Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38975874

ABSTRACT

KRAS inhibitors demonstrate clinical efficacy in pancreatic ductal adenocarcinoma (PDAC); however, resistance is common. Among patients with KRASG12C-mutant PDAC treated with adagrasib or sotorasib, mutations in PIK3CA and KRAS, and amplifications of KRASG12C, MYC, MET, EGFR, and CDK6 emerged at acquired resistance. In PDAC cell lines and organoid models treated with the KRASG12D inhibitor MRTX1133, epithelial-to-mesenchymal transition and PI3K-AKT-mTOR signaling associate with resistance to therapy. MRTX1133 treatment of the KrasLSL-G12D/+;Trp53LSL-R172H/+;p48-Cre (KPC) mouse model yielded deep tumor regressions, but drug resistance ultimately emerged, accompanied by amplifications of Kras, Yap1, Myc, and Cdk6/Abcb1a/b, and co-evolution of drug-resistant transcriptional programs. Moreover, in KPC and PDX models, mesenchymal and basal-like cell states displayed increased response to KRAS inhibition compared to the classical state. Combination treatment with KRASG12D inhibition and chemotherapy significantly improved tumor control in PDAC mouse models. Collectively, these data elucidate co-evolving resistance mechanisms to KRAS inhibition and support multiple combination therapy strategies.

2.
Nat Cancer ; 4(1): 62-80, 2023 01.
Article in English | MEDLINE | ID: mdl-36585453

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is considered non-immunogenic, with trials showing its recalcitrance to PD1 and CTLA4 immune checkpoint therapies (ICTs). Here, we sought to systematically characterize the mechanisms underlying de novo ICT resistance and to identify effective therapeutic options for PDAC. We report that agonist 41BB and antagonist LAG3 ICT alone and in combination, increased survival and antitumor immunity, characterized by modulating T cell subsets with antitumor activity, increased T cell clonality and diversification, decreased immunosuppressive myeloid cells and increased antigen presentation/decreased immunosuppressive capability of myeloid cells. Translational analyses confirmed the expression of 41BB and LAG3 in human PDAC. Since single and dual ICTs were not curative, T cell-activating ICTs were combined with a CXCR1/2 inhibitor targeting immunosuppressive myeloid cells. Triple therapy resulted in durable complete responses. Given similar profiles in human PDAC and the availability of these agents for clinical testing, our findings provide a testable hypothesis for this lethal disease.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Carcinoma, Pancreatic Ductal/drug therapy , Myeloid Cells/pathology , Pancreatic Neoplasms/drug therapy , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Receptors, Interleukin-8A/immunology , Pancreatic Neoplasms
3.
Cancer Discov ; 12(10): 2330-2349, 2022 10 05.
Article in English | MEDLINE | ID: mdl-35849783

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has few effective treatments. Immunotherapy, an attractive alternative strategy, remains challenging with the lack of knowledge on the tumor-infiltrating lymphocyte (TIL) landscape in PDAC. To generate a reference of T-cell subpopulations, we profiled 80,000 T cells from 57 PDAC samples, 22 uninvolved/normal samples, and cultured TIL using single-cell transcriptomic and T-cell receptor analysis. These data revealed 20 cell states and heterogeneous distributions of TIL populations. The CD8+ TIL contained a putative transitional GZMK+ population based on T-cell receptor clonotype sharing, and cell-state trajectory analysis showed similarity to a GZMB+PRF1+ cytotoxic and a CXCL13+ dysfunctional population. Statistical analysis suggested that certain TIL states, such as dysfunctional and inhibitory populations, often occurred together. Finally, analysis of cultured TIL revealed that high-frequency clones from effector populations were preferentially expanded. These data provide a framework for understanding the PDAC TIL landscape for future TIL use in immunotherapy for PDAC. SIGNIFICANCE: To improve the efficacy of immunotherapy in PDAC, there is a great need to understand the PDAC TIL landscape. This study represents a reference of PDAC TIL subpopulations and their relationships and provides a foundation upon which to base future immunotherapeutic efforts. This article is highlighted in the In This Issue feature, p. 2221.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/therapy , Humans , Lymphocytes, Tumor-Infiltrating , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Receptors, Antigen, T-Cell , Pancreatic Neoplasms
4.
Nature ; 592(7853): 302-308, 2021 04.
Article in English | MEDLINE | ID: mdl-33762732

