Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Cancer ; 2(1): 66-82, 2021 01.
Article in English | MEDLINE | ID: mdl-33738458

ABSTRACT

Despite objective responses to PARP inhibition and improvements in progression-free survival compared to standard chemotherapy in patients with BRCA-associated triple-negative breast cancer (TNBC), benefits are transitory. Using high dimensional single-cell profiling of human TNBC, here we demonstrate that macrophages are the predominant infiltrating immune cell type in BRCA-associated TNBC. Through multi-omics profiling we show that PARP inhibitors enhance both anti- and pro-tumor features of macrophages through glucose and lipid metabolic reprogramming driven by the sterol regulatory element-binding protein 1 (SREBP-1) pathway. Combined PARP inhibitor therapy with CSF-1R blocking antibodies significantly enhanced innate and adaptive anti-tumor immunity and extends survival in BRCA-deficient tumors in vivo and is mediated by CD8+ T-cells. Collectively, our results uncover macrophage-mediated immune suppression as a liability of PARP inhibitor treatment and demonstrate combined PARP inhibition and macrophage targeting therapy induces a durable reprogramming of the tumor microenvironment, thus constituting a promising therapeutic strategy for TNBC.


Subject(s)
Poly(ADP-ribose) Polymerase Inhibitors , Triple Negative Breast Neoplasms , BRCA1 Protein/genetics , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Humans , Macrophages , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Tumor Microenvironment
2.
Nat Genet ; 53(3): 342-353, 2021 03.
Article in English | MEDLINE | ID: mdl-33558758

ABSTRACT

Technological and computational advances in genomics and interactomics have made it possible to identify how disease mutations perturb protein-protein interaction (PPI) networks within human cells. Here, we show that disease-associated germline variants are significantly enriched in sequences encoding PPI interfaces compared to variants identified in healthy participants from the projects 1000 Genomes and ExAC. Somatic missense mutations are also significantly enriched in PPI interfaces compared to noninterfaces in 10,861 tumor exomes. We computationally identified 470 putative oncoPPIs in a pan-cancer analysis and demonstrate that oncoPPIs are highly correlated with patient survival and drug resistance/sensitivity. We experimentally validate the network effects of 13 oncoPPIs using a systematic binary interaction assay, and also demonstrate the functional consequences of two of these on tumor cell growth. In summary, this human interactome network framework provides a powerful tool for prioritization of alleles with PPI-perturbing mutations to inform pathobiological mechanism- and genotype-based therapeutic discovery.


Subject(s)
Computational Biology/methods , Mutation , Neoplasms/genetics , Protein Interaction Maps/genetics , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Arginine/genetics , Arginine/metabolism , Disease/genetics , Genome, Human , Histones/genetics , Histones/metabolism , Humans , Pharmacogenomic Testing , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Reproducibility of Results , Serine/genetics , Serine/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
3.
Hum Mol Genet ; 29(R2): R177-R185, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32601683

ABSTRACT

An individual's inherited genetic makeup and acquired genomic variants may account for a significant portion of observable variability in therapy efficacy and toxicity. Pharmacogenomics (PGx) is the concept that treatments can be modified to account for these differences to increase chances of therapeutic efficacy while minimizing risk of adverse effects. This is particularly applicable to oncology in which treatment may be multimodal. Each tumor type has a unique genomic signature that lends to inclusion of targeted therapy but may be associated with cumulative toxicity, such as cardiotoxicity, and can impact quality of life. A greater understanding of therapeutic agents impacted by PGx and subsequent implementation has the potential to improve outcomes and reduce risk of drug-induced adverse effects.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxicity/pathology , Cardiovascular Diseases/pathology , Molecular Targeted Therapy/adverse effects , Neoplasms/drug therapy , Pharmacogenetics , Pharmacogenomic Variants , Cardiotoxicity/etiology , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Precision Medicine , Risk Assessment
4.
Hum Mol Genet ; 29(R2): R186-R196, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32620943

