic biology‐based engineered cells for drug delivery. We first introduce synthetic biology strategies to engineer cells for biological drug delivery and discuss the benefits in terms of specificity, intelligence, and controllability. Furthermore, we highlight the cutting‐edge advancements at the convergence of synthetic biology and nanotechnology in drug delivery. Nanotechnology expands the engineering design and construction concepts of synthetic biology, and synthetic biology drives the development for biotechnology‐driven nanomaterial synthesis. In the future, synthetic biology‐based engineered cells may be developed to be more modular, standardized, and intelligent, leading to significant breakthroughs in the construction of advanced drug delivery systems.
Article
One driver of the high failure rates of clinical trials for therapeutic cancer vaccines is likely the inability to sufficiently engage conventional dendritic cells (cDCs), the antigen-presenting cell (APC) subset that is specialized in priming antitumor T cells. Here, we demonstrate that, relative to vaccination with an injectable mesoporous silica rod (MPS) vaccine alone (Vax), combining MPS vaccines with CD122-biased IL-2/anti-IL-2 antibody complexes (IL-2cx) drives ~3-fold expansion of cDCs at the vaccination sites, vaccine-draining lymph nodes, and spleens of treated mice. Furthermore, relative to Vax alone, Vax+IL-2cx led to a ~3-fold increase in the numbers of CD8 ⁺ T cells and ~15-fold increase in the numbers of NK cells at the vaccination site. Notably, with both the model protein antigen OVA as well as various peptide neoantigens, Vax+IL-2cx induced ~5 to 30-fold greater numbers of circulating antigen-specific CD8 ⁺ T cells relative to Vax alone. We further demonstrate that Vax+IL-2cx leads to significantly improved efficacy in the MC38 colon carcinoma model relative to either monotherapy alone, driving complete regressions in 50% of mice in a cDC-dependent manner. Relative to vaccine alone, Vax+IL-2cx led to comparable numbers of CD8 ⁺ T cells, but markedly greater numbers of NK cells and activated cDCs in the B16F10 melanoma tumor microenvironment post-therapy. Taken together, these findings suggest that the administration of factors that engage both the cDC-CD8 ⁺ T cell and cDC-NK cell axes can boost the potency of therapeutic cancer vaccines.
Article
Full-text available
Antiviral CD8⁺ T cell responses are characterized by an initial activation/priming of T lymphocytes followed by a massive proliferation, subset differentiation, population contraction and the development of a stable memory pool. The transcription factor BATF3 has been shown to play a central role in the development of conventional dendritic cells, which in turn are critical for optimal priming of CD8⁺ T cells. Here we show that BATF3 was expressed transiently within the first days after T cell priming and had long-lasting T cell–intrinsic effects. T cells that lacked Batf3 showed normal expansion and differentiation, yet succumbed to an aggravated contraction and had a diminished memory response. Vice versa, BATF3 overexpression in CD8⁺ T cells promoted their survival and transition to memory. Mechanistically, BATF3 regulated T cell apoptosis and longevity via the proapoptotic factor BIM. By programing CD8⁺ T cell survival and memory, BATF3 is a promising molecule to optimize adoptive T cell therapy in patients.
