Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cancer Discov ; 14(6): 1018-1047, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38581685

ABSTRACT

Understanding the role of the tumor microenvironment (TME) in lung cancer is critical to improving patient outcomes. We identified four histology-independent archetype TMEs in treatment-naïve early-stage lung cancer using imaging mass cytometry in the TRACERx study (n = 81 patients/198 samples/2.3 million cells). In immune-hot adenocarcinomas, spatial niches of T cells and macrophages increased with clonal neoantigen burden, whereas such an increase was observed for niches of plasma and B cells in immune-excluded squamous cell carcinomas (LUSC). Immune-low TMEs were associated with fibroblast barriers to immune infiltration. The fourth archetype, characterized by sparse lymphocytes and high tumor-associated neutrophil (TAN) infiltration, had tumor cells spatially separated from vasculature and exhibited low spatial intratumor heterogeneity. TAN-high LUSC had frequent PIK3CA mutations. TAN-high tumors harbored recently expanded and metastasis-seeding subclones and had a shorter disease-free survival independent of stage. These findings delineate genomic, immune, and physical barriers to immune surveillance and implicate neutrophil-rich TMEs in metastasis. SIGNIFICANCE: This study provides novel insights into the spatial organization of the lung cancer TME in the context of tumor immunogenicity, tumor heterogeneity, and cancer evolution. Pairing the tumor evolutionary history with the spatially resolved TME suggests mechanistic hypotheses for tumor progression and metastasis with implications for patient outcome and treatment. This article is featured in Selected Articles from This Issue, p. 897.


Subject(s)
Lung Neoplasms , Tumor Microenvironment , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Tumor Microenvironment/immunology , T-Lymphocytes/immunology , Myeloid Cells/immunology , Female , Male , Immune Evasion
2.
J Exp Clin Cancer Res ; 43(1): 88, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38515178

ABSTRACT

BACKGROUND: This study explores the repurposing of Auranofin (AF), an anti-rheumatic drug, for treating non-small cell lung cancer (NSCLC) adenocarcinoma and pancreatic ductal adenocarcinoma (PDAC). Drug repurposing in oncology offers a cost-effective and time-efficient approach to developing new cancer therapies. Our research focuses on evaluating AF's selective cytotoxicity against cancer cells, identifying RNAseq-based biomarkers to predict AF response, and finding the most effective co-therapeutic agents for combination with AF. METHODS: Our investigation employed a comprehensive drug screening of AF in combination with eleven anticancer agents in cancerous PDAC and NSCLC patient-derived organoids (n = 7), and non-cancerous pulmonary organoids (n = 2). Additionally, we conducted RNA sequencing to identify potential biomarkers for AF sensitivity and experimented with various drug combinations to optimize AF's therapeutic efficacy. RESULTS: The results revealed that AF demonstrates a preferential cytotoxic effect on NSCLC and PDAC cancer cells at clinically relevant concentrations below 1 µM, sparing normal epithelial cells. We identified Carbonic Anhydrase 12 (CA12) as a significant RNAseq-based biomarker, closely associated with the NF-κB survival signaling pathway, which is crucial in cancer cell response to oxidative stress. Our findings suggest that cancer cells with low CA12 expression are more susceptible to AF treatment. Furthermore, the combination of AF with the AKT inhibitor MK2206 was found to be particularly effective, exhibiting potent and selective cytotoxic synergy, especially in tumor organoid models classified as intermediate responders to AF, without adverse effects on healthy organoids. CONCLUSION: Our research offers valuable insights into the use of AF for treating NSCLC and PDAC. It highlights AF's cancer cell selectivity, establishes CA12 as a predictive biomarker for AF sensitivity, and underscores the enhanced efficacy of AF when combined with MK2206 and other therapeutics. These findings pave the way for further exploration of AF in cancer treatment, particularly in identifying patient populations most likely to benefit from its use and in optimizing combination therapies for improved patient outcomes.


