Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Methods Cell Biol ; 180: 25-37, 2023.
Article in English | MEDLINE | ID: mdl-37890930

ABSTRACT

Mutation-associated neoantigens are key targets of tumor-specific T cells and thus play a major role in driving responses to immune checkpoint blockade (ICB) therapy in tumors with high mutational burden. However, only a small number of mutated peptides are actually presented by MHC molecules and only a minority can induce T cell responses. In addition, the recognition of these neoantigens by T cells is limited by the level of expression of the mutated gene product in the tumor cells. Preclinical studies have shown that radiation can convert the irradiated tumor into an in situ vaccine, leading to the priming of tumor-specific T cells and to the rejection of otherwise ICB-resistant tumors. There is now preclinical and clinical evidence that radiation can upregulate the expression of genes containing immunogenic mutations and expose them to the immune system. Therefore, the identification of neoantigens upregulated by radiation could help to predict which patients might benefit from treatment with combinations of radiotherapy and ICB and could also be incorporated into personalized neoantigen vaccination strategies. In this chapter, we present the pipeline that we used to identify relevant radiation-upregulated neoantigens in a poorly immunogenic mouse model of metastatic breast cancer.


Subject(s)
Antigens, Neoplasm , Neoplasms , Animals , Mice , Humans , Antigens, Neoplasm/genetics , Antigens, Neoplasm/chemistry , Neoplasms/genetics , Neoplasms/radiotherapy , T-Lymphocytes , Mutation , Peptides
2.
Nat Commun ; 14(1): 5146, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37620372

ABSTRACT

Radiation therapy (RT) increases tumor response to CTLA-4 inhibition (CTLA4i) in mice and in some patients, yet deep responses are rare. To identify rational combinations of immunotherapy to improve responses we use models of triple negative breast cancer highly resistant to immunotherapy in female mice. We find that CTLA4i promotes the expansion of CD4+ T helper cells, whereas RT enhances T cell clonality and enriches for CD8+ T cells with an exhausted phenotype. Combination therapy decreases regulatory CD4+ T cells and increases effector memory, early activation and precursor exhausted CD8+ T cells. A combined gene signature comprising these three CD8+ T cell clusters is associated with survival in patients. Here we show that targeting additional immune checkpoints expressed by intratumoral T cells, including PD1, is not effective, whereas CD40 agonist therapy recruits resistant tumors into responding to the combination of RT and CTLA4i, indicating the need to target different immune compartments.


Subject(s)
CD8-Positive T-Lymphocytes , Triple Negative Breast Neoplasms , Female , Animals , Mice , Humans , Immunotherapy , CD40 Antigens , Combined Modality Therapy , Triple Negative Breast Neoplasms/radiotherapy
3.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33476307

ABSTRACT

Neoantigens generated by somatic nonsynonymous mutations are key targets of tumor-specific T cells, but only a small number of mutations predicted to be immunogenic are presented by MHC molecules on cancer cells. Vaccination studies in mice and patients have shown that the majority of neoepitopes that elicit T cell responses fail to induce significant antitumor activity, for incompletely understood reasons. We report that radiotherapy upregulates the expression of genes containing immunogenic mutations in a poorly immunogenic mouse model of triple-negative breast cancer. Vaccination with neoepitopes encoded by these genes elicited CD8+ and CD4+ T cells that, whereas ineffective in preventing tumor growth, improved the therapeutic efficacy of radiotherapy. Mechanistically, neoantigen-specific CD8+ T cells preferentially killed irradiated tumor cells. Neoantigen-specific CD4+ T cells were required for the therapeutic efficacy of vaccination and acted by producing Th1 cytokines, killing irradiated tumor cells, and promoting epitope spread. Such a cytotoxic activity relied on the ability of radiation to upregulate class II MHC molecules as well as the death receptors FAS/CD95 and DR5 on the surface of tumor cells. These results provide proof-of-principle evidence that radiotherapy works in concert with neoantigen vaccination to improve tumor control.


