scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Personalized In Vitro and In Vivo Cancer Models to Guide Precision Medicine

TL;DR: A robust precision cancer care platform that integrates whole-exome sequencing with a living biobank that enables high-throughput drug screens on patient-derived tumor organoids is described, which promotes the discovery of novel therapeutic approaches that can be assessed in clinical trials and provides personalized therapeutic options where standard clinical options have been exhausted.
Abstract: Precision medicine is an approach that takes into account the influence of individuals' genes, environment, and lifestyle exposures to tailor interventions. Here, we describe the development of a robust precision cancer care platform that integrates whole-exome sequencing with a living biobank that enables high-throughput drug screens on patient-derived tumor organoids. To date, 56 tumor-derived organoid cultures and 19 patient-derived xenograft (PDX) models have been established from the 769 patients enrolled in an Institutional Review Board-approved clinical trial. Because genomics alone was insufficient to identify therapeutic options for the majority of patients with advanced disease, we used high-throughput drug screening to discover effective treatment strategies. Analysis of tumor-derived cells from four cases, two uterine malignancies and two colon cancers, identified effective drugs and drug combinations that were subsequently validated using 3-D cultures and PDX models. This platform thereby promotes the discovery of novel therapeutic approaches that can be assessed in clinical trials and provides personalized therapeutic options for individual patients where standard clinical options have been exhausted.Significance: Integration of genomic data with drug screening from personalized in vitro and in vivo cancer models guides precision cancer care and fuels next-generation research. Cancer Discov; 7(5); 462-77. ©2017 AACR.See related commentary by Picco and Garnett, p. 456This article is highlighted in the In This Issue feature, p. 443.
Citations
More filters
Journal ArticleDOI
23 Feb 2018-Science
TL;DR: Responses to anticancer agents ex vivo in organoids and PDO-based orthotopic mouse tumor xenograft models with the responses of the patients in clinical trials are compared to suggest that PDOs can recapitulate patient responses in the clinic and could be implemented in personalized medicine programs.
Abstract: Patient-derived organoids (PDOs) have recently emerged as robust preclinical models; however, their potential to predict clinical outcomes in patients has remained unclear. We report on a living biobank of PDOs from metastatic, heavily pretreated colorectal and gastroesophageal cancer patients recruited in phase 1/2 clinical trials. Phenotypic and genotypic profiling of PDOs showed a high degree of similarity to the original patient tumors. Molecular profiling of tumor organoids was matched to drug-screening results, suggesting that PDOs could complement existing approaches in defining cancer vulnerabilities and improving treatment responses. We compared responses to anticancer agents ex vivo in organoids and PDO-based orthotopic mouse tumor xenograft models with the responses of the patients in clinical trials. Our data suggest that PDOs can recapitulate patient responses in the clinic and could be implemented in personalized medicine programs.

1,099 citations

Journal ArticleDOI
TL;DR: In this Review, Drost and Clevers discuss the recent advances in organoid models of cancer and how they can be exploited to drive the translation of basic cancer research into novel patient-specific treatment regimens in the clinic.
Abstract: The recent advances in in vitro 3D culture technologies, such as organoids, have opened new avenues for the development of novel, more physiological human cancer models. Such preclinical models are essential for more efficient translation of basic cancer research into novel treatment regimens for patients with cancer. Wild-type organoids can be grown from embryonic and adult stem cells and display self-organizing capacities, phenocopying essential aspects of the organs they are derived from. Genetic modification of organoids allows disease modelling in a setting that approaches the physiological environment. Additionally, organoids can be grown with high efficiency from patient-derived healthy and tumour tissues, potentially enabling patient-specific drug testing and the development of individualized treatment regimens. In this Review, we evaluate tumour organoid protocols and how they can be utilized as an alternative model for cancer research.

955 citations

Journal ArticleDOI
TL;DR: Common approaches to 3D culture are reviewed, the significance of 3D cultures in drug resistance and drug repositioning is discussed and some of the challenges of applying 3D cell cultures to high-throughput drug discovery are addressed.
Abstract: Drug development is a lengthy and costly process that proceeds through several stages from target identification to lead discovery and optimization, preclinical validation and clinical trials culminating in approval for clinical use. An important step in this process is high-throughput screening (HTS) of small compound libraries for lead identification. Currently, the majority of cell-based HTS is being carried out on cultured cells propagated in two-dimensions (2D) on plastic surfaces optimized for tissue culture. At the same time, compelling evidence suggests that cells cultured in these non-physiological conditions are not representative of cells residing in the complex microenvironment of a tissue. This discrepancy is thought to be a significant contributor to the high failure rate in drug discovery, where only a low percentage of drugs investigated ever make it through the gamut of testing and approval to the market. Thus, three-dimensional (3D) cell culture technologies that more closely resemble in vivo cell environments are now being pursued with intensity as they are expected to accommodate better precision in drug discovery. Here we will review common approaches to 3D culture, discuss the significance of 3D cultures in drug resistance and drug repositioning and address some of the challenges of applying 3D cell cultures to high-throughput drug discovery.