Article
Full-text available
Conventional type 1 dendritic cells (cDC1)¹ are thought to perform antigen cross-presentation, which is required to prime CD8⁺ T cells2,3, whereas cDC2 are specialized for priming CD4⁺ T cells4,5. CD4⁺ T cells are also considered to help CD8⁺ T cell responses through a variety of mechanisms6–11, including a process whereby CD4⁺ T cells ‘license’ cDC1 for CD8⁺ T cell priming¹². However, this model has not been directly tested in vivo or in the setting of help-dependent tumour rejection. Here we generated an Xcr1Cre mouse strain to evaluate the cellular interactions that mediate tumour rejection in a model requiring CD4⁺ and CD8⁺ T cells. As expected, tumour rejection required cDC1 and CD8⁺ T cell priming required the expression of major histocompatibility class I molecules by cDC1. Unexpectedly, early priming of CD4⁺ T cells against tumour-derived antigens also required cDC1, and this was not simply because they transport antigens to lymph nodes for processing by cDC2, as selective deletion of major histocompatibility class II molecules in cDC1 also prevented early CD4⁺ T cell priming. Furthermore, deletion of either major histocompatibility class II or CD40 in cDC1 impaired tumour rejection, consistent with a role for cognate CD4⁺ T cell interactions and CD40 signalling in cDC1 licensing. Finally, CD40 signalling in cDC1 was critical not only for CD8⁺ T cell priming, but also for initial CD4⁺ T cell activation. Thus, in the setting of tumour-derived antigens, cDC1 function as an autonomous platform capable of antigen processing and priming for both CD4⁺ and CD8⁺ T cells and of the direct orchestration of their cross-talk that is required for optimal anti-tumour immunity.
Article
Full-text available
Differentiation of proinflammatory CD4+ conventional T cells (Tconv) is critical for productive antitumor responses yet their elicitation remains poorly understood. We comprehensively characterized myeloid cells in tumor draining lymph nodes (tdLN) of mice and identified two subsets of conventional type-2 dendritic cells (cDC2) that traffic from tumor to tdLN and present tumor-derived antigens to CD4+ Tconv, but then fail to support antitumor CD4+ Tconv differentiation. Regulatory T cell (Treg) depletion enhanced their capacity to elicit strong CD4+ Tconv responses and ensuing antitumor protection. Analogous cDC2 populations were identified in patients, and as in mice, their abundance relative to Treg predicts protective ICOS+ PD-1lo CD4+ Tconv phenotypes and survival. Further, in melanoma patients with low Treg abundance, intratumoral cDC2 density alone correlates with abundant CD4+ Tconv and with responsiveness to anti-PD-1 therapy. Together, this highlights a pathway that restrains cDC2 and whose reversal enhances CD4+ Tconv abundance and controls tumor growth.
Article
Full-text available
Conventional type 1 dendritic cells (cDC1) are critical for antitumor immunity, and their abundance within tumors is associated with immune-mediated rejection and the success of immunotherapy. Here, we show that cDC1 accumulation in mouse tumors often depends on natural killer (NK) cells that produce the cDC1 chemoattractants CCL5 and XCL1. Similarly, in human cancers, intratumoral CCL5, XCL1, and XCL2 transcripts closely correlate with gene signatures of both NK cells and cDC1 and are associated with increased overall patient survival. Notably, tumor production of prostaglandin E2 (PGE2) leads to evasion of the NK cell-cDC1 axis in part by impairing NK cell viability and chemokine production, as well as by causing downregulation of chemokine receptor expression in cDC1. Our findings reveal a cellular and molecular checkpoint for intratumoral cDC1 recruitment that is targeted by tumor-derived PGE2 for immune evasion and that could be exploited for cancer therapy.
Article
Full-text available
Antibody blockade of the inhibitory CTLA-4 pathway has led to clinical benefit in a subset of patients with metastatic melanoma. Anti-CTLA-4 enhances T cell responses, including production of IFN-γ, which is a critical cytokine for host immune responses. However, the role of IFN-γ signaling in tumor cells in the setting of anti-CTLA-4 therapy remains unknown. Here, we demonstrate that patients identified as non-responders to anti-CTLA-4 (ipilimumab) have tumors with genomic defects in IFN-γ pathway genes. Furthermore, mice bearing melanoma tumors with knockdown of IFN-γ receptor 1 (IFNGR1) have impaired tumor rejection upon anti-CTLA-4 therapy. These data highlight that loss of the IFN-γ signaling pathway is associated with primary resistance to anti-CTLA-4 therapy. Our findings demonstrate the importance of tumor genomic data, especially IFN-γ related genes, as prognostic information for patients selected to receive treatment with immune checkpoint therapy.