ABSTRACT

Our knowledge of copy number evolution during the expansion of primary breast tumours is limited1,2. Here, to investigate this process, we developed a single-cell, single-molecule DNA-sequencing method and performed copy number analysis of 16,178 single cells from 8 human triple-negative breast cancers and 4 cell lines. The results show that breast tumours and cell lines comprise a large milieu of subclones (7-22) that are organized into a few (3-5) major superclones. Evolutionary analysis suggests that after clonal TP53 mutations, multiple loss-of-heterozygosity events and genome doubling, there was a period of transient genomic instability followed by ongoing copy number evolution during the primary tumour expansion. By subcloning single daughter cells in culture, we show that tumour cells rediversify their genomes and do not retain isogenic properties. These data show that triple-negative breast cancers continue to evolve chromosome aberrations and maintain a reservoir of subclonal diversity during primary tumour growth.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Clone Cells/metabolism , Clone Cells/pathology , Evolution, Molecular , Base Sequence , Cell Line, Tumor , Cell Lineage , Chromosome Aberrations , DNA Copy Number Variations/genetics , DNA Mutational Analysis , Genomic Instability/genetics , Humans , Loss of Heterozygosity/genetics , Models, Genetic , Mutation Rate , Single Molecule Imaging , Single-Cell Analysis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
5.
Nat Biotechnol ; 39(5): 599-608, 2021 05.
Article in English | MEDLINE | ID: mdl-33462507

ABSTRACT

Single-cell transcriptomic analysis is widely used to study human tumors. However, it remains challenging to distinguish normal cell types in the tumor microenvironment from malignant cells and to resolve clonal substructure within the tumor. To address these challenges, we developed an integrative Bayesian segmentation approach called copy number karyotyping of aneuploid tumors (CopyKAT) to estimate genomic copy number profiles at an average genomic resolution of 5 Mb from read depth in high-throughput single-cell RNA sequencing (scRNA-seq) data. We applied CopyKAT to analyze 46,501 single cells from 21 tumors, including triple-negative breast cancer, pancreatic ductal adenocarcinoma, anaplastic thyroid cancer, invasive ductal carcinoma and glioblastoma, to accurately (98%) distinguish cancer cells from normal cell types. In three breast tumors, CopyKAT resolved clonal subpopulations that differed in the expression of cancer genes, such as KRAS, and signatures, including epithelial-to-mesenchymal transition, DNA repair, apoptosis and hypoxia. These data show that CopyKAT can aid in the analysis of scRNA-seq data in a variety of solid human tumors.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Pancreatic Ductal/genetics , Clonal Evolution , DNA Copy Number Variations/genetics , Transcriptome/genetics , Breast Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic , Genomics/trends , High-Throughput Nucleotide Sequencing , Humans , Mutation/genetics , Single-Cell Analysis , Tumor Microenvironment/genetics
6.
J Exp Med ; 217(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32491160

ABSTRACT

CD8+ T cells are master effectors of antitumor immunity, and their presence at tumor sites correlates with favorable outcomes. However, metabolic constraints imposed by the tumor microenvironment (TME) can dampen their ability to control tumor progression. We describe lipid accumulation in the TME areas of pancreatic ductal adenocarcinoma (PDA) populated by CD8+ T cells infiltrating both murine and human tumors. In this lipid-rich but otherwise nutrient-poor TME, access to using lipid metabolism becomes particularly valuable for sustaining cell functions. Here, we found that intrapancreatic CD8+ T cells progressively accumulate specific long-chain fatty acids (LCFAs), which, rather than provide a fuel source, impair their mitochondrial function and trigger major transcriptional reprogramming of pathways involved in lipid metabolism, with the subsequent reduction of fatty acid catabolism. In particular, intrapancreatic CD8+ T cells specifically exhibit down-regulation of the very-long-chain acyl-CoA dehydrogenase (VLCAD) enzyme, which exacerbates accumulation of LCFAs and very-long-chain fatty acids (VLCFAs) that mediate lipotoxicity. Metabolic reprogramming of tumor-specific T cells through enforced expression of ACADVL enabled enhanced intratumoral T cell survival and persistence in an engineered mouse model of PDA, overcoming one of the major hurdles to immunotherapy for PDA.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Fatty Acids/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Tumor Microenvironment , Acyl-CoA Dehydrogenase, Long-Chain/biosynthesis , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Animals , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Down-Regulation , Fatty Acids/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Mutant Strains , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology
7.
Nat Commun ; 11(1): 1839, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32296058