Subject(s)
Adenocarcinoma , Antineoplastic Agents , Carbonic Anhydrases , Carcinoma, Non-Small-Cell Lung , Carcinoma, Pancreatic Ductal , Lung Neoplasms , Pancreatic Neoplasms , Humans , Auranofin/pharmacology , Auranofin/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Proto-Oncogene Proteins c-akt/metabolism , Lung Neoplasms/genetics , Drug Repositioning , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Lung/pathology , Biomarkers , Organoids/metabolism
3.
J Immunother Cancer ; 11(11)2023 11.
Article in English | MEDLINE | ID: mdl-37914385

ABSTRACT

BACKGROUND: Checkpoint inhibitor (CPI) immunotherapies have provided durable clinical responses across a range of solid tumor types for some patients with cancer. Nonetheless, response rates to CPI vary greatly between cancer types. Resolving intratumor transcriptomic changes induced by CPI may improve our understanding of the mechanisms of sensitivity and resistance. METHODS: We assembled a cohort of longitudinal pre-therapy and on-therapy samples from 174 patients treated with CPI across six cancer types by leveraging transcriptomic sequencing data from five studies. RESULTS: Meta-analyses of published RNA markers revealed an on-therapy pattern of immune reinvigoration in patients with breast cancer, which was not discernible pre-therapy, providing biological insight into the impact of CPI on the breast cancer immune microenvironment. We identified 98 breast cancer-specific correlates of CPI response, including 13 genes which are known IO targets, such as toll-like receptors TLR1, TLR4, and TLR8, that could hold potential as combination targets for patients with breast cancer receiving CPI treatment. Furthermore, we demonstrate that a subset of response genes identified in breast cancer are already highly expressed pre-therapy in melanoma, and additionally we establish divergent RNA dynamics between breast cancer and melanoma following CPI treatment, which may suggest distinct immune microenvironments between the two cancer types. CONCLUSIONS: Overall, delineating longitudinal RNA dynamics following CPI therapy sheds light on the mechanisms underlying diverging response trajectories, and identifies putative targets for combination therapy.


Subject(s)
Breast Neoplasms , Melanoma , Humans , Female , Melanoma/drug therapy , Immunotherapy/adverse effects , Combined Modality Therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Tumor Microenvironment/genetics
4.
Front Pharmacol ; 13: 1083017, 2022.
Article in English | MEDLINE | ID: mdl-36712670

ABSTRACT

Organoids have become a prominent model system in pulmonary research. The ability to establish organoid cultures directly from patient tissue has expanded the repertoire of physiologically relevant preclinical model systems. In addition to their derivation from adult lung stem/progenitor cells, lung organoids can be derived from fetal tissue or induced pluripotent stem cells to fill a critical gap in modelling pulmonary development in vitro. Recent years have seen important progress in the characterisation and refinement of organoid culture systems. Here, we address several open questions in the field, including how closely organoids recapitulate the tissue of origin, how well organoids recapitulate patient cohorts, and how well organoids capture diversity within a patient. We advocate deeper characterisation of models using single cell technologies, generation of more diverse organoid biobanks and further standardisation of culture media.

5.
Cancer Discov ; 11(8): 1923-1937, 2021 08.
Article in English | MEDLINE | ID: mdl-33837064

ABSTRACT

Targeted therapies, chemotherapy, and immunotherapy are used to treat patients with mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal cancer. The clinical effectiveness of targeted therapy and chemotherapy is limited by resistance and drug toxicities, and about half of patients receiving immunotherapy have disease that is refractory to immune checkpoint inhibitors. Loss of Werner syndrome ATP-dependent helicase (WRN) is a synthetic lethality in dMMR/MSI-H cells. To inform the development of WRN as a therapeutic target, we performed WRN knockout or knockdown in 60 heterogeneous dMMR colorectal cancer preclinical models, demonstrating that WRN dependency is an almost universal feature and a robust marker for patient selection. Furthermore, models of resistance to clinically relevant targeted therapy, chemotherapy, and immunotherapy retain WRN dependency. These data show the potential of therapeutically targeting WRN in patients with dMMR/MSI-H colorectal cancer and support WRN as a therapeutic option for patients with dMMR/MSI-H cancers refractory to current treatment strategies. SIGNIFICANCE: We found that a large, diverse set of dMMR/MSI-H colorectal cancer preclinical models, including models of treatment-refractory disease, are WRN-dependent. Our results support WRN as a promising synthetic-lethal target in dMMR/MSI-H colorectal cancer tumors as a monotherapy or in combination with targeted agents, chemotherapy, or immunotherapy.This article is highlighted in the In This Issue feature, p. 1861.


