Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Nat Immunol ; 18(12): 1342-1352, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29058703

ABSTRACT

T cells reorganize their metabolic profiles after being activated, but the systemic metabolic effect of sustained activation of the immune system has remained unexplored. Here we report that augmented T cell responses in Pdcd1-/- mice, which lack the inhibitory receptor PD-1, induced a metabolic serum signature characterized by depletion of amino acids. We found that the depletion of amino acids in serum was due to the accumulation of amino acids in activated Pdcd1-/- T cells in the lymph nodes. A systemic decrease in tryptophan and tyrosine led to substantial deficiency in the neurotransmitters serotonin and dopamine in the brain, which resulted in behavioral changes dominated by anxiety-like behavior and exacerbated fear responses. Together these data indicate that excessive activation of T cells causes a systemic metabolomic shift with consequences that extend beyond the immune system.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Fear/physiology , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/genetics , T-Lymphocytes/immunology , Amino Acids/blood , Animals , Brain/metabolism , Dopamine/deficiency , Interferon-gamma/blood , Kynurenine/blood , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/deficiency , Serotonin/deficiency , T-Lymphocytes/metabolism , Tryptophan/metabolism , Tyrosine/metabolism
2.
Nature ; 599(7885): 471-476, 2021 11.
Article in English | MEDLINE | ID: mdl-34732892

ABSTRACT

Small, soluble metabolites not only are essential intermediates in intracellular biochemical processes, but can also influence neighbouring cells when released into the extracellular milieu1-3. Here we identify the metabolite and neurotransmitter GABA as a candidate signalling molecule synthesized and secreted by activated B cells and plasma cells. We show that B cell-derived GABA promotes monocyte differentiation into anti-inflammatory macrophages that secrete interleukin-10 and inhibit CD8+ T cell killer function. In mice, B cell deficiency or B cell-specific inactivation of the GABA-generating enzyme GAD67 enhances anti-tumour responses. Our study reveals that, in addition to cytokines and membrane proteins, small metabolites derived from B-lineage cells have immunoregulatory functions, which may be pharmaceutical targets allowing fine-tuning of immune responses.


Subject(s)
B-Lymphocytes/metabolism , Interleukin-10/immunology , Macrophages/metabolism , Neoplasms/immunology , gamma-Aminobutyric Acid/metabolism , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Female , Gene Deletion , Glutamate Decarboxylase/deficiency , Glutamate Decarboxylase/genetics , Humans , Inflammation/immunology , Inflammation/prevention & control , Macrophages/immunology , Male , Mice , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , gamma-Aminobutyric Acid/biosynthesis
3.
J Am Chem Soc ; 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38848460

ABSTRACT

Cancer immune therapies, particularly programmed cell death protein 1 (PD-1) blockade immunotherapy, falter in aged individuals due to compromised T-cell immunity. Spermidine, a biogenic polyamine that declines along with aging, shows promise in restoring antitumor immunity by enhancing mitochondrial fatty acid oxidation (FAO). Herein, we report a spermidine-based chemoproteomic probe (probe 2) that enables profiling of spermidine-binding proteins and screening for small-molecule enhancers of mitochondrial FAO. Chemoproteomic profiling by the probe revealed 140 proteins engaged in cellular interaction with spermidine, with a significant majority being mitochondrial proteins. Hydroxyl coenzyme A (CoA) dehydrogenase subunits α (HADHA) and other lipid metabolism-linked proteins are among the mitochondrial proteins that have attracted considerable interest. Screening spermidine analogs with the probe led to the discovery of compound 13, which interacts with these lipid metabolism-linked proteins and activates HADHA. This simple and biostable synthetic compound we named "spermimic" mirrors spermidine's ability to enhance mitochondrial bioenergetics and displays similar effectiveness in augmenting PD-1 blockade therapy in mice. This study lays the foundation for developing small-molecule activators of antitumor immunity, offering potential in combination cancer immunotherapy.