ABSTRACT

Complex tumor microenvironmental (TME) features influence the outcome of cancer immunotherapy (IO). Here we perform immunogenomic analyses on 67 intratumor sub-regions of a PD-1 inhibitor-resistant melanoma tumor and 2 additional metastases arising over 8 years, to characterize TME interactions. We identify spatially distinct evolution of copy number alterations influencing local immune composition. Sub-regions with chromosome 7 gain display a relative lack of leukocyte infiltrate but evidence of neutrophil activation, recapitulated in The Cancer Genome Atlas (TCGA) samples, and associated with lack of response to IO across three clinical cohorts. Whether neutrophil activation represents cause or consequence of local tumor necrosis requires further study. Analyses of T-cell clonotypes reveal the presence of recurrent priming events manifesting in a dominant T-cell clonotype over many years. Our findings highlight the links between marked levels of genomic and immune heterogeneity within the physical space of a tumor, with implications for biomarker evaluation and immunotherapy response.


Subject(s)
Genomics/methods , Melanoma/metabolism , Biomarkers, Tumor/genetics , DNA Copy Number Variations/genetics , Humans , Melanoma/genetics , Mutation/genetics , Neutrophil Activation/genetics , Neutrophil Activation/physiology , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology
8.
Trends Mol Med ; 24(10): 821-822, 2018 10.
Article in English | MEDLINE | ID: mdl-30077643

ABSTRACT

The role of tissue-resident memory T (TRM) cells in breast cancer progression has been difficult to study. Savas et al. [1] (Nat. Med. 2018;24:986-993) used single-cell RNA sequencing to identify TRM cells in triple-negative breast cancer patients and demonstrated their prognostic value for predicting survival.


Subject(s)
Breast Neoplasms , Immunologic Memory , CD8-Positive T-Lymphocytes , Disease Progression , Humans , Prognosis
9.
Cell ; 172(1-2): 205-217.e12, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29307488

ABSTRACT

Ductal carcinoma in situ (DCIS) is an early-stage breast cancer that infrequently progresses to invasive ductal carcinoma (IDC). Genomic evolution has been difficult to delineate during invasion due to intratumor heterogeneity and the low number of tumor cells in the ducts. To overcome these challenges, we developed Topographic Single Cell Sequencing (TSCS) to measure genomic copy number profiles of single tumor cells while preserving their spatial context in tissue sections. We applied TSCS to 1,293 single cells from 10 synchronous patients with both DCIS and IDC regions in addition to exome sequencing. Our data reveal a direct genomic lineage between in situ and invasive tumor subpopulations and further show that most mutations and copy number aberrations evolved within the ducts prior to invasion. These results support a multiclonal invasion model, in which one or more clones escape the ducts and migrate into the adjacent tissues to establish the invasive carcinomas.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Clonal Evolution , Adult , Aged , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Cell Movement , Exome , Female , Humans , Middle Aged , Mutation , Neoplasm Invasiveness , Sequence Analysis, DNA , Single-Cell Analysis
10.
J Mol Diagn ; 18(1): 100-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26631873

ABSTRACT

BRAF(V600E) is a common mutation in papillary thyroid carcinoma (PTC) correlated with aggressive features. Our objective was to assess the feasibility and accuracy of a novel RNA-based blood assay to identify individuals with a high-risk tumor mutation in patients with PTC. Patients with benign or malignant thyroid disorders were included between September 2013 and July 2014 before either thyroidectomy (n = 62) or treatment of recurrent or metastatic PTC (n = 8). RNA was isolated from peripheral blood lymphocytes and reverse transcribed and followed by two rounds of nested PCR amplification with a restriction digest specific for wild-type BRAF. BRAF(V600E) levels were quantified with standardization curves. Circulating BRAF(V600E) levels were compared with BRAF mutation status from surgical pathologic DNA-based tissue assays. Testing characteristics and receiving-operator curve using tissue results as the gold standard were assessed. Matched blood and tissue assays for BRAF(V600E) were performed on 70 patients with PTC (stages I to IV, n = 48) or other (n = 22) thyroid tumors. Sixty-three percent of PTC patients tested positive for BRAF(V600E) with conventional tissue assays on surgical specimens. The correlation between the RNA-based blood assay and tissue BRAF status was 0.71. PTC patients harbor detectable BRAF(V600E) circulating tumor cells. This blood assay is feasible and has potential as a biomarker for prognosis, surveillance, clinical decision making, and assessment of treatment response to BRAF-targeted therapies.