Subject(s)
Antigens, Neoplasm/pharmacology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , Th1 Cells/immunology , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Female , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/radiation effects , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Radiotherapy , Th1 Cells/pathology , Vaccination
4.
Oncotarget ; 11(4): 452-461, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-32064049

ABSTRACT

Exercise is associated with favorable changes in circulating immune cells and improved survival in early-stage breast cancer patients, but the mechansims remain to be fully elucidated. Preclinical studies indicate that physical activity started before tumor injection reduces tumor incidence and progression. Here we tested whether exercise has anti-tumor effects in mice with established 4T1 mammary carcinoma, a mouse model of triple negative breast cancer. Exercise slowed tumor progression and reduced the tumor-induced accumulation of myeloid-derived suppressor cells (MDSCs). The reduction in MDSCs was accompanied by a relative increase in natural killer and CD8 T cell activation, suggesting that exercise restores a favorable immune environment. Consistently, exercise improved responses to a combination of programmed cell death protein 1 (PD-1) blockade and focal radiotherapy. These data support further investigations of exercise in breast cancer patients treated with combinations of immunotherapy and cytotoxic agents to improve cancer outcomes.

5.
Cancer Immunol Res ; 8(4): 465-478, 2020 04.
Article in English | MEDLINE | ID: mdl-32047024

ABSTRACT

The ability of focal radiotherapy to promote priming of tumor-specific CD8+ T cells and increase responses to immunotherapy is dependent on infiltration of the tumor by Batf3-dependent conventional dendritic cell type 1 (cDC1) cells. Such infiltration is driven by radiotherapy-induced IFN type I (IFN-I). Other signals may also modulate cDC1 infiltration of irradiated tumors. Here we found increased expression of adenosine-generating enzymes CD38 and CD73 in irradiated mouse and human breast cancer cells and increased adenosine in mouse tumors following radiotherapy. CD73 blockade alone had no effect. CD73 blockade with radiotherapy restored radiotherapy-induced cDC1 infiltration of tumors in settings where radiotherapy induction of IFN-I was suboptimal. In the absence of radiotherapy-induced IFN-I, blockade of CD73 was required for rejection of the irradiated tumor and for systemic tumor control (abscopal effect) in the context of cytotoxic T-lymphocyte-associated protein 4 blockade. These results suggest that CD73 may be a radiation-induced checkpoint, and that CD73 blockade in combination with radiotherapy and immune checkpoint blockade might improve patient response to therapy.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Adenosine/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Interferon Type I/immunology , Neoplasms/radiotherapy , 5'-Nucleotidase/immunology , Animals , Cell Line, Tumor , Female , Humans , Interferon Type I/radiation effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology
8.
Genome Med ; 11(1): 40, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31221199

ABSTRACT

The expression of antigens that are recognized by self-reactive T cells is essential for immune-mediated tumor rejection by immune checkpoint blockade (ICB) therapy. Growing evidence suggests that mutation-associated neoantigens drive ICB responses in tumors with high mutational burden. In most patients, only a few of the mutations in the cancer exome that are predicted to be immunogenic are recognized by T cells. One factor that limits this recognition is the level of expression of the mutated gene product in cancer cells. Substantial preclinical data show that radiation can convert the irradiated tumor into a site for priming of tumor-specific T cells, that is, an in situ vaccine, and can induce responses in otherwise ICB-resistant tumors. Critical for radiation-elicited T-cell activation is the induction of viral mimicry, which is mediated by the accumulation of cytosolic DNA in the irradiated cells, with consequent activation of the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon (IFN) genes (STING) pathway and downstream production of type I IFN and other pro-inflammatory cytokines. Recent data suggest that radiation can also enhance cancer cell antigenicity by upregulating the expression of a large number of genes that are involved in the response to DNA damage and cellular stress, thus potentially exposing immunogenic mutations to the immune system. Here, we discuss how the principles of antigen presentation favor the presentation of peptides that are derived from newly synthesized proteins in irradiated cells. These concepts support a model that incorporates the presence of immunogenic mutations in genes that are upregulated by radiation to predict which patients might benefit from treatment with combinations of radiotherapy and ICB.