4.
Microvasc Res ; 151: 104596, 2024 01.
Article in English | MEDLINE | ID: mdl-37625620

ABSTRACT

In the later stages of angiogenesis, the vascular sprout transitions into a functional vessel by fusing with a target vessel. Although this process appears to routinely occur in embryonic tissue, the biologic rules for sprout fusion and lumenization in adult regenerating tissue are unknown. To investigate this process, we grafted portions of the regenerating post-pneumonectomy lung onto the chick chorioallantoic membrane (CAM). Grafts from all 4 lobes of the post-pneumonectomy right lung demonstrated peri-graft angiogenesis as reflected by fluorescent plasma markers; however, fluorescent microsphere perfusion primarily occurred in the lobe of the lung that is the dominant site of post-pneumonectomy angiogenesis-namely, the cardiac lobe. Vascularization of the cardiac lobe grafts was confirmed by active tissue growth (p < .05). Functional vascular connections between the cardiac lobe and the CAM vascular network were demonstrated by confocal fluorescence microscopy as well as corrosion casting and scanning electron microscopy (SEM). Bulk transcriptional profiling of the cardiac lobe demonstrated the enhanced expression of many genes relative to alveolar epithelial cell (CD11b-/CD31-) control cells, but only the upregulation of Ereg and Fgf6 compared to the less well-vascularized right upper lobe. The growth of actively regenerating non-neoplastic adult tissue on the CAM demonstrates that functional lumenization can occur between species (mouse and chick) and across the developmental spectrum (adult and embryo).


Subject(s)
Chorioallantoic Membrane , Neovascularization, Physiologic , Mice , Animals , Chorioallantoic Membrane/blood supply , Chickens , Neovascularization, Pathologic , Lung
5.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34088845

ABSTRACT

CD8+ T cells play a central role in antitumor immune responses that kill cancer cells directly. In aged individuals, CD8+ T cell immunity is strongly suppressed, which is associated with cancer and other age-related diseases. The mechanism underlying this age-related decrease in immune function remains largely unknown. This study investigated the role of T cell function in age-related unresponsiveness to PD-1 blockade cancer therapy. We found inefficient generation of CD44lowCD62Llow CD8+ T cell subset (P4) in draining lymph nodes of tumor-bearing aged mice. In vitro stimulation of naive CD8+ T cells first generated P4 cells, followed by effector/memory T cells. The P4 cells contained a unique set of genes related to enzymes involved in one-carbon (1C) metabolism, which is critical to antigen-specific T cell activation and mitochondrial function. Consistent with this finding, 1C-metabolism-related gene expression and mitochondrial respiration were down-regulated in aged CD8+ T cells compared with young CD8+ T cells. In aged OVA-specific T cell receptor (TCR) transgenic mice, ZAP-70 was not activated, even after inoculation with OVA-expressing tumor cells. The attenuation of TCR signaling appeared to be due to elevated expression of CD45RB phosphatase in aged CD8+ T cells. Surprisingly, strong stimulation by nonself cell injection into aged PD-1-deficient mice restored normal levels of CD45RB and ameliorated the emergence of P4 cells and 1C metabolic enzyme expression in CD8+ T cells, and antitumor activity. These findings indicate that impaired induction of the P4 subset may be responsible for the age-related resistance to PD-1 blockade, which can be rescued by strong TCR stimulation.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Hyaluronan Receptors/immunology , L-Selectin/immunology , Neoplasms, Experimental/immunology , Aging/genetics , Animals , Cell Line, Tumor , Hyaluronan Receptors/genetics , L-Selectin/genetics , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/immunology
6.
Proc Natl Acad Sci U S A ; 117(38): 23674-23683, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32907933

ABSTRACT

The gut microbiome has garnered attention as an effective target to boost immunity and improve cancer immunotherapy. We found that B cell-defective (BCD) mice, such as µ-membrane targeted deletion (µMT) and activation-induced cytidine deaminase (AID) knockouts (KOs), have elevated antitumor immunity under specific pathogen-free but not germ-free conditions. Microbial dysbiosis in these BCD mice enriched the type I IFN (IFN) signature in mucosal CD8+ T cells, resulting in up-regulation of the type I IFN-inducible protein stem cell antigen-1 (Sca-1). Among CD8+ T cells, naïve cells predominantly circulate from the gut to the periphery, and those that had migrated from the mesenteric lymph nodes (mLNs) to the periphery had significantly higher expression of Sca-1. The gut-educated Sca-1+ naïve subset is endowed with enhanced mitochondrial activity and antitumor effector potential. The heterogeneity and functional versatility of the systemic naïve CD8+ T cell compartment was revealed by single-cell analysis and functional assays of CD8+ T cell subpopulations. These results indicate one of the potential mechanisms through which microbial dysbiosis regulates antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , Interferon Type I/immunology , Neoplasms, Experimental/immunology , Animals , Antigens, Ly/immunology , Antigens, Ly/metabolism , B-Lymphocytes , Cell Line, Tumor , Cells, Cultured , Dysbiosis/immunology , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Interferon Type I/metabolism , Lymph Nodes/cytology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology
7.
Int Immunol ; 33(1): 17-26, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32622347