Subject(s)
Colorectal Neoplasms/therapy , DNA Mismatch Repair , Werner Syndrome Helicase/genetics , Colorectal Neoplasms/genetics , Drug Therapy , Humans , Immunotherapy , Molecular Targeted Therapy
6.
Front Endocrinol (Lausanne) ; 12: 627819, 2021.
Article in English | MEDLINE | ID: mdl-33776923

ABSTRACT

Gastroenteropancreatic neuroendocrine carcinoma (GEP-NEC) is a poorly understood disease with limited treatment options. A better understanding of this disease would greatly benefit from the availability of representative preclinical models. Here, we present the potential of tumor organoids, three-dimensional cultures of tumor cells, to model GEP-NEC. We established three GEP-NEC organoid lines, originating from the stomach and colon, and characterized them using DNA sequencing and immunohistochemistry. Organoids largely resembled the original tumor in expression of synaptophysin, chromogranin and Ki-67. Models derived from tumors containing both neuroendocrine and non-neuroendocrine components were at risk of overgrowth by non-neuroendocrine tumor cells. Organoids were derived from patients treated with cisplatin and everolimus and for the three patients studied, organoid chemosensitivity paralleled clinical response. We demonstrate the feasibility of establishing NEC organoid lines and their potential applications. Organoid culture has the potential to greatly extend the repertoire of preclinical models for GEP-NEC, supporting drug development for this difficult-to-treat tumor type.


Subject(s)
Intestinal Neoplasms/pathology , Models, Biological , Neuroendocrine Tumors/pathology , Organoids/pathology , Pancreatic Neoplasms/pathology , Stomach Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Cisplatin/therapeutic use , DNA Mismatch Repair/drug effects , Everolimus/pharmacology , Everolimus/therapeutic use , Gene Dosage , Humans , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/genetics , Ki-67 Antigen/metabolism , Mutation/genetics , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/genetics , Organoids/drug effects , Organoids/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Exome Sequencing
7.
Cell Rep ; 31(5): 107588, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32375033

ABSTRACT

Clinical implementation of tumor organoids for personalized medicine requires that pure tumor organoids can be reliably established. Here, we present our experience with organoid cultures from >70 non-small cell lung cancer (NSCLC) samples. We systematically evaluate several methods to identify tumor purity of organoids established from intrapulmonary tumors. Eighty percent of organoids from intrapulmonary lesions have a normal copy number profile, suggesting overgrowth by normal airway organoids (AOs). This is further supported by the failure to detect mutations found in the original tumor in organoids. Histomorphology alone is insufficient to determine tumor purity, but when combined with p63 immunostaining, tumor and normal AOs can be distinguished. Taking into account overgrowth by normal AOs, the establishment rate of pure NSCLC organoids is 17%. Therefore, current methods are insufficient to establish pure NSCLC organoids from intrapulmonary lesions. We discourage their use unless steps are taken to prevent overgrowth by normal AOs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Organoids/pathology , Precision Medicine , Humans , Lung Neoplasms/genetics , Mutation/genetics , Precision Medicine/methods
9.
Nat Med ; 26(4): 566-576, 2020 04.
Article in English | MEDLINE | ID: mdl-32251400

ABSTRACT

PD-1 plus CTLA-4 blockade is highly effective in advanced-stage, mismatch repair (MMR)-deficient (dMMR) colorectal cancers, yet not in MMR-proficient (pMMR) tumors. We postulated a higher efficacy of neoadjuvant immunotherapy in early-stage colon cancers. In the exploratory NICHE study (ClinicalTrials.gov: NCT03026140), patients with dMMR or pMMR tumors received a single dose of ipilimumab and two doses of nivolumab before surgery, the pMMR group with or without celecoxib. The primary objective was safety and feasibility; 40 patients with 21 dMMR and 20 pMMR tumors were treated, and 3 patients received nivolumab monotherapy in the safety run-in. Treatment was well tolerated and all patients underwent radical resections without delays, meeting the primary endpoint. Of the patients who received ipilimumab + nivolumab (20 dMMR and 15 pMMR tumors), 35 were evaluable for efficacy and translational endpoints. Pathological response was observed in 20/20 (100%; 95% exact confidence interval (CI): 86-100%) dMMR tumors, with 19 major pathological responses (MPRs, ≤10% residual viable tumor) and 12 pathological complete responses. In pMMR tumors, 4/15 (27%; 95% exact CI: 8-55%) showed pathological responses, with 3 MPRs and 1 partial response. CD8+PD-1+ T cell infiltration was predictive of response in pMMR tumors. These data indicate that neoadjuvant immunotherapy may have the potential to become the standard of care for a defined group of colon cancer patients when validated in larger studies with at least 3 years of disease-free survival data.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents, Immunological/adverse effects , Colonic Neoplasms/therapy , DNA Mismatch Repair/genetics , Immunotherapy/adverse effects , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/administration & dosage , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Combined Modality Therapy , DNA Mismatch Repair/drug effects , Digestive System Surgical Procedures , Drug Administration Schedule , Feasibility Studies , Female , Humans , Immunotherapy/methods , Ipilimumab/administration & dosage , Ipilimumab/adverse effects , Male , Middle Aged , Neoadjuvant Therapy/adverse effects , Neoadjuvant Therapy/methods , Neoplasm Staging , Nivolumab/administration & dosage , Nivolumab/adverse effects , Treatment Failure
11.
Nat Protoc ; 15(1): 15-39, 2020 01.
Article in English | MEDLINE | ID: mdl-31853056