Article
Helping NK cells find their way MICA and MICB proteins can be expressed on tumors and act as “kill me” signals to the immune system. But tumors often disguise themselves by shedding these proteins, which prevents specialized natural killer (NK) cells from recognizing and destroying the cancer. Ferrari de Andrade et al. engineered antibodies directed against the site responsible for the proteolytic shedding of MICA and MICB (see the Perspective by Cerwenka and Lanier). The approach effectively locked MICA and MICB onto tumors so that NK cells could spot them for elimination. The antibodies exhibited preclinical efficacy in multiple tumor models, including humanized melanoma. Furthermore, the strategy reduced lung cancer metastasis after NK cell–mediated tumor lysis. Science , this issue p. 1537 ; see also p. 1460
Article
Effector T cells have the capability of recognizing and killing cancer cells. However, whether tumors can become immune resistant through exclusion of effector T cells from the tumor microenvironment is not known. By using a tumor model resembling non-T cell-inflamed human tumors, we assessed whether adoptive T cell transfer might overcome failed spontaneous priming. Flow cytometric assays combined with intra-vital imaging indicated failed trafficking of effector T cells into tumors. Mechanistically, this was due to the absence of CXCL9/10, which we found to be produced by CD103+ dendritic cells (DCs) in T cell-inflamed tumors. Our data indicate that lack of CD103+ DCs within the tumor microenvironment dominantly resists the effector phase of an anti-tumor T cell response, contributing to immune escape.
Article
Cancer immunotherapy can induce long lasting responses in patients with metastatic cancers of a wide range of histologies. Broadening the clinical applicability of these treatments requires an improved understanding of the mechanisms limiting cancer immunotherapy. The interactions between the immune system and cancer cells are continuous, dynamic, and evolving from the initial establishment of a cancer cell to the development of metastatic disease, which is dependent on immune evasion. As the molecular mechanisms of resistance to immunotherapy are elucidated, actionable strategies to prevent or treat them may be derived to improve clinical outcomes for patients.
Article
Intratumoral dendritic cells (DC) bearing CD103 in mice or CD141 in humans drive intratumoral CD8(+) T cell activation. Using multiple strategies, we identified a critical role for these DC in trafficking tumor antigen to lymph nodes (LN), resulting in both direct CD8(+) T cell stimulation and antigen hand-off to resident myeloid cells. These effects all required CCR7. Live imaging demonstrated direct presentation to T cells in LN, and CCR7 loss specifically in these cells resulted in defective LN T cell priming and increased tumor outgrowth. CCR7 expression levels in human tumors correlate with signatures of CD141(+) DC, intratumoral T cells, and better clinical outcomes. This work identifies an ongoing pathway to T cell priming, which should be harnessed for tumor therapies.
Article
Background Approximately 75% of objective responses to anti–programmed death 1 (PD-1) therapy in patients with melanoma are durable, lasting for years, but delayed relapses have been noted long after initial objective tumor regression despite continuous therapy. Mechanisms of immune escape in this context are unknown. Methods We analyzed biopsy samples from paired baseline and relapsing lesions in four patients with metastatic melanoma who had had an initial objective tumor regression in response to anti–PD-1 therapy (pembrolizumab) followed by disease progression months to years later. Results Whole-exome sequencing detected clonal selection and outgrowth of the acquired resistant tumors and, in two of the four patients, revealed resistance-associated loss-of-function mutations in the genes encoding interferon-receptor–associated Janus kinase 1 (JAK1) or Janus kinase 2 (JAK2), concurrent with deletion of the wild-type allele. A truncating mutation in the gene encoding the antigen-presenting protein beta-2-microglobulin (B2M) was identified in a third patient. JAK1 and JAK2 truncating mutations resulted in a lack of response to interferon gamma, including insensitivity to its antiproliferative effects on cancer cells. The B2M truncating mutation led to loss of surface expression of major histocompatibility complex class I. Conclusions In this study, acquired resistance to PD-1 blockade immunotherapy in patients with melanoma was associated with defects in the pathways involved in interferon-receptor signaling and in antigen presentation. (Funded by the National Institutes of Health and others.)