ABSTRACT

Energy metabolism plays an important role in proliferating cells. Recent reports indicate that metabolic regulation or metabolic products can control immune cell differentiation, fate and reactions. Cancer immunotherapy based on blockade of programmed cell death protein 1 (PD-1) has been used worldwide, but a significant fraction of patients remain unresponsive. Therefore, clarifying the mechanisms and overcoming the unresponsiveness are urgent issues. Because cancer immunity consists of interactions between the cancer and host immune cells, there has recently been a focus on the metabolic interactions and/or competition between the tumor and the immune system to address these issues. Cancer cells render their microenvironment immunosuppressive, driving T-cell dysfunction or exhaustion, which is advantageous for cancer cell survival. However, accumulating mechanistic evidence of T-cell and cancer cell metabolism has gradually revealed that controlling the metabolic pathways of either type of cell can overcome T-cell dysfunction and reprogram the metabolic balance in the tumor microenvironment. Here, we summarize the role of immune metabolism in T-cell-based immune surveillance and cancer immune escape. This new concept has boosted the development of combination therapy and predictive biomarkers in cancer immunotherapy with immune checkpoint inhibitors.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Cell Differentiation/immunology , Combined Modality Therapy , Energy Metabolism/immunology , Humans , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tumor Escape/immunology , Tumor Microenvironment/immunology
8.
Int Immunol ; 32(8): 519-531, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32219331

ABSTRACT

Chronic hepatitis B is now controllable when treated with nucleoside reverse transcriptase inhibitors (NRTIs), which inhibit hepatitis B virus (HBV) replication. However, once the NRTIs are discontinued, most patients relapse, necessitating lifelong NRTIs treatment. HBV infection relapse is assumed to be caused by the persistent existence of covalently closed circular DNA (cccDNA) in the nuclei of infected hepatocytes. The mechanism by which cccDNA-positive hepatocytes escape immune surveillance during NRTIs treatment remains elusive. Entecavir (ETV), a commonly used NRTI, post-transcriptionally up-regulates programmed cell death-ligand 1 (PD-L1), an immune checkpoint molecule, on the cell surface of hepatocytes regardless of HBV infection. Up-regulation by ETV depends on up-regulation of CKLF-like MARVEL transmembrane domain-containing 6, a newly identified potent regulator of PD-L1 expression on the cell surface. ETV-treated hepatic cells suppressed the activity of primary CD3 T cells and programmed cell death protein-1 (PD-1)-over-expressed Jurkat cells. Finally, ETV induces PD-L1 in primary hepatocytes infected by HBV. These results provide evidence that ETV considerably up-regulates PD-L1 on the cell surface of infected hepatocytes, which may be one of the mechanisms by which infected hepatocytes subvert immune surveillance.


Subject(s)
Antiviral Agents/pharmacology , B7-H1 Antigen/immunology , Guanine/analogs & derivatives , Hepatocytes/drug effects , MARVEL Domain-Containing Proteins/immunology , Up-Regulation/drug effects , B7-H1 Antigen/genetics , Cell Line, Tumor , Guanine/pharmacology , Hepatocytes/immunology , Humans , Surface Properties , Up-Regulation/immunology
9.
Int J Clin Oncol ; 25(5): 790-800, 2020 May.
Article in English | MEDLINE | ID: mdl-31900651