ABSTRACT

T cells are key players in cancer immunotherapy, but strategies to expand tumor-reactive cells and study their interactions with tumor cells at the level of an individual patient are limited. Here we describe the generation and functional assessment of tumor-reactive T cells based on cocultures of tumor organoids and autologous peripheral blood lymphocytes. The procedure consists of an initial coculture of 2 weeks, in which tumor-reactive T cells are first expanded in the presence of (IFNγ-stimulated) autologous tumor cells. Subsequently, T cells are evaluated for their capacity to carry out effector functions (IFNγ secretion and degranulation) after recognition of tumor cells, and their capacity to kill tumor organoids. This strategy is unique in its use of peripheral blood as a source of tumor-reactive T cells in an antigen-agnostic manner. In 2 weeks, tumor-reactive CD8+ T-cell populations can be obtained from ~33-50% of samples from patients with non-small-cell lung cancer (NSCLC) and microsatellite-instable colorectal cancer (CRC). This enables the establishment of ex vivo test systems for T-cell-based immunotherapy at the level of the individual patient.


Subject(s)
Coculture Techniques/methods , Neoplasms/pathology , Organoids/pathology , T-Lymphocytes/cytology , Humans
12.
Sci Transl Med ; 11(513)2019 10 09.
Article in English | MEDLINE | ID: mdl-31597751

ABSTRACT

There is a clear and unmet clinical need for biomarkers to predict responsiveness to chemotherapy for cancer. We developed an in vitro test based on patient-derived tumor organoids (PDOs) from metastatic lesions to identify nonresponders to standard-of-care chemotherapy in colorectal cancer (CRC). In a prospective clinical study, we show the feasibility of generating and testing PDOs for evaluation of sensitivity to chemotherapy. Our PDO test predicted response of the biopsied lesion in more than 80% of patients treated with irinotecan-based therapies without misclassifying patients who would have benefited from treatment. This correlation was specific to irinotecan-based chemotherapy, however, and the PDOs failed to predict outcome for treatment with 5-fluorouracil plus oxaliplatin. Our data suggest that PDOs could be used to prevent cancer patients from undergoing ineffective irinotecan-based chemotherapy.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Organoids/cytology , Antineoplastic Agents/therapeutic use , Capecitabine/therapeutic use , Colorectal Neoplasms/drug therapy , Female , Fluorouracil/therapeutic use , Humans , Irinotecan/therapeutic use , Oxaliplatin/therapeutic use , Prospective Studies , Treatment Outcome
13.
EMBO J ; 38(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30643021

ABSTRACT

Organoids are self-organizing 3D structures grown from stem cells that recapitulate essential aspects of organ structure and function. Here, we describe a method to establish long-term-expanding human airway organoids from broncho-alveolar resections or lavage material. The pseudostratified airway organoids consist of basal cells, functional multi-ciliated cells, mucus-producing secretory cells, and CC10-secreting club cells. Airway organoids derived from cystic fibrosis (CF) patients allow assessment of CFTR function in an organoid swelling assay. Organoids established from lung cancer resections and metastasis biopsies retain tumor histopathology as well as cancer gene mutations and are amenable to drug screening. Respiratory syncytial virus (RSV) infection recapitulates central disease features, dramatically increases organoid cell motility via the non-structural viral NS2 protein, and preferentially recruits neutrophils upon co-culturing. We conclude that human airway organoids represent versatile models for the in vitro study of hereditary, malignant, and infectious pulmonary disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cystic Fibrosis/pathology , Epithelial Cells/pathology , Organ Culture Techniques/methods , Organoids/pathology , Respiratory Syncytial Virus Infections/pathology , Respiratory System/pathology , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cells, Cultured , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Drug Screening Assays, Antitumor , Epithelial Cells/metabolism , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Organoids/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/isolation & purification , Respiratory System/metabolism , Xenograft Model Antitumor Assays
14.
Nat Med ; 25(1): 89-94, 2019 01.
Article in English | MEDLINE | ID: mdl-30510250