ABSTRACT

Immune checkpoint blockade (ICB) has become a standard of care in a subset of solid tumors. Although cancer survivorship has extended, rates of durable response of ICB remain poor; furthermore, cardiac adverse effects are emerging, which impact several mechanical aspects of the heart. Cardio-oncology programs implement a clinical assessment to curtail cardiovascular disease progression but are limited to the current clinical parameters used in cardiology. Pharmacogenomics provides the potential to unveil heritable and somatic genetic variations for guiding precision immunotherapy treatment to reduce the risk of immune-related cardiotoxicity. A better understanding of pharmacogenomics will optimize the current treatment selection and dosing of immunotherapy. Here, we summarize the recent pharmacogenomics studies in immunotherapy responsiveness and its related cardiotoxicity and highlight how patient genetics and epigenetics can facilitate researchers and clinicians in designing new approaches for precision immunotherapy. We highlight and discuss how single-cell technologies, human-induced pluripotent stem cells and systems pharmacogenomics accelerate future studies of precision cardio-oncology.


Subject(s)
Cardiotoxicity/pathology , Cardiovascular Diseases/pathology , Immunotherapy/adverse effects , Molecular Targeted Therapy/adverse effects , Neoplasms/drug therapy , Pharmacogenetics , Pharmacogenomic Variants , Cardiotoxicity/etiology , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/genetics , Cardiovascular Diseases/immunology , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Precision Medicine , Risk Assessment
5.
Genome Res ; 30(1): 62-71, 2020 01.
Article in English | MEDLINE | ID: mdl-31871067

ABSTRACT

Missense variant interpretation is challenging. Essential regions for protein function are conserved among gene-family members, and genetic variants within these regions are potentially more likely to confer risk to disease. Here, we generated 2871 gene-family protein sequence alignments involving 9990 genes and performed missense variant burden analyses to identify novel essential protein regions. We mapped 2,219,811 variants from the general population into these alignments and compared their distribution with 76,153 missense variants from patients. With this gene-family approach, we identified 465 regions enriched for patient variants spanning 41,463 amino acids in 1252 genes. As a comparison, by testing the same genes individually, we identified fewer patient variant enriched regions, involving only 2639 amino acids and 215 genes. Next, we selected de novo variants from 6753 patients with neurodevelopmental disorders and 1911 unaffected siblings and observed an 8.33-fold enrichment of patient variants in our identified regions (95% C.I. = 3.90-Inf, P-value = 2.72 × 10-11). By using the complete ClinVar variant set, we found that missense variants inside the identified regions are 106-fold more likely to be classified as pathogenic in comparison to benign classification (OR = 106.15, 95% C.I = 70.66-Inf, P-value < 2.2 × 10-16). All pathogenic variant enriched regions (PERs) identified are available online through "PER viewer," a user-friendly online platform for interactive data mining, visualization, and download. In summary, our gene-family burden analysis approach identified novel PERs in protein sequences. This annotation can empower variant interpretation.


Subject(s)
Chromosome Mapping , Genetic Predisposition to Disease , Genetic Variation , Multigene Family , Alleles , Amino Acid Sequence , Amino Acid Substitution , Computational Biology/methods , Female , Genome-Wide Association Study , Humans , Male , Mutation, Missense , Software , User-Computer Interface
6.
Cancer Discov ; 9(6): 722-737, 2019 06.
Article in English | MEDLINE | ID: mdl-31015319

ABSTRACT

Combinatorial clinical trials of PARP inhibitors with immunotherapies are ongoing, yet the immunomodulatory effects of PARP inhibition have been incompletely studied. Here, we sought to dissect the mechanisms underlying PARP inhibitor-induced changes in the tumor microenvironment of BRCA1-deficient triple-negative breast cancer (TNBC). We demonstrate that the PARP inhibitor olaparib induces CD8+ T-cell infiltration and activation in vivo, and that CD8+ T-cell depletion severely compromises antitumor efficacy. Olaparib-induced T-cell recruitment is mediated through activation of the cGAS/STING pathway in tumor cells with paracrine activation of dendritic cells and is more pronounced in HR-deficient compared with HR-proficient TNBC cells and in vivo models. CRISPR-mediated knockout of STING in cancer cells prevents proinflammatory signaling and is sufficient to abolish olaparib-induced T-cell infiltration in vivo. These findings elucidate an additional mechanism of action of PARP inhibitors and provide a rationale for combining PARP inhibition with immunotherapies for the treatment of TNBC. SIGNIFICANCE: This work demonstrates cross-talk between PARP inhibition and the tumor microenvironment related to STING/TBK1/IRF3 pathway activation in cancer cells that governs CD8+ T-cell recruitment and antitumor efficacy. The data provide insight into the mechanism of action of PARP inhibitors in BRCA-associated breast cancer.This article is highlighted in the In This Issue feature, p. 681.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Membrane Proteins/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/etiology , Triple Negative Breast Neoplasms/metabolism , BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , Biomarkers , CD8-Positive T-Lymphocytes/drug effects , Female , Humans , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
7.
Cancer Discov ; 7(8): 852-867, 2017 08.
Article in English | MEDLINE | ID: mdl-28408401

ABSTRACT

Effective therapies for non-small cell lung cancer (NSCLC) remain challenging despite an increasingly comprehensive understanding of somatically altered oncogenic pathways. It is now clear that therapeutic agents with potential to impact the tumor immune microenvironment potentiate immune-orchestrated therapeutic benefit. Herein, we evaluated the immunoregulatory properties of histone deacetylase (HDAC) and bromodomain inhibitors, two classes of drugs that modulate the epigenome, with a focus on key cell subsets that are engaged in an immune response. By evaluating human peripheral blood and NSCLC tumors, we show that the selective HDAC6 inhibitor ricolinostat promotes phenotypic changes that support enhanced T-cell activation and improved function of antigen-presenting cells. The bromodomain inhibitor JQ1 attenuated CD4+FOXP3+ T regulatory cell suppressive function and synergized with ricolinostat to facilitate immune-mediated tumor growth arrest, leading to prolonged survival of mice with lung adenocarcinomas. Collectively, our findings highlight the immunomodulatory effects of two epigenetic modifiers that, together, promote T cell-mediated antitumor immunity and demonstrate their therapeutic potential for treatment of NSCLC.Significance: Selective inhibition of HDACs and bromodomain proteins modulates tumor-associated immune cells in a manner that favors improved T-cell function and reduced inhibitory cellular mechanisms. These effects facilitated robust antitumor responses in tumor-bearing mice, demonstrating the therapeutic potential of combining these epigenetic modulators for the treatment of NSCLC. Cancer Discov; 7(8); 852-67. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 783.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Pyrimidines/administration & dosage , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Humans , Hydroxamic Acids/adverse effects , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Middle Aged , Pyrimidines/adverse effects , Xenograft Model Antitumor Assays
8.
Nature ; 543(7645): 428-432, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28273064

ABSTRACT

Although the main focus of immuno-oncology has been manipulating the adaptive immune system, harnessing both the innate and adaptive arms of the immune system might produce superior tumour reduction and elimination. Tumour-associated macrophages often have net pro-tumour effects, but their embedded location and their untapped potential provide impetus to discover strategies to turn them against tumours. Strategies that deplete (anti-CSF-1 antibodies and CSF-1R inhibition) or stimulate (agonistic anti-CD40 or inhibitory anti-CD47 antibodies) tumour-associated macrophages have had some success. We hypothesized that pharmacologic modulation of macrophage phenotype could produce an anti-tumour effect. We previously reported that a first-in-class selective class IIa histone deacetylase (HDAC) inhibitor, TMP195, influenced human monocyte responses to the colony-stimulating factors CSF-1 and CSF-2 in vitro. Here, we utilize a macrophage-dependent autochthonous mouse model of breast cancer to demonstrate that in vivo TMP195 treatment alters the tumour microenvironment and reduces tumour burden and pulmonary metastases by modulating macrophage phenotypes. TMP195 induces the recruitment and differentiation of highly phagocytic and stimulatory macrophages within tumours. Furthermore, combining TMP195 with chemotherapy regimens or T-cell checkpoint blockade in this model significantly enhances the durability of tumour reduction. These data introduce class IIa HDAC inhibition as a means to harness the anti-tumour potential of macrophages to enhance cancer therapy.


Subject(s)
Breast Neoplasms/drug therapy , Histone Deacetylase Inhibitors/classification , Histone Deacetylase Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Macrophages/drug effects , Macrophages/immunology , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Breast Neoplasms/blood supply , Breast Neoplasms/enzymology , Breast Neoplasms/immunology , Cell Differentiation/drug effects , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Histone Deacetylase Inhibitors/therapeutic use , Humans , Lung Neoplasms/immunology , Macrophage Activation/drug effects , Macrophages/cytology , Mice , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Phagocytosis/drug effects , Tumor Burden/drug effects , Tumor Burden/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...