ABSTRACT

Programmed cell death 1 (PD-1) signal receptor blockade has revolutionized the field of cancer therapy. Despite their considerable potential for treating certain cancers, drugs targeting PD-1 still present two main drawbacks: the substantial number of unresponsive patients and/or patients showing recurrences, and side effects associated with the autoimmune response. These drawbacks highlight the need for further investigation of the mechanisms underlying the therapeutic effects, as well as the need to develop novel biomarkers to predict the lack of treatment response and to monitor potential adverse events. Combination therapy is a promising approach to improve the efficacy of PD-1 blockade therapy. Considering the increasing number of patients with cancer worldwide, solving the above issues is central to the field of cancer immunotherapy. In this review, we discuss these issues and clinical perspectives associated with PD-1 blockade cancer immunotherapy.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , CTLA-4 Antigen/immunology , Combined Modality Therapy , Humans , Immunotherapy/adverse effects , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/metabolism
10.
Proc Natl Acad Sci U S A ; 114(5): E761-E770, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096382

ABSTRACT

Although immunotherapy by PD-1 blockade has dramatically improved the survival rate of cancer patients, further improvement in efficacy is required to reduce the fraction of less sensitive patients. In mouse models of PD-1 blockade therapy, we found that tumor-reactive cytotoxic T lymphocytes (CTLs) in draining lymph nodes (DLNs) carry increased mitochondrial mass and more reactive oxygen species (ROS). We show that ROS generation by ROS precursors or indirectly by mitochondrial uncouplers synergized the tumoricidal activity of PD-1 blockade by expansion of effector/memory CTLs in DLNs and within the tumor. These CTLs carry not only the activation of mechanistic target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) but also an increment of their downstream transcription factors such as PPAR-gamma coactivator 1α (PGC-1α) and T-bet. Furthermore, direct activators of mTOR, AMPK, or PGC-1α also synergized the PD-1 blockade therapy whereas none of above-mentioned chemicals alone had any effects on tumor growth. These findings will pave a way to developing novel combinatorial therapies with PD-1 blockade.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Mitochondria/metabolism , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/immunology , AMP-Activated Protein Kinases/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Biphenyl Compounds , Cell Line, Tumor , Cytokines/immunology , Lymph Nodes/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Morpholines/pharmacology , Morpholines/therapeutic use , Neoplasms/immunology , Neoplasms/metabolism , Oxygen Consumption , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Pyrones/pharmacology , Pyrones/therapeutic use , Reactive Oxygen Species/metabolism , T-Lymphocytes, Cytotoxic/metabolism , TOR Serine-Threonine Kinases/metabolism , Thiophenes/pharmacology , Thiophenes/therapeutic use , Triazines/pharmacology , Triazines/therapeutic use
11.
Gan To Kagaku Ryoho ; 47(1): 16-20, 2020 Jan.
Article in Japanese | MEDLINE | ID: mdl-32381855

ABSTRACT

Immune checkpoint blockade inhibition is a therapy which interferes with inhibitory signals placed upon immune cells, thereby eliciting anti-tumor responses. Although programmed death-1(PD-1)blockade therapy has been shown to be highly effective in clinical use, certain population of patients still fail to respond. Therefore, it is critical to determine how therapeutic efficacy of checkpoint inhibition can be enhanced. Recently, it has been shown that intracellular metabolism plays an important role in T cell differentiation and function. Because an effective tumor response relies on the differentiation of tumor-responsive effector cytotoxic T lymphocytes(CTLs), understanding such mechanisms will be essential for developing an improved therapeutic approach. Experiments on tumor-bearing mice have displayed strong anti-tumor responses upon combination therapy ofPD -1 blockade and enhanced mitochondrial metabolism, through increased production ofreactive oxygen species(ROS); upregulation ofmechanistic target ofrapamycin(mTOR)and AMP-activated protein kinase(AMPK); and upregulation ofperoxisome proliferator-activated receptor gamma coactivator 1-alpha(PGC-1a). In addition, the use of bezafibrate, a peroxisome proliferator-activated receptor(PPAR)agonist, in combination with PD-1 blockade resulted in improved effector function in CTLs. In this review, we will explore the mechanism behind T cell metabolism under the context ofcheckpoint blockade therapy, as well as how modulating T cell metabolism yields synergistic anti-tumor effects in combination with PD-1 blockade therapy.