ABSTRACT

Infiltration of human cancers by T cells is generally interpreted as a sign of immune recognition, and there is a growing effort to reactivate dysfunctional T cells at such tumor sites1. However, these efforts only have value if the intratumoral T cell receptor (TCR) repertoire of such cells is intrinsically tumor reactive, and this has not been established in an unbiased manner for most human cancers. To address this issue, we analyzed the intrinsic tumor reactivity of the intratumoral TCR repertoire of CD8+ T cells in ovarian and colorectal cancer-two tumor types for which T cell infiltrates form a positive prognostic marker2,3. Data obtained demonstrate that a capacity to recognize autologous tumor is limited to approximately 10% of intratumoral CD8+ T cells. Furthermore, in two of four patient samples tested, no tumor-reactive TCRs were identified, despite infiltration of their tumors by T cells. These data indicate that the intrinsic capacity of intratumoral T cells to recognize adjacent tumor tissue can be rare and variable, and suggest that clinical efforts to reactivate intratumoral T cells will benefit from approaches that simultaneously increase the quality of the intratumoral TCR repertoire.


Subject(s)
Neoplasms/immunology , Neoplasms/pathology , Receptors, Antigen, T-Cell/metabolism , CD8-Positive T-Lymphocytes/immunology , Humans , Jurkat Cells , Lymphocytes, Tumor-Infiltrating/immunology , Phenotype , Reproducibility of Results
15.
Cell ; 174(6): 1586-1598.e12, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30100188

ABSTRACT

Cancer immunotherapies have shown substantial clinical activity for a subset of patients with epithelial cancers. Still, technological platforms to study cancer T-cell interactions for individual patients and understand determinants of responsiveness are presently lacking. Here, we establish and validate a platform to induce and analyze tumor-specific T cell responses to epithelial cancers in a personalized manner. We demonstrate that co-cultures of autologous tumor organoids and peripheral blood lymphocytes can be used to enrich tumor-reactive T cells from peripheral blood of patients with mismatch repair-deficient colorectal cancer and non-small-cell lung cancer. Furthermore, we demonstrate that these T cells can be used to assess the efficiency of killing of matched tumor organoids. This platform provides an unbiased strategy for the isolation of tumor-reactive T cells and provides a means by which to assess the sensitivity of tumor cells to T cell-mediated attack at the level of the individual patient.


Subject(s)
Leukocytes, Mononuclear/cytology , T-Lymphocytes/immunology , Aged , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Culture Techniques , Coculture Techniques , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , In Vitro Techniques , Interferon-gamma/pharmacology , Leukocytes, Mononuclear/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphocyte Activation/drug effects , Male , Middle Aged , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Tumor Cells, Cultured
16.
Cell Chem Biol ; 24(9): 1092-1100, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28757181

ABSTRACT

Tumor organoids are 3D cultures of cancer cells that can be derived on an individual patient basis with a high success rate. This creates opportunities to build large biobanks with relevant patient material that can be used to perform drug screens and facilitate drug development. The high take rate will also allow side-by-side comparison to evaluate the translational potential of this model system to the patient. These tumors-in-a-dish can be established for a variety of tumor types including colorectal, pancreas, stomach, prostate, and breast cancers. In this review, we highlight what is currently known about tumor organoid culture, the advantages and challenges of the model system, compare it with other pre-clinical cancer models, and evaluate its value for drug development.


Subject(s)
Drug Discovery , Models, Biological , Neoplasms/therapy , Animals , Antineoplastic Agents/therapeutic use , CRISPR-Cas Systems , Cell Culture Techniques , Cystic Fibrosis/therapy , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Xenograft Model Antitumor Assays
17.
JAMA Oncol ; 2(11): 1490-1495, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27491050

ABSTRACT

IMPORTANCE: Checkpoint blockade therapy targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and the programmed cell death protein 1 pathways (PD-1/PD-L1) have achieved success in treating a number of malignancies. However, only a subset of patients responds to these therapies, and optimization of patient selection for treatment is imperative to avoid adverse effects without clinical benefit and keep costs manageable. OBSERVATIONS: The past few years have witnessed checkpoint inhibition becoming a first-line treatment option with US Food and Drug Administration approvals for various tumor types. Genomic analyses (whole genome, exome, and transcriptome) have been instrumental in identifying a genetic profile associated with sensitivity to checkpoint inhibitors. Therapy outcome is determined at various levels: (1) the degree of tumor "foreignness," as reflected by mutational burden and expression of viral genes, (2) the composition and activity of a preexisting immune infiltrate, and (3) mechanisms of tumor escape from immune surveillance. In addition, there are opportunities for genomic analyses of genetic polymorphisms and the gut microbiome that may be associated with clinical response to therapy. CONCLUSIONS AND RELEVANCE: Genomics provides powerful tools for the identification of biomarkers for response to immune checkpoint blockade, given their potential to analyze multiple parameters simultaneously in an unbiased manner. This offers the opportunity for genomics- and transcriptomics-based selection of patients for rationally designed therapy with immune checkpoint inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Immunologic Factors/therapeutic use , Neoplasms/drug therapy , Transcriptome , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Genomics , Humans , Immunologic Factors/pharmacology , Neoplasms/immunology , Neoplasms/metabolism , Patient Selection , Treatment Outcome
18.
Proc Natl Acad Sci U S A ; 112(43): 13308-11, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26460009

ABSTRACT

Tumor organoids are 3D cultures of cancer cells. They can be derived from the tumor of each individual patient, thereby providing an attractive ex vivo assay to tailor treatment. Using patient-derived tumor organoids for this purpose requires that organoids derived from biopsies maintain the genetic diversity of the in vivo tumor. In this study tumor biopsies were obtained from 14 patients with metastatic colorectal cancer (i) to test the feasibility of organoid culture from metastatic biopsy specimens and (ii) to compare the genetic diversity of patient-derived tumor organoids and the original tumor biopsy. Genetic analysis was performed using SOLiD sequencing for 1,977 cancer-relevant genes. Copy number profiles were generated from sequencing data using CopywriteR. Here we demonstrate that organoid cultures can be established from tumor biopsies of patients with metastatic colorectal cancer with a success rate of 71%. Genetic analysis showed that organoids reflect the metastasis from which they were derived. Ninety percent of somatic mutations were shared between organoids and biopsies from the same patient, and the DNA copy number profiles of organoids and the corresponding original tumor show a correlation of 0.89. Most importantly, none of the mutations that were found exclusively in either the tumor or organoid culture are in driver genes or genes amenable for drug targeting. These findings support further exploration of patient-derived organoids as an ex vivo platform to personalize anticancer treatment.


Subject(s)
Cell Culture Techniques/methods , Colorectal Neoplasms/genetics , Genetic Variation/genetics , Neoplasm Metastasis/genetics , Organoids/cytology , Organoids/growth & development , Antineoplastic Protocols/standards , Base Sequence , Colorectal Neoplasms/drug therapy , Genes, Neoplasm/genetics , Humans , Molecular Sequence Data , Organoids/chemistry , Precision Medicine/methods , Sequence Analysis, DNA
20.
J Neurol Neurosurg Psychiatry ; 84(4): 379-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22942215

ABSTRACT

In the majority of the normal population, the left hemisphere is dominant for language. In epilepsy, a higher proportion of 'atypical' language representation is encountered. This can follow one of three patterns: (1) altered interhemispheric representation, where the spectrum of lateralisation is shifted to the right; (2) interhemispheric dissociation of linguistic subfunctions; or (3) intrahemispheric changes in representation. Knowledge of these patterns is essential for avoiding postoperative language deficits in epilepsy patients undergoing surgery. Several predictors of atypical language representation exist. It is more prevalent in left-handed individuals. Lesions in rough proximity to classical language areas are more associated with atypical language, although in some cases, remote lesions, such as in the hippocampus, can also lead to altered language representation. The more disruptive the lesion, the more likely atypical language is to be found. Widespread and frequent interictal epileptiform discharges are also associated with atypical language. Atypical language representation is more likely to be present when injury or epilepsy onset occurred at a young age. Thus, a subgroup of patients can be defined in whom atypical language representation is more likely to be found.


Subject(s)
Epilepsy/physiopathology , Functional Laterality/physiology , Language , Adolescent , Adult , Age of Onset , Aged , Child , Child, Preschool , Dominance, Cerebral/physiology , Epilepsy/epidemiology , Epilepsy/pathology , Humans , Middle Aged , Reference Values , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...