Subject(s)
Antigen Presentation/radiation effects , Antigens, Neoplasm/genetics , Neoplasms/immunology , Radiotherapy/adverse effects , Animals , Antigens, Neoplasm/immunology , Humans , Mutation , Neoplasms/genetics , Neoplasms/radiotherapy
9.
Nat Med ; 24(12): 1845-1851, 2018 12.
Article in English | MEDLINE | ID: mdl-30397353

ABSTRACT

Focal radiation therapy enhances systemic responses to anti-CTLA-4 antibodies in preclinical studies and in some patients with melanoma1-3, but its efficacy in inducing systemic responses (abscopal responses) against tumors unresponsive to CTLA-4 blockade remained uncertain. Radiation therapy promotes the activation of anti-tumor T cells, an effect dependent on type I interferon induction in the irradiated tumor4-6. The latter is essential for achieving abscopal responses in murine cancers6. The mechanisms underlying abscopal responses in patients treated with radiation therapy and CTLA-4 blockade remain unclear. Here we report that radiation therapy and CTLA-4 blockade induced systemic anti-tumor T cells in chemo-refractory metastatic non-small-cell lung cancer (NSCLC), where anti-CTLA-4 antibodies had failed to demonstrate significant efficacy alone or in combination with chemotherapy7,8. Objective responses were observed in 18% of enrolled patients, and 31% had disease control. Increased serum interferon-ß after radiation and early dynamic changes of blood T cell clones were the strongest response predictors, confirming preclinical mechanistic data. Functional analysis in one responding patient showed the rapid in vivo expansion of CD8 T cells recognizing a neoantigen encoded in a gene upregulated by radiation, supporting the hypothesis that one explanation for the abscopal response is radiation-induced exposure of immunogenic mutations to the immune system.


Subject(s)
CD8-Positive T-Lymphocytes/radiation effects , CTLA-4 Antigen/antagonists & inhibitors , Ipilimumab/administration & dosage , Lung Neoplasms/therapy , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cell Line, Tumor , Combined Modality Therapy , Drug Resistance, Neoplasm/radiation effects , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Male , Middle Aged , Radiotherapy
10.
PLoS One ; 13(9): e0202512, 2018.
Article in English | MEDLINE | ID: mdl-30204750

ABSTRACT

Extra-cellular galectin-9 (gal-9) is an immuno-modulatory protein with predominant immunosuppressive effects. Inappropriate production of gal-9 has been reported in several human malignancies and viral diseases like nasopharyngeal, pancreatic and renal carcinomas, metastatic melanomas and chronic active viral hepatitis. Therefore therapeutic antibodies neutralizing extra-cellular gal-9 are expected to contribute to immune restoration in these pathological conditions. Two novel monoclonal antibodies targeting gal-9 -Gal-Nab 1 and 2-have been produced and characterized in this study. We report a protective effect of Gal-Nab1 and Gal-Nab2 on the apoptotic cell death induced by gal-9 in primary T cells. In addition, they inhibit late phenotypic changes observed in peripheral T cells that survive gal-9-induced apoptosis. Gal-Nab1 and Gal-Nab2 bind nearly identical, overlapping linear epitopes contained in the 213-224 amino-acid segments of gal-9. Nevertheless, they have some distinct functional characteristics suggesting that their three-dimensional epitopes are distinct. These differences are best demonstrated when gal-9 is applied on Jurkat cells where Gal-Nab1 is less efficient than Gal-Nab2 in the prevention of apoptotic cell death. In addition, Gal-Nab1 stimulates non-lethal phosphatidylserine translocation at the plasma membrane and calcium mobilization triggered by gal-9 in these cells. Both Gal-Nab1 and 2 cross-react with murine gal-9. They bind its natural as well as its recombinant form. This cross-species recognition will be an advantage for their assessment in pre-clinical tumor models.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Epitopes/immunology , Galectins/chemistry , T-Lymphocytes/cytology , Animals , Apoptosis/drug effects , Biological Transport , Calcium/metabolism , Galectins/adverse effects , Galectins/immunology , Humans , Immunization , Jurkat Cells , Mice , Phosphatidylserines/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
11.
Semin Cancer Biol ; 52(Pt 2): 125-134, 2018 10.
Article in English | MEDLINE | ID: mdl-29258856

ABSTRACT

Over the past few years, multiple immune checkpoint blockers (ICBs) have achieved unprecedented clinical success and have been approved by regulatory agencies for the treatment of an increasing number of malignancies. However, only a limited fraction of patients responds to ICBs employed as a standalone intervention, calling for the development of combinatorial regimens. Radiation therapy (RT) stands out as a very promising candidate for this purpose. Indeed, RT mediates antineoplastic effects not only by cytotoxic and cytostatic mechanisms, but also by modulating immunological functions, both locally (within the irradiated field) and systemically. As combinatorial regimens involving RT and ICBs are being developed and clinically tested at an accelerating pace, it is paramount to identify biomarkers that reliably predict the likelihood of individual patients to respond. Here, we discuss emerging biomarkers that may potentially predict the response of cancer patients to RT plus ICBs.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/immunology , Neoplasms/radiotherapy , Neoplasms/therapy , Animals , Antineoplastic Agents/immunology , Humans , Neoplasms/immunology , Radiotherapy/methods
12.
Cancer Immunol Res ; 6(2): 139-150, 2018 02.
Article in English | MEDLINE | ID: mdl-29180535