Subject(s)
T-Lymphocytes, Cytotoxic , Animals , Antineoplastic Agents , Humans , Lymphocyte Activation , Neoplasms , Programmed Cell Death 1 Receptor
12.
Br J Cancer ; 120(1): 115-127, 2019 01.
Article in English | MEDLINE | ID: mdl-30382166

ABSTRACT

BACKGROUND: V-domain Ig suppressor of T cell activation (VISTA) is a novel inhibitory immune-checkpoint protein. VISTA expression on tumour cells and the associated regulatory mechanisms remain unclear. We investigated VISTA expression and function in tumour cells, and evaluated its mechanism and activity. METHODS: VISTA in tumour cells was assessed by tissue microarray analysis, immunohistochemical staining and western blot. A series of in vitro assays were used to determine the function of tumour-expressed VISTA. In vivo efficacy was evaluated in syngeneic models. RESULTS: VISTA was highly expressed in human ovarian and endometrial cancers. Upregulation of VISTA in endometrial cancer was related to the methylation status of the VISTA promoter. VISTA in tumour cells suppressed T cell proliferation and cytokine production in vitro, and decreased the tumour-infiltrating CD8+ T cells in vivo. Anti-VISTA antibody prolonged the survival of tumour-bearing mice. CONCLUSIONS: This is the first demonstration that VISTA is highly expressed in human ovarian and endometrial cancer cells, and that anti-VISTA antibody treatment significantly prolongs the survival of mice bearing tumours expressing high levels of VISTA. The data suggest that VISTA is a novel immunosuppressive factor within the tumour microenvironment, as well as a new target for cancer immunotherapy.


Subject(s)
B7 Antigens/genetics , Endometrial Neoplasms/genetics , Ovarian Neoplasms/genetics , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Female , Humans , Immunotherapy , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
13.
J Immunol ; 198(3): 1056-1065, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28003379

ABSTRACT

The human invariant NK (iNK) TCR is largely composed of the invariant TCR Vα24-Jα18 chain and semivariant TCR Vß11 chains with variable CDR3ß sequences. The direct role of CDR3ß in Ag recognition has been studied extensively. Although it was noted that CDR3ß can interact with CDR3α, how this interaction might indirectly influence Ag recognition is not fully elucidated. We observed that the third position of Vß11 CDR3 can encode an Arg or Ser residue as a result of somatic rearrangement. Clonotypic analysis of the two iNK TCR types with a single amino acid substitution revealed that the staining intensity by anti-Vα24 Abs depends on whether Ser or Arg is encoded. When stained with an anti-Vα24-Jα18 Ab, human primary invariant NKT cells could be divided into Vα24 low- and high-intensity subsets, and Arg-encoding TCR Vß11 chains were more frequently isolated from the Vα24 low-intensity subpopulation compared with the Vα24 high-intensity subpopulation. The Arg/Ser substitution also influenced Ag recognition as determined by CD1d multimer staining and CD1d-restricted functional responses. Importantly, in silico modeling validated that this Ser-to-Arg mutation could alter the structure of the CDR3ß loop, as well as the CDR3α loop. Collectively, these results indicate that the Arg/Ser encoded at the third CDR3ß residue can effectively modulate the overall structure of, and Ag recognition by, human iNK TCRs.


Subject(s)
Antigens/immunology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/immunology , Antigens, CD1d/immunology , Complementarity Determining Regions/chemistry , Humans , Molecular Dynamics Simulation
14.
Curr Top Microbiol Immunol ; 410: 75-97, 2017.
Article in English | MEDLINE | ID: mdl-28929192

ABSTRACT

Immunity developed to defend our bodies from foreign particles, including bacteria and viruses. Although effector cells responsible for acquired immunity, mainly T cells, and B cells, are able to distinguish self from non-self, they sometimes attack the body's tissues because of imperfect central tolerance. Several immune check points developed to limit overactivation of these cells. One of the most important immune checkpoints is programmed cell death-1 (PD-1), which is expressed mainly on activated lymphocytes. As its ligands (PD-Ls) are expressed widely in the body and affect the responses against self and foreign antigens, controlling PD-1/PD-L interactions enables the management of several immune-related diseases such as autoimmune disease, virus infection, and cancers. Currently, the strategy of PD-1/ PD-L1 blockade has already been applied to clinical cancer therapy, providing evidences that PD-1 signal is one of the main factors of cancer immune escape in humans. The dramatic efficacy of PD-1 blockade in cancer immunotherapy, promises the control of other immune diseases by PD-1 signal modulation. In this review, we summarize the history of PD-1, subsequent basic studies, and their application to the clinic.