911 citations


Cites background from "Personalized In Vitro and In Vivo C..."

  • ...…tumoroids derived from patient cancer tissues that contain tumor cells and stroma cells of the tumor microenvironment are poised to provide advanced and more realistic 3D culture platforms for personalized drug evaluation and development (Xu et al., 2014; Stadler et al., 2015; Pauli et al., 2017)....

    [...]

  • ...…form hybrid culture systems that combine the advantages of both systems to form a complex microenvironment for 3D cell culture. to perform drug screens and facilitate drug development in a patient-centered manner (Stadler et al., 2015; Nath and Devi, 2016; Pauli et al., 2017; Weeber et al., 2017)....

    [...]

  • ...to perform drug screens and facilitate drug development in a patient-centered manner (Stadler et al., 2015; Nath and Devi, 2016; Pauli et al., 2017; Weeber et al., 2017)....

    [...]

  • ...In particular, tumoroids derived from patient samples are promising techniques for drug screening and drug development in precision medicine (Stadler et al., 2015; Nath and Devi, 2016; Pauli et al., 2017; Weeber et al., 2017)....

    [...]

  • ...Beyond such organoids, tumoroids derived from patient cancer tissues that contain tumor cells and stroma cells of the tumor microenvironment are poised to provide advanced and more realistic 3D culture platforms for personalized drug evaluation and development (Xu et al., 2014; Stadler et al., 2015; Pauli et al., 2017)....

    [...]

Journal ArticleDOI
TL;DR: This Review discusses key advances in the design and development of RNAi drugs leading up to this landmark achievement, the state of the current clinical pipeline and prospects for future advances, including novel RNAi pathway agents utilizing mechanisms beyond post-translational RNAi silencing.
Abstract: The RNA interference (RNAi) pathway regulates mRNA stability and translation in nearly all human cells. Small double-stranded RNA molecules can efficiently trigger RNAi silencing of specific genes, but their therapeutic use has faced numerous challenges involving safety and potency. However, August 2018 marked a new era for the field, with the US Food and Drug Administration approving patisiran, the first RNAi-based drug. In this Review, we discuss key advances in the design and development of RNAi drugs leading up to this landmark achievement, the state of the current clinical pipeline and prospects for future advances, including novel RNAi pathway agents utilizing mechanisms beyond post-translational RNAi silencing.

779 citations

Journal ArticleDOI
TL;DR: Emerging evidence indicates that epigenetic regulations and noncoding RNAs may play important roles in breast cancer development and may contribute to the heterogeneity and metastatic aspects of breast cancer, especially for triple-negative breast cancer.
Abstract: As the most commonly occurring cancer in women worldwide, breast cancer poses a formidable public health challenge on a global scale. Breast cancer consists of a group of biologically and molecularly heterogeneous diseases originated from the breast. While the risk factors associated with this cancer varies with respect to other cancers, genetic predisposition, most notably mutations in BRCA1 or BRCA2 gene, is an important causative factor for this malignancy. Breast cancers can begin in different areas of the breast, such as the ducts, the lobules, or the tissue in between. Within the large group of diverse breast carcinomas, there are various denoted types of breast cancer based on their invasiveness relative to the primary tumor sites. It is important to distinguish between the various subtypes because they have different prognoses and treatment implications. As there are remarkable parallels between normal development and breast cancer progression at the molecular level, it has been postulated that breast cancer may be derived from mammary cancer stem cells. Normal breast development and mammary stem cells are regulated by several signaling pathways, such as estrogen receptors (ERs), HER2, and Wnt/β-catenin signaling pathways, which control stem cell proliferation, cell death, cell differentiation, and cell motility. Furthermore, emerging evidence indicates that epigenetic regulations and noncoding RNAs may play important roles in breast cancer development and may contribute to the heterogeneity and metastatic aspects of breast cancer, especially for triple-negative breast cancer. This review provides a comprehensive survey of the molecular, cellular and genetic aspects of breast cancer.