Subject(s)
Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma/blood , Carcinoma/genetics , Proto-Oncogene Proteins B-raf/blood , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/blood , Thyroid Neoplasms/genetics , Carcinoma/surgery , Carcinoma, Papillary , Female , Humans , Lymphocytes/cytology , Male , Middle Aged , Mutation/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Cancer, Papillary , Thyroid Neoplasms/surgery , Thyroidectomy
11.
Mol Cancer Ther ; 13(12): 3210-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25319388

ABSTRACT

BRAF inhibitors (BRAFi) have led to clinical benefit in patients with melanoma. The development of a blood-based assay to detect and quantify BRAF levels in these patients has diagnostic, prognostic, and predictive capabilities that could guide treatment decisions. Blood BRAF(V600E) detection and quantification were performed on samples from 128 patients with stage II (19), III (67), and IV (42) melanoma. Tissue BRAF analysis was performed in all patients with stage IV disease and in selected patients with stage II and III disease. Clinical outcomes were correlated to initial BRAF levels as well as BRAF level dynamics. Serial analysis was performed on 17 stage IV melanoma patients treated with BRAFi and compared with tumor measurements by RECIST. The assay was highly sensitive (96%) and specific (95%) in the stage IV setting, using a blood level of 4.8 pg as "positive." BRAF levels typically decreased following BRAFi. A subset of these patients (5) had an increase in BRAF(V600E) values 42 to 112 days before clinical or radiographic disease progression (PD). From 86 patients with resected, stage II or III melanoma, 39 had evidence of disease relapse (45.3%). Furthermore, BRAF mutation in the blood after surgical resection in these patients was not associated with a difference in relapse risk, although tissue BRAF status was only available for a subset of patients. In summary, we have developed a highly sensitive and specific, blood-based assay to detect BRAF(V600) mutation in patients with melanoma.


Subject(s)
DNA Mutational Analysis/methods , Melanoma/diagnosis , Melanoma/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Amino Acid Substitution , Cell Line, Tumor , Codon , DNA Mutational Analysis/standards , Genotype , Humans , Leukocytes, Mononuclear , Melanoma/drug therapy , Molecular Targeted Therapy , Neoplasm Staging , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Reproducibility of Results , Sensitivity and Specificity
12.
PLoS One ; 9(7): e101286, 2014.
Article in English | MEDLINE | ID: mdl-24983357

ABSTRACT

While response rates to BRAF inhibitiors (BRAFi) are high, disease progression emerges quickly. One strategy to delay the onset of resistance is to target anti-apoptotic proteins such as BCL-2, known to be associated with a poor prognosis. We analyzed BCL-2 family member expression levels of 34 samples from 17 patients collected before and 10 to 14 days after treatment initiation with either vemurafenib or dabrafenib/trametinib combination. The observed changes in mRNA and protein levels with BRAFi treatment led us to hypothesize that combining BRAFi with a BCL-2 inhibitor (the BH3-mimetic navitoclax) would improve outcome. We tested this hypothesis in cell lines and in mice. Pretreatment mRNA levels of BCL-2 negatively correlated with maximal tumor regression. Early increases in mRNA levels were seen in BIM, BCL-XL, BID and BCL2-W, as were decreases in MCL-1 and BCL2A. No significant changes were observed with BCL-2. Using reverse phase protein array (RPPA), significant increases in protein levels were found in BIM and BID. No changes in mRNA or protein correlated with response. Concurrent BRAF (PLX4720) and BCL2 (navitoclax) inhibition synergistically reduced viability in BRAF mutant cell lines and correlated with down-modulation of MCL-1 and BIM induction after PLX4720 treatment. In xenograft models, navitoclax enhanced the efficacy of PLX4720. The combination of a selective BRAF inhibitor with a BH3-mimetic promises to be an important therapeutic strategy capable of enhancing the clinical efficacy of BRAF inhibition in many patients that might otherwise succumb quickly to de novo resistance. Trial registrations: ClinicalTrials.gov NCT01006980; ClinicalTrials.gov NCT01107418; ClinicalTrials.gov NCT01264380; ClinicalTrials.gov NCT01248936; ClinicalTrials.gov NCT00949702; ClinicalTrials.gov NCT01072175.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Adult , Aged , Aniline Compounds/administration & dosage , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Imidazoles/administration & dosage , Indoles/administration & dosage , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Middle Aged , Mutation, Missense , Neoplasm Metastasis , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Sulfonamides/administration & dosage , Vemurafenib
SELECTION OF CITATIONS
SEARCH DETAIL
...