Subject(s)
B7-H1 Antigen , Immunity , Programmed Cell Death 1 Receptor , Humans , Immunotherapy
15.
J Biomed Sci ; 24(1): 26, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28376884

ABSTRACT

Immunotherapy has recently emerged as the fourth pillar of cancer treatment, joining surgery, radiation, and chemotherapy. While early immunotherapies focused on accelerating T-cell activity, current immune-checkpoint inhibitors take the brakes off the anti-tumor immune responses. Successful clinical trials with PD-1 monoclonal antibodies and other immune-checkpoint inhibitors have opened new avenues in cancer immunology. However, the failure of a large subset of cancer patients to respond to these new immunotherapies has led to intensified research on combination therapies and predictive biomarkers. Here we summarize the development of PD-1-blockade immunotherapy and current issues in its clinical use.


Subject(s)
Antineoplastic Agents/pharmacology , Immunologic Factors/therapeutic use , Immunotherapy , Neoplasms/therapy , Programmed Cell Death 1 Receptor/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/metabolism , Humans , Signal Transduction
16.
J Immunol ; 194(7): 3487-500, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25710913

ABSTRACT

TCRα- and ß-chains cooperatively recognize peptide-MHC complexes. It has been shown that a "chain-centric" TCR hemichain can, by itself, dictate MHC-restricted Ag specificity without requiring major contributions from the paired TCR counterchain. Little is known, however, regarding the relative contributions and roles of chain-centric and its counter, non-chain-centric, hemichains in determining T cell avidity. We comprehensively analyzed a thymically unselected T cell repertoire generated by transducing the α-chain-centric HLA-A*02:01(A2)/MART127-35 TCRα, clone SIG35α, into A2-matched and unmatched postthymic T cells. Regardless of their HLA-A2 positivity, a substantial subset of peripheral T cells transduced with SIG35α gained reactivity for A2/MART127-35. Although the generated A2/MART127-35-specific T cells used various TRBV genes, TRBV27 predominated with >10(2) highly diverse and unique clonotypic CDR3ß sequences. T cells individually reconstituted with various A2/MART127-35 TRBV27 TCRß genes along with SIG35α possessed a wide range (>2 log orders) of avidity. Approximately half possessed avidity higher than T cells expressing clone DMF5, a naturally occurring A2/MART127-35 TCR with one of the highest affinities. Importantly, similar findings were recapitulated with other self-Ags. Our results indicate that, although a chain-centric TCR hemichain determines Ag specificity, the paired counterchain can regulate avidity over a broad range (>2 log orders) without compromising Ag specificity. TCR chain centricity can be exploited to generate a thymically unselected Ag-specific T cell repertoire, which can be used to isolate high-avidity antitumor T cells and their uniquely encoded TCRs rarely found in the periphery because of tolerance.


Subject(s)
Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Immunophenotyping , MART-1 Antigen/chemistry , MART-1 Antigen/genetics , MART-1 Antigen/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding , Protein Multimerization , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transduction, Genetic
17.
J Autoimmun ; 68: 39-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26748722

ABSTRACT

Invariant natural killer T (iNKT) cells are a subset of T lymphocytes that recognize lipid ligands presented by monomorphic CD1d. Human iNKT T cell receptor (TCR) is largely composed of invariant Vα24 (Vα24i) TCRα chain and semi-variant Vß11 TCRß chain, where complementarity-determining region (CDR)3ß is the sole variable region. One of the characteristic features of iNKT cells is that they retain autoreactivity even after the thymic selection. However, the molecular features of human iNKT TCR CDR3ß sequences that regulate autoreactivity remain unknown. Since the numbers of iNKT cells with detectable autoreactivity in peripheral blood is limited, we introduced the Vα24i gene into peripheral T cells and generated a de novo human iNKT TCR repertoire. By stimulating the transfected T cells with artificial antigen presenting cells (aAPCs) presenting self-ligands, we enriched strongly autoreactive iNKT TCRs and isolated a large panel of human iNKT TCRs with a broad range autoreactivity. From this panel of unique iNKT TCRs, we deciphered three CDR3ß sequence motifs frequently encoded by strongly-autoreactive iNKT TCRs: a VD region with 2 or more acidic amino acids, usage of the Jß2-5 allele, and a CDR3ß region of 13 amino acids in length. iNKT TCRs encoding 2 or 3 sequence motifs also exhibit higher autoreactivity than those encoding 0 or 1 motifs. These data facilitate our understanding of the molecular basis for human iNKT cell autoreactivity involved in immune responses associated with human disease.