609 citations

References
More filters
Journal ArticleDOI
TL;DR: Vemurafenib produced improved rates of overall and progression-free survival in patients with previously untreated melanoma with the BRAF V600E mutation in a phase 3 randomized clinical trial.
Abstract: At 6 months, overall survival was 84% (95% confidence interval [CI], 78 to 89) in the vemurafenib group and 64% (95% CI, 56 to 73) in the dacarbazine group. In the interim analysis for overall survival and final analysis for progression-free survival, vemurafenib was associated with a relative reduction of 63% in the risk of death and of 74% in the risk of either death or disease progression, as compared with dacarbazine (P<0.001 for both comparisons). After review of the interim analysis by an independent data and safety monitoring board, crossover from dacarbazine to vemurafenib was recommended. Response rates were 48% for vemurafenib and 5% for dacarbazine. Common adverse events associated with vemurafenib were arthralgia, rash, fatigue, alopecia, keratoacanthoma or squamous-cell carcinoma, photosensitivity, nausea, and diarrhea; 38% of patients required dose modification because of toxic effects. Conclusions Vemurafenib produced improved rates of overall and progression-free survival in patients with previously untreated melanoma with the BRAF V600E mutation. (Funded by Hoffmann–La Roche; BRIM-3 ClinicalTrials.gov number, NCT01006980.)

6,773 citations

Journal ArticleDOI
29 Mar 2012-Nature
TL;DR: The results indicate that large, annotated cell-line collections may help to enable preclinical stratification schemata for anticancer agents and the generation of genetic predictions of drug response in the preclinical setting and their incorporation into cancer clinical trial design could speed the emergence of ‘personalized’ therapeutic regimens.
Abstract: The systematic translation of cancer genomic data into knowledge of tumour biology and therapeutic possibilities remains challenging. Such efforts should be greatly aided by robust preclinical model systems that reflect the genomic diversity of human cancers and for which detailed genetic and pharmacological annotation is available. Here we describe the Cancer Cell Line Encyclopedia (CCLE): a compilation of gene expression, chromosomal copy number and massively parallel sequencing data from 947 human cancer cell lines. When coupled with pharmacological profiles for 24 anticancer drugs across 479 of the cell lines, this collection allowed identification of genetic, lineage, and gene-expression-based predictors of drug sensitivity. In addition to known predictors, we found that plasma cell lineage correlated with sensitivity to IGF1 receptor inhibitors; AHR expression was associated with MEK inhibitor efficacy in NRAS-mutant lines; and SLFN11 expression predicted sensitivity to topoisomerase inhibitors. Together, our results indicate that large, annotated cell-line collections may help to enable preclinical stratification schemata for anticancer agents. The generation of genetic predictions of drug response in the preclinical setting and their incorporation into cancer clinical trial design could speed the emergence of 'personalized' therapeutic regimens.

6,417 citations


"Personalized In Vitro and In Vivo C..." refers background in this paper

  • ...Large-scale drug screens of cell line panels—such as the NCI60 by the National Cancer Institute or the Cancer Cell Line Encyclopedia (CCLE)—have addressed compound sensitivity in cancer cells to identify mechanisms of growth inhibition and tumor-cell death (4, 5)....

    [...]

Journal ArticleDOI
Michael S. Lawrence1, Petar Stojanov1, Petar Stojanov2, Paz Polak1, Paz Polak3, Paz Polak2, Gregory V. Kryukov1, Gregory V. Kryukov2, Gregory V. Kryukov3, Kristian Cibulskis1, Andrey Sivachenko1, Scott L. Carter1, Chip Stewart1, Craig H. Mermel1, Craig H. Mermel2, Steven A. Roberts4, Adam Kiezun1, Peter S. Hammerman1, Peter S. Hammerman2, Aaron McKenna5, Aaron McKenna1, Yotam Drier, Lihua Zou1, Alex H. Ramos1, Trevor J. Pugh2, Trevor J. Pugh1, Nicolas Stransky1, Elena Helman6, Elena Helman1, Jaegil Kim1, Carrie Sougnez1, Lauren Ambrogio1, Elizabeth Nickerson1, Erica Shefler1, Maria L. Cortes1, Daniel Auclair1, Gordon Saksena1, Douglas Voet1, Michael S. Noble1, Daniel DiCara1, Pei Lin1, Lee Lichtenstein1, David I. Heiman1, Timothy Fennell1, Marcin Imielinski1, Marcin Imielinski2, Bryan Hernandez1, Eran Hodis1, Eran Hodis2, Sylvan C. Baca2, Sylvan C. Baca1, Austin M. Dulak1, Austin M. Dulak2, Jens G. Lohr2, Jens G. Lohr1, Dan A. Landau7, Dan A. Landau2, Dan A. Landau1, Catherine J. Wu2, Jorge Melendez-Zajgla, Alfredo Hidalgo-Miranda, Amnon Koren2, Amnon Koren1, Steven A. McCarroll2, Steven A. McCarroll1, Jaume Mora8, Ryan S. Lee9, Ryan S. Lee2, Brian D. Crompton9, Brian D. Crompton2, Robert C. Onofrio1, Melissa Parkin1, Wendy Winckler1, Kristin G. Ardlie1, Stacey Gabriel1, Charles W. M. Roberts9, Charles W. M. Roberts2, Jaclyn A. Biegel10, Kimberly Stegmaier2, Kimberly Stegmaier9, Kimberly Stegmaier1, Adam J. Bass1, Adam J. Bass2, Levi A. Garraway2, Levi A. Garraway1, Matthew Meyerson2, Matthew Meyerson1, Todd R. Golub, Dmitry A. Gordenin4, Shamil R. Sunyaev2, Shamil R. Sunyaev3, Shamil R. Sunyaev1, Eric S. Lander2, Eric S. Lander1, Eric S. Lander6, Gad Getz1, Gad Getz2 
11 Jul 2013-Nature
TL;DR: A fundamental problem with cancer genome studies is described: as the sample size increases, the list of putatively significant genes produced by current analytical methods burgeons into the hundreds and the list includes many implausible genes, suggesting extensive false-positive findings that overshadow true driver events.
Abstract: Major international projects are underway that are aimed at creating a comprehensive catalogue of all the genes responsible for the initiation and progression of cancer. These studies involve the sequencing of matched tumour-normal samples followed by mathematical analysis to identify those genes in which mutations occur more frequently than expected by random chance. Here we describe a fundamental problem with cancer genome studies: as the sample size increases, the list of putatively significant genes produced by current analytical methods burgeons into the hundreds. The list includes many implausible genes (such as those encoding olfactory receptors and the muscle protein titin), suggesting extensive false-positive findings that overshadow true driver events. We show that this problem stems largely from mutational heterogeneity and provide a novel analytical methodology, MutSigCV, for resolving the problem. We apply MutSigCV to exome sequences from 3,083 tumour-normal pairs and discover extraordinary variation in mutation frequency and spectrum within cancer types, which sheds light on mutational processes and disease aetiology, and in mutation frequency across the genome, which is strongly correlated with DNA replication timing and also with transcriptional activity. By incorporating mutational heterogeneity into the analyses, MutSigCV is able to eliminate most of the apparent artefactual findings and enable the identification of genes truly associated with cancer.

4,411 citations

Journal ArticleDOI
TL;DR: The MuTect algorithm for calling somatic point mutations enables subclonal analysis of the whole-genome or whole-exome sequencing data being generated in large-scale cancer genomics projects as discussed by the authors.
Abstract: The MuTect algorithm for calling somatic point mutations enables subclonal analysis of the whole-genome or whole-exome sequencing data being generated in large-scale cancer genomics projects.

3,773 citations

Journal ArticleDOI
TL;DR: Crizotinib is superior to standard chemotherapy in patients with previously treated, advanced non-small-cell lung cancer with ALK rearrangement and greater improvement in global quality of life with crizotinIB than with chemotherapy.
Abstract: BACKGROUND: In single-group studies, chromosomal rearrangements of the anaplastic lymphoma kinase gene (ALK ) have been associated with marked clinical responses to crizotinib, an oral tyrosine kinase inhibitor targeting ALK. Whether crizotinib is superior to standard chemotherapy with respect to efficacy is unknown. METHODS: We conducted a phase 3, open-label trial comparing crizotinib with chemotherapy in 347 patients with locally advanced or metastatic ALK-positive lung cancer who had received one prior platinum-based regimen. Patients were randomly assigned to receive oral treatment with crizotinib (250 mg) twice daily or intravenous chemotherapy with either pemetrexed (500 mg per square meter of body-surface area) or docetaxel (75 mg per square meter) every 3 weeks. Patients in the chemotherapy group who had disease progression were permitted to cross over to crizotinib as part of a separate study. The primary end point was progression-free survival. RESULTS: The median progression-free survival was 7.7 months in the crizotinib group and 3.0 months in the chemotherapy group (hazard ratio for progression or death with crizotinib, 0.49; 95% confidence interval [CI], 0.37 to 0.64; P<0.001). The response rates were 65% (95% CI, 58 to 72) with crizotinib, as compared with 20% (95% CI, 14 to 26) with chemotherapy (P<0.001). An interim analysis of overall survival showed no significant improvement with crizotinib as compared with chemotherapy (hazard ratio for death in the crizotinib group, 1.02; 95% CI, 0.68 to 1.54; P=0.54). Common adverse events associated with crizotinib were visual disorder, gastrointestinal side effects, and elevated liver aminotransferase levels, whereas common adverse events with chemotherapy were fatigue, alopecia, and dyspnea. Patients reported greater reductions in symptoms of lung cancer and greater improvement in global quality of life with crizotinib than with chemotherapy. CONCLUSIONS: Crizotinib is superior to standard chemotherapy in patients with previously treated, advanced non-small-cell lung cancer with ALK rearrangement. (Funded by Pfizer; ClinicalTrials.gov number, NCT00932893.) Copyright © 2013 Massachusetts Medical Society.

3,074 citations

Related Papers (5)