ABSTRACT

Immune checkpoint inhibitors activate T cells to reject tumors. Unique tumor mutations are key T-cell targets, but a comprehensive understanding of the nature of a successful antitumor T-cell response is lacking. To investigate the T-cell receptor (TCR) repertoire associated with treatment success versus failure, we used a well-characterized mouse carcinoma that is rejected by CD8 T cells in mice treated with radiotherapy (RT) and anti-CTLA-4 in combination, but not as monotherapy, and comprehensively analyzed tumor-infiltrating lymphocytes (TILs) by high-throughput sequencing of the TCRΒ CDR3 region. The combined treatment increased TIL density and CD8/CD4 ratio. Assessment of the frequency of T-cell clones indicated that anti-CTLA-4 resulted in fewer clones and a more oligoclonal repertoire compared with untreated tumors. In contrast, RT increased the CD8/CD4 ratio and broadened the TCR repertoire, and when used in combination with anti-CTLA-4, these selected T-cell clones proliferated. Hierarchical clustering of CDR3 sequences showed a treatment-specific clustering of TCRs that were shared by different mice. Abundant clonotypes were commonly shared between animals and yet treatment-specific. Analysis of amino-acid sequence similarities revealed a significant increase in the number and richness of dominant CDR3 motifs in tumors treated with RT + anti-CTLA-4 compared with control. The repertoire of TCRs reactive with a single tumor antigen recognized by CD8+ T cells was heterogeneous but highly clonal, irrespective of treatment. Overall, data support a model whereby a diverse TCR repertoire is required to achieve tumor rejection and may underlie the synergy between RT and CTLA-4 blockade. Cancer Immunol Res; 6(2); 139-50. ©2017 AACR.


Subject(s)
Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Breast Neoplasms/immunology , Breast Neoplasms/radiotherapy , CTLA-4 Antigen/immunology , Combined Modality Therapy , Female , Humans , Mice , Random Allocation , Xenograft Model Antitumor Assays
13.
Oncoimmunology ; 6(11): e1373237, 2017.
Article in English | MEDLINE | ID: mdl-29147629

ABSTRACT

Immune checkpoint blockers (ICBs) are literally revolutionizing the clinical management of an ever more diversified panel of oncological indications. Although considerable attention persists around the inhibition of cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1) signaling, several other co-inhibitory T-cell receptors are being evaluated as potential targets for the development of novel ICBs. Moreover, substantial efforts are being devoted to the identification of biomarkers that reliably predict the likelihood of each patient to obtain clinical benefits from ICBs in the absence of severe toxicity. Tailoring the delivery of specific ICBs or combinations thereof to selected patient populations in the context of precision medicine programs constitutes indeed a major objective of the future of ICB-based immunotherapy. Here, we discuss recent preclinical and clinical advances on the development of ICBs for oncological indications.

14.
Phys Rev Lett ; 118(26): 267201, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28707912

ABSTRACT

The quantum spin liquid material herbertsmithite is described by an antiferromagnetic Heisenberg model on the kagome lattice with a non-negligible Dzyaloshinskii-Moriya interaction (DMI). A well-established phase transition to the q=0 long-range order occurs in this model when the DMI strength increases, but the precise nature of a small-DMI phase remains controversial. Here, we describe a new phase obtained from Schwinger-boson mean-field theory that is stable at small DMI, and which can explain the dispersionless spectrum seen in the inelastic neutron scattering experiment by Han et al. [Nature (London) 492, 406 (2012)NATUAS0028-083610.1038/nature11659]. It is a time-reversal symmetry breaking Z_{2} spin liquid, with the unique property of a small and constant spin gap in an extended region of the Brillouin zone. The phase diagram as a function of DMI and spin size is given, and dynamical spin structure factors are presented.

15.
Front Immunol ; 8: 229, 2017.
Article in English | MEDLINE | ID: mdl-28348554

ABSTRACT

The immunostimulatory properties of radiation therapy (RT) have recently generated widespread interest due to preclinical and clinical evidence that tumor-localized RT can sometimes induce antitumor immune responses mediating regression of non-irradiated metastases (abscopal effect). The ability of RT to activate antitumor T cells explains the synergy of RT with immune checkpoint inhibitors, which has been well documented in mouse tumor models and is supported by observations of more frequent abscopal responses in patients refractory to immunotherapy who receive RT during immunotherapy. However, abscopal responses following RT remain relatively rare in the clinic, and antitumor immune responses are not effectively induced by RT against poorly immunogenic mouse tumors. This suggests that in order to improve the pro-immunogenic effects of RT, it is necessary to identify and overcome the barriers that pre-exist and/or are induced by RT in the tumor microenvironment. On the one hand, RT induces an immunogenic death of cancer cells associated with release of powerful danger signals that are essential to recruit and activate dendritic cells (DCs) and initiate antitumor immune responses. On the other hand, RT can promote the generation of immunosuppressive mediators that hinder DCs activation and impair the function of effector T cells. In this review, we discuss current evidence that several inhibitory pathways are induced and modulated in irradiated tumors. In particular, we will focus on factors that regulate and limit radiation-induced immunogenicity and emphasize current research on actionable targets that could increase the effectiveness of radiation-induced in situ tumor vaccination.

16.
J Biol Chem ; 290(27): 16797-811, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25947381

ABSTRACT

Galectin-9 (gal-9) is a multifunctional ß-galactoside-binding lectin, frequently released in the extracellular medium, where it acts as a pleiotropic immune modulator. Despite its overall immunosuppressive effects, a recent study has reported bimodal action of gal-9 on human resting blood T cells with apoptosis occurring in the majority of them, followed by a wave of activation and expansion of Th1 cells in the surviving population. Our knowledge of the signaling events triggered by exogenous gal-9 in T cells remains limited. One of these events is cytosolic calcium (Ca(2+)) release reported in some murine and human T cells. The aim of this study was to investigate the contribution of Ca(2+) mobilization to apoptotic and nonapoptotic effects of exogenous gal-9 in human T cells. We found that the T cell receptor (TCR)-CD3 complex and the Lck kinase were required for Ca(2+) mobilization but not for apoptosis induction in Jurkat cells. These data were confirmed in human CD4(+) T cells from peripheral blood as follows: a specific Lck chemical inhibitor abrogated Ca(2+) mobilization but not apoptosis induction. Moreover, Lck activity was also required for the production of Th1-type cytokines, i.e. interleukin-2 and interferon-γ, which resulted from gal-9 stimulation in peripheral CD4(+) T cells. These findings indicate that gal-9 acts on T cells by two distinct pathways as follows: one mimicking antigen-specific activation of the TCR with a mandatory contribution of proximal elements of the TCR complex, especially Lck, and another resulting in apoptosis that is independent of this complex.


Subject(s)
Apoptosis , CD3 Complex/metabolism , Galectins/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , CD3 Complex/genetics , Calcium/metabolism , Cytokines/genetics , Cytokines/metabolism , Galectins/genetics , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology
17.
Phys Rev Lett ; 108(20): 207204, 2012 May 18.
Article in English | MEDLINE | ID: mdl-23003183

ABSTRACT

Inspired by the recent discovery of a new instability towards a chiral phase of the classical Heisenberg model on the kagome lattice, we propose a specific chiral spin liquid that reconciles different, well-established results concerning both the classical and quantum models. This proposal is analyzed in an extended mean-field Schwinger boson framework encompassing time reversal symmetry breaking phases, which allows both a classical and a quantum phase description. At low temperatures, we find that quantum fluctuations favor this chiral phase, which is stable against small perturbations of second- and third-neighbor interactions. For spin-1/2, this phase may be, beyond the mean field, a chiral gapped spin liquid. Such a phase is consistent with the density matrix renormalization group results of Yan et al. [Science 332, 1173 (2011)]. Mysterious features of the low-lying excitations of exact diagonalization spectra also find an explanation in this framework. Moreover, thermal fluctuations compete with quantum ones and induce a transition from this flux phase to a planar zero flux phase at a nonzero value of the renormalized temperature (T/S2), reconciling these results with those obtained for the classical system.

SELECTION OF CITATIONS
SEARCH DETAIL
...