Subject(s)
Amino Acid Motifs , Autoimmunity , Complementarity Determining Regions/genetics , Receptors, Natural Killer Cell/immunology , Receptors, Natural Killer Cell/metabolism , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Cell Line , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/metabolism , Gene Expression , Humans , Immunophenotyping , Phenotype , Protein Multimerization , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
18.
Rinsho Ketsueki ; 57(10): 2224-2231, 2016.
Article in Japanese | MEDLINE | ID: mdl-27795534

ABSTRACT

Immune checkpoint inhibitors, especially anti-programmed cell death-1 (PD-1) antibodies, have revolutionized cancer therapy. A PD-1 antibody, nivolumab, was the first of these agents to be approved by the Pharmaceuticals and Medical Devices Agency (PMDA) of Japan, as a new cancer drug for melanoma, in July 2014. While PD-1 mAb therapy has so far been approved only for untreated malignant melanomas and non-small cell lung cancer, many clinical studies on various types of cancer have been conducted worldwide. Immune checkpoint inhibitors target lymphocytes rather than cancer cells, and evoke an anti-tumor immune reaction. Since the activated lymphocytes recognize various tumor-associated antigens including a mutated antigen, immune checkpoint inhibitors exhibit continuous long-term effectiveness, despite the generation of genetic mutations in cancer cells. As compared with previous cancer treatments, immune checkpoint inhibitors show superior efficacy against tumors with fewer side effects. Therefore, these novel immune checkpoint inhibitor agents are anticipated to become a 4th cancer treatment option following surgery, chemotherapy, and radiation therapy. Herein, we review the main clinical results of PD-1 mAb cancer immunotherapy obtained to date and discuss issues relevant to administering this form of treatment.


Subject(s)
Neoplasms/immunology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Animals , Biomarkers, Tumor/blood , Clinical Trials as Topic , Humans , Immunotherapy , Molecular Targeted Therapy , Neoplasms/diagnosis , Prognosis
20.
J Clin Invest ; 134(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38557498

ABSTRACT

BACKGROUNDPrecise stratification of patients with non-small cell lung cancer (NSCLC) is needed for appropriate application of PD-1/PD-L1 blockade therapy.METHODSWe measured soluble forms of the immune-checkpoint molecules PD-L1, PD-1, and CTLA-4 in plasma of patients with advanced NSCLC before PD-1/PD-L1 blockade. A prospective biomarker-finding trial (cohort A) included 50 previously treated patients who received nivolumab. A retrospective observational study was performed for patients treated with any PD-1/PD-L1 blockade therapy (cohorts B and C), cytotoxic chemotherapy (cohort D), or targeted therapy (cohort E). Plasma samples from all patients were assayed for soluble immune-checkpoint molecules with a highly sensitive chemiluminescence-based assay.RESULTSNonresponsiveness to PD-1/PD-L1 blockade therapy was associated with higher concentrations of these soluble immune factors among patients with immune-reactive (hot) tumors. Such an association was not apparent for patients treated with cytotoxic chemotherapy or targeted therapy. Integrative analysis of tumor size, PD-L1 expression in tumor tissue (tPD-L1), and gene expression in tumor tissue and peripheral CD8+ T cells revealed that high concentrations of the 3 soluble immune factors were associated with hyper or terminal exhaustion of antitumor immunity. The combination of soluble PD-L1 (sPD-L1) and sCTLA-4 efficiently discriminated responsiveness to PD-1/PD-L1 blockade among patients with immune-reactive tumors.CONCLUSIONCombinations of soluble immune factors might be able to identify patients unlikely to respond to PD-1/PD-L1 blockade as a result of terminal exhaustion of antitumor immunity. Our data suggest that such a combination better predicts, along with tPD-L1, for the response of patients with NSCLC.TRIAL REGISTRATIONUMIN000019674.FUNDINGThis study was funded by Ono Pharmaceutical Co. Ltd. and Sysmex Corporation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Immune Checkpoint Inhibitors , Lung Neoplasms , Humans , B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Immunologic Factors/blood , Immunologic Factors/chemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Programmed Cell Death 1 Receptor , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL