201
|
Langenberg KPS, Looze EJ, Molenaar JJ. The Landscape of Pediatric Precision Oncology: Program Design, Actionable Alterations, and Clinical Trial Development. Cancers (Basel) 2021; 13:4324. [PMID: 34503139 PMCID: PMC8431194 DOI: 10.3390/cancers13174324] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022] Open
Abstract
Over the last years, various precision medicine programs have been developed for pediatric patients with high-risk, relapsed, or refractory malignancies, selecting patients for targeted treatment through comprehensive molecular profiling. In this review, we describe characteristics of these initiatives, demonstrating the feasibility and potential of molecular-driven precision medicine. Actionable events are identified in a significant subset of patients, although comparing results is complicated due to the lack of a standardized definition of actionable alterations and the different molecular profiling strategies used. The first biomarker-driven trials for childhood cancer have been initiated, but until now the effect of precision medicine on clinical outcome has only been reported for a small number of patients, demonstrating clinical benefit in some. Future perspectives include the incorporation of novel approaches such as liquid biopsies and immune monitoring as well as innovative collaborative trial design including combination strategies, and the development of agents specifically targeting aberrations in childhood malignancies.
Collapse
Affiliation(s)
- Karin P. S. Langenberg
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (E.J.L.); (J.J.M.)
| | - Eleonora J. Looze
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (E.J.L.); (J.J.M.)
| | - Jan J. Molenaar
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (E.J.L.); (J.J.M.)
- Department of Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
202
|
Bērziņa S, Harrison A, Taly V, Xiao W. Technological Advances in Tumor-On-Chip Technology: From Bench to Bedside. Cancers (Basel) 2021; 13:cancers13164192. [PMID: 34439345 PMCID: PMC8394443 DOI: 10.3390/cancers13164192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Various 3D in vitro tumor models are rapidly advancing cancer research. Unlike animal models, they can be produced quickly and are amenable to high-throughput studies. Growing tumor spheroids in microfluidic tumor-on-chip platforms has particularly elevated the capabilities of such models. Tumor-on-chip devices can mimic multiple aspects of the dynamic in vivo tumor microenvironment in a precisely controlled manner. Moreover, new technologies for the on- and off-chip analysis of these tumor mimics are continuously emerging. There is thus an urgent need to review the latest developments in this rapidly progressing field. Here, we present an overview of the technological advances in tumor-on-chip technology by reviewing state-of-the-art tools for on-chip analysis. In particular, we evaluate the potential for tumor-on-chip technology to guide personalized cancer therapies. We strive to appeal to cancer researchers and biomedical engineers alike, informing on current progress, while provoking thought on the outstanding developments needed to achieve clinical-stage research. Abstract Tumor-on-chip technology has cemented its importance as an in vitro tumor model for cancer research. Its ability to recapitulate different elements of the in vivo tumor microenvironment makes it promising for translational medicine, with potential application in enabling personalized anti-cancer therapies. Here, we provide an overview of the current technological advances for tumor-on-chip generation. To further elevate the functionalities of the technology, these approaches need to be coupled with effective analysis tools. This aspect of tumor-on-chip technology is often neglected in the current literature. We address this shortcoming by reviewing state-of-the-art on-chip analysis tools for microfluidic tumor models. Lastly, we focus on the current progress in tumor-on-chip devices using patient-derived samples and evaluate their potential for clinical research and personalized medicine applications.
Collapse
|
203
|
Jordan AM. Molecularly profiled trials: toward a framework of actions for the "nil actionables". Br J Cancer 2021; 125:473-478. [PMID: 34040178 PMCID: PMC8150144 DOI: 10.1038/s41416-021-01423-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 02/02/2023] Open
Abstract
The sequencing of tumour or blood samples is increasingly used to stratify patients into clinical trials of molecularly targeted agents, and this approach has frequently demonstrated clinical benefit for those who are deemed eligible. But what of those who have no clear and evident molecular driver? What of those deemed to have "nil actionable" mutations? How might we deliver better therapeutic opportunities for those left behind in the clamour toward stratified therapeutics? And what significant learnings lie hidden in the data we amass but do not interrogate and understand? This Perspective article suggests a holistic approach to the future treatment of such patients, and sets a framework through which significant additional patient benefit might be achieved. In order to deliver upon this framework, it encourages and invites the clinical community to engage more enthusiastically and share learnings with colleagues in the early drug discovery community, in order to deliver a step change in patient care.
Collapse
|
204
|
Horak P, Leichsenring J, Goldschmid H, Kreutzfeldt S, Kazdal D, Teleanu V, Endris V, Gieldon L, Allgäuer M, Volckmar AL, Dikow N, Renner M, Kirchner M, Penzel R, Ploeger C, Brandt R, Seker-Cin H, Budczies J, Heilig CE, Neumann O, Schaaf CP, Schirmacher P, Fröhling S, Stenzinger A. Assigning evidence to actionability: An introduction to variant interpretation in precision cancer medicine. Genes Chromosomes Cancer 2021; 61:303-313. [PMID: 34331337 DOI: 10.1002/gcc.22987] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/25/2021] [Indexed: 12/15/2022] Open
Abstract
Modern concepts in precision cancer medicine are based on increasingly complex genomic analyses and require standardized criteria for the functional evaluation and reporting of detected genomic alterations in order to assess their clinical relevance. In this article, we propose and address the necessary steps in systematic variant evaluation consisting of bioinformatic analysis, functional annotation and clinical interpretation, focusing on the latter two aspects. We discuss the role and clinical application of current variant classification systems and point out their scope and limitations. Finally, we highlight the significance of the molecular tumor board as a platform for clinical decision-making based on genomic analyses.
Collapse
Affiliation(s)
- Peter Horak
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Jonas Leichsenring
- Institut für Pathologie, Zytologie und molekulare Diagnostik, Regiomed Klinikum Coburg, Coburg, Germany.,Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hannah Goldschmid
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Simon Kreutzfeldt
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
| | - Daniel Kazdal
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany
| | - Veronica Teleanu
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Volker Endris
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Laura Gieldon
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Michael Allgäuer
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Anna-Lena Volckmar
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Nicola Dikow
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Marcus Renner
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
| | - Martina Kirchner
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Roland Penzel
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Carolin Ploeger
- Center for Personalized Medicine (ZPM), Heidelberg, Germany.,Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Regine Brandt
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Huriye Seker-Cin
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jan Budczies
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany
| | - Christoph E Heilig
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
| | - Olaf Neumann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Peter Schirmacher
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Center for Personalized Medicine (ZPM), Heidelberg, Germany.,Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Albrecht Stenzinger
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.,Center for Personalized Medicine (ZPM), Heidelberg, Germany.,Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany
| |
Collapse
|
205
|
De Falco V, Poliero L, Vitello PP, Ciardiello D, Vitale P, Zanaletti N, Giunta EF, Terminiello M, Caputo V, Carlino F, Di Liello R, Ventriglia A, Famiglietti V, Martinelli E, Morgillo F, Orditura M, De Vita F, Fasano M, Napolitano S, Martini G, Della Corte CM, Franco R, Altucci L, Ciardiello F, Troiani T. Feasibility of next-generation sequencing in clinical practice: results of a pilot study in the Department of Precision Medicine at the University of Campania 'Luigi Vanvitelli'. ESMO Open 2021; 5:S2059-7029(20)30067-3. [PMID: 32234730 PMCID: PMC7174013 DOI: 10.1136/esmoopen-2020-000675] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/28/2022] Open
Abstract
Background The emerging role of next-generation sequencing (NGS) targeted panels is revolutionising our approach to cancer patients, providing information on gene alterations helpful for diagnosis and clinical decision, in a short time and with acceptable costs. Materials and methods In this work, we evaluated the clinical application of FoundationOne CDx test, a hybrid capture-based NGS. This test identifies alterations in 324 genes, tumour mutational burden and genomic signatures as microsatellite instability. The decision to obtain the NGS assay for a particular patient was done according to investigator’s choice. Results Overall, 122 tumour specimens were analysed, of which 84 (68.85%) succeeded. The success rate was influenced by type of specimen formalin-fixed paraffin embedded (FFPE block vs FFPE slides), by origin of the sample (surgery vs biopsy) and by time of fixation (<5 years vs ≥5 years). The most frequent subgroups of effective reports derived from colorectal cancer (25 samples), non-small-cell lung cancer (16 samples), ovarian cancer (10 samples), biliary tract cancer (9 samples), breast cancer (7 samples), gastric cancer (7 samples). The most frequent alterations found in whole population referred to TP53 (45.9%), KRAS (19.6%) and APC (13.9%). Furthermore, we performed an analysis of patients in whom this comprehensive genomic profiling (CGP) had a relevance for the patient’s disease. Conclusions On our opinion, CGP could be proposed in clinical practice in order to select patients that could most benefit from the analysis proposed, like patients with good performance status without any available treatments or with unexpected resistance to a therapy.
Collapse
Affiliation(s)
- Vincenzo De Falco
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Luca Poliero
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Pietro Paolo Vitello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Davide Ciardiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Pasquale Vitale
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Nicoletta Zanaletti
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Emilio Francesco Giunta
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Marinella Terminiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Vincenza Caputo
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Francesca Carlino
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Raimondo Di Liello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Anna Ventriglia
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Vincenzo Famiglietti
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Erika Martinelli
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Floriana Morgillo
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Michele Orditura
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Ferdinando De Vita
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Morena Fasano
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Stefania Napolitano
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Giulia Martini
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Carminia Maria Della Corte
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Renato Franco
- Department of Mental, Physical Health and Preventive Medicine, University of Campania Luigi Vanvitelli, Napoli, Italy
| | - Lucia Altucci
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Fortunato Ciardiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Teresa Troiani
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| |
Collapse
|
206
|
Gil DA, Deming DA, Skala MC. Volumetric growth tracking of patient-derived cancer organoids using optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2021; 12:3789-3805. [PMID: 34457380 PMCID: PMC8367263 DOI: 10.1364/boe.428197] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 05/02/2023]
Abstract
Patient-derived cancer organoids (PCOs) are in vitro organotypic models that reflect in vivo drug response, thus PCOs are an accessible model for cancer drug screening in a clinically relevant timeframe. However, current methods to assess the response of PCOs are limited. Here, a custom swept-source optical coherence tomography (OCT) system was used to rapidly evaluate volumetric growth and drug response in PCOs. This system was optimized for an inverted imaging geometry to enable high-throughput imaging of PCOs. An automated image analysis framework was developed to perform 3D single-organoid tracking of PCOs across multiple time points over 48 hours. Metabolic inhibitors and cancer therapies decreased PCOs volumetric growth rate compared to control PCOs. Single-organoid tracking improved sensitivity to drug treatment compared to a pooled analysis of changes in organoid volume. OCT provided a more accurate assessment of organoid volume compared to a volume estimation method based on 2D projections. Single-organoid tracking with OCT also identified heterogeneity in drug response between solid and hollow PCOs. This work demonstrates that OCT and 3D single-organoid tracking are attractive tools to monitor volumetric growth and drug response in PCOs, providing rapid, non-destructive methods to quantify heterogeneity in PCOs.
Collapse
Affiliation(s)
- Daniel A. Gil
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53704, USA
- Morgridge Institute for Research, Madison, WI 53704, USA
| | - Dustin A. Deming
- University of Wisconsin Carbone Cancer Center, Madison, WI 53704, USA
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin, Madison, WI 53704, USA
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI 53704, USA
| | - Melissa C. Skala
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53704, USA
- Morgridge Institute for Research, Madison, WI 53704, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI 53704, USA
| |
Collapse
|
207
|
Petak I, Kamal M, Dirner A, Bieche I, Doczi R, Mariani O, Filotas P, Salomon A, Vodicska B, Servois V, Varkondi E, Gentien D, Tihanyi D, Tresca P, Lakatos D, Servant N, Deri J, du Rusquec P, Hegedus C, Bello Roufai D, Schwab R, Dupain C, Valyi-Nagy IT, Le Tourneau C. A computational method for prioritizing targeted therapies in precision oncology: performance analysis in the SHIVA01 trial. NPJ Precis Oncol 2021; 5:59. [PMID: 34162980 PMCID: PMC8222375 DOI: 10.1038/s41698-021-00191-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 05/13/2021] [Indexed: 01/25/2023] Open
Abstract
Precision oncology is currently based on pairing molecularly targeted agents (MTA) to predefined single driver genes or biomarkers. Each tumor harbors a combination of a large number of potential genetic alterations of multiple driver genes in a complex system that limits the potential of this approach. We have developed an artificial intelligence (AI)-assisted computational method, the digital drug-assignment (DDA) system, to prioritize potential MTAs for each cancer patient based on the complex individual molecular profile of their tumor. We analyzed the clinical benefit of the DDA system on the molecular and clinical outcome data of patients treated in the SHIVA01 precision oncology clinical trial with MTAs matched to individual genetic alterations or biomarkers of their tumor. We found that the DDA score assigned to MTAs was significantly higher in patients experiencing disease control than in patients with progressive disease (1523 versus 580, P = 0.037). The median PFS was also significantly longer in patients receiving MTAs with high (1000+ <) than with low (<0) DDA scores (3.95 versus 1.95 months, P = 0.044). Our results indicate that AI-based systems, like DDA, are promising new tools for oncologists to improve the clinical benefit of precision oncology.
Collapse
Affiliation(s)
- Istvan Petak
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, USA.
- Oncompass Medicine, Budapest, Hungary.
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institute Curie, Paris & Saint-Cloud, France
| | | | - Ivan Bieche
- Pharmacogenomics unit, Institut Curie, Paris, France
| | | | - Odette Mariani
- Department of Biopathology, Institut Curie, Paris, France
| | | | - Anne Salomon
- Department of Biopathology, Institut Curie, Paris, France
| | | | | | | | - David Gentien
- Translational Research Department, Institut Curie, Paris, France
| | | | - Patricia Tresca
- Department of Drug Development and Innovation (D3i), Institute Curie, Paris & Saint-Cloud, France
| | | | | | | | - Pauline du Rusquec
- Department of Drug Development and Innovation (D3i), Institute Curie, Paris & Saint-Cloud, France
| | | | - Diana Bello Roufai
- Department of Drug Development and Innovation (D3i), Institute Curie, Paris & Saint-Cloud, France
| | | | - Celia Dupain
- Department of Drug Development and Innovation (D3i), Institute Curie, Paris & Saint-Cloud, France
| | - Istvan T Valyi-Nagy
- Central Hospital of Southern Pest-National Institute for Hematology and Infectious Diseases, Budapest, Hungary.
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institute Curie, Paris & Saint-Cloud, France.
- INSERM U900 Research Unit, Paris & Saint-Cloud, France.
- Paris-Saclay University, Paris, France.
| |
Collapse
|
208
|
Criteria-based curation of a therapy-focused compendium to support treatment recommendations in precision oncology. NPJ Precis Oncol 2021; 5:58. [PMID: 34162978 PMCID: PMC8222322 DOI: 10.1038/s41698-021-00194-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/26/2021] [Indexed: 11/09/2022] Open
Abstract
While several resources exist that interpret therapeutic significance of genomic alterations in cancer, many regional real-world issues limit access to drugs. There is a need for a pragmatic, evidence-based, context-adapted tool to guide clinical management based on molecular biomarkers. To this end, we have structured a compendium of approved and experimental therapies with associated biomarkers following a survey of drug regulatory databases, existing knowledge bases, and published literature. Each biomarker-disease-therapy triplet was categorised using a tiering system reflective of key therapeutic considerations: approved and reimbursed therapies with respect to a jurisdiction (Tier 1), evidence of efficacy or approval in another jurisdiction (Tier 2), evidence of antitumour activity (Tier 3), and plausible biological rationale (Tier 4). Two resistance categories were defined: lack of efficacy (Tier R1) or antitumor activity (Tier R2). Based on this framework, we curated a digital resource focused on drugs relevant in the Australian healthcare system (TOPOGRAPH: Therapy Oriented Precision Oncology Guidelines for Recommending Anticancer Pharmaceuticals). As of November 2020, TOPOGRAPH comprised 2810 biomarker-disease-therapy triplets in 989 expert-appraised entries, including 373 therapies, 199 biomarkers, and 106 cancer types. In the 345 therapies catalogued, 84 (24%) and 65 (19%) were designated Tiers 1 and 2, respectively, while 271 (79%) therapies were supported by preclinical studies, early clinical trials, retrospective studies, or case series (Tiers 3 and 4). A companion algorithm was also developed to support rational, context-appropriate treatment selection informed by molecular biomarkers. This framework can be readily adapted to build similar resources in other jurisdictions to support therapeutic decision-making.
Collapse
|
209
|
Vidula N, Niemierko A, Malvarosa G, Yuen M, Lennerz J, Iafrate AJ, Wander SA, Spring L, Juric D, Isakoff S, Younger J, Moy B, Ellisen LW, Bardia A. Tumor Tissue- versus Plasma-based Genotyping for Selection of Matched Therapy and Impact on Clinical Outcomes in Patients with Metastatic Breast Cancer. Clin Cancer Res 2021; 27:3404-3413. [PMID: 33504549 DOI: 10.1158/1078-0432.ccr-20-3444] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/08/2020] [Accepted: 01/22/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Actionable mutations can guide genotype-directed matched therapy. We evaluated the utility of tissue-based and plasma-based genotyping for the identification of actionable mutations and selection of matched therapy in patients with metastatic breast cancer (MBC). EXPERIMENTAL DESIGN Patients with MBC who underwent tissue genotyping (institutional platform, 91-gene assay) or plasma-based cell-free DNA (cfDNA, Guardant360, 73-gene assay) between January 2016 and December 2017 were included. A chart review of records to identify subtype, demographics, treatment, outcomes, and tissue genotyping or cfDNA results was performed. The incidence of actionable mutations and the selection of matched therapy in tissue genotyping or cfDNA cohorts was determined. The impact of matched therapy status on overall survival (OS) in tissue genotyping or cfDNA subgroups was determined with Cox regression analysis. RESULTS Of 252 patients who underwent cfDNA testing, 232 (92%) had detectable mutations, 196 (78%) had actionable mutations, and 86 (34%) received matched therapy. Of 118 patients who underwent tissue genotyping, 90 (76%) had detectable mutations, 59 (50%) had actionable mutations, and 13 (11%) received matched therapy. For cfDNA patients with actionable mutations, matched versus nonmatched therapy was associated with better OS [HR 0.41, 95% confidence interval (CI): 0.23-0.73, P = 0.002], and this remained significant in a multivariable analysis correcting for age, subtype, visceral metastases, and brain metastases (HR = 0.46, 95% CI: 0.26-0.83, P = 0.010). CONCLUSIONS Plasma-based genotyping identified high rates of actionable mutations, which was associated with significant application of matched therapy and better OS in patients with MBC.See related commentary by Rugo and Huppert, p. 3275.
Collapse
Affiliation(s)
- Neelima Vidula
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.
| | - Andrzej Niemierko
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Giuliana Malvarosa
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Megan Yuen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Jochen Lennerz
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - A John Iafrate
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Seth A Wander
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Laura Spring
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Dejan Juric
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Steven Isakoff
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Jerry Younger
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Leif W Ellisen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
210
|
Bagaev A, Kotlov N, Nomie K, Svekolkin V, Gafurov A, Isaeva O, Osokin N, Kozlov I, Frenkel F, Gancharova O, Almog N, Tsiper M, Ataullakhanov R, Fowler N. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 2021; 39:845-865.e7. [PMID: 34019806 DOI: 10.1016/j.ccell.2021.04.014] [Citation(s) in RCA: 549] [Impact Index Per Article: 183.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/14/2020] [Accepted: 04/23/2021] [Indexed: 12/18/2022]
Abstract
The clinical use of molecular targeted therapy is rapidly evolving but has primarily focused on genomic alterations. Transcriptomic analysis offers an opportunity to dissect the complexity of tumors, including the tumor microenvironment (TME), a crucial mediator of cancer progression and therapeutic outcome. TME classification by transcriptomic analysis of >10,000 cancer patients identifies four distinct TME subtypes conserved across 20 different cancers. The TME subtypes correlate with patient response to immunotherapy in multiple cancers, with patients possessing immune-favorable TME subtypes benefiting the most from immunotherapy. Thus, the TME subtypes act as a generalized immunotherapy biomarker across many cancer types due to the inclusion of malignant and microenvironment components. A visual tool integrating transcriptomic and genomic data provides a global tumor portrait, describing the tumor framework, mutational load, immune composition, anti-tumor immunity, and immunosuppressive escape mechanisms. Integrative analyses plus visualization may aid in biomarker discovery and the personalization of therapeutic regimens.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Nathan Fowler
- BostonGene, Waltham, MA 02453, USA; Department of Lymphoma and Myeloma, Unit 0429, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
211
|
Mock A, Plath M, Moratin J, Tapken MJ, Jäger D, Krauss J, Fröhling S, Hess J, Zaoui K. EGFR and PI3K Pathway Activities Might Guide Drug Repurposing in HPV-Negative Head and Neck Cancers. Front Oncol 2021; 11:678966. [PMID: 34178665 PMCID: PMC8226088 DOI: 10.3389/fonc.2021.678966] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/13/2021] [Indexed: 12/18/2022] Open
Abstract
While genetic alterations in Epidermal growth factor receptor (EGFR) and PI3K are common in head and neck squamous cell carcinomas (HNSCC), their impact on oncogenic signaling and cancer drug sensitivities remains elusive. To determine their consequences on the transcriptional network, pathway activities of EGFR, PI3K, and 12 additional oncogenic pathways were inferred in 498 HNSCC samples of The Cancer Genome Atlas using PROGENy. More than half of HPV-negative HNSCC showed a pathway activation in EGFR or PI3K. An amplification in EGFR and a mutation in PI3KCA resulted in a significantly higher activity of the respective pathway (p = 0.017 and p = 0.007). Interestingly, both pathway activations could only be explained by genetic alterations in less than 25% of cases indicating additional molecular events involved in the downstream signaling. Suitable in vitro pathway models could be identified in a published drug screen of 45 HPV-negative HNSCC cell lines. An active EGFR pathway was predictive for the response to the PI3K inhibitor buparlisib (p = 6.36E-03) and an inactive EGFR and PI3K pathway was associated with efficacy of the B-cell lymphoma (BCL) inhibitor navitoclax (p = 9.26E-03). In addition, an inactive PI3K pathway correlated with a response to multiple Histone deacetylase inhibitor (HDAC) inhibitors. These findings require validation in preclinical models and clinical studies.
Collapse
Affiliation(s)
- Andreas Mock
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany.,Division of Translational Medical Oncology, NCT Heidelberg, German Cancer Center (DKFZ), Heidelberg, Germany
| | - Michaela Plath
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Julius Moratin
- Department of Oral and Cranio-Maxillofacial Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Maria Johanna Tapken
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Jürgen Krauss
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, NCT Heidelberg, German Cancer Center (DKFZ), Heidelberg, Germany
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Molecular Mechanisms of Head and Neck Tumors, DKFZ, Heidelberg, Germany
| | - Karim Zaoui
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
212
|
Martinez F, Brucks E, Otsuji J, Mehnoor H, Arif-Tiwari H, Babiker HM, Recio-Boiles A. Using Advanced Molecular Profiling to Identify the Origin of and Tailor Treatment for an Intracranial Mass of Unknown Primary. JCO Precis Oncol 2021; 5:981-987. [PMID: 34136743 PMCID: PMC8202556 DOI: 10.1200/po.20.00243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 03/11/2021] [Accepted: 04/28/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
| | - Eric Brucks
- Department of Medicine, University of Arizona, Tucson, AZ
| | - Janelle Otsuji
- Department of Pathology, University of Arizona, Tucson, AZ
| | | | - Hina Arif-Tiwari
- Department of Medical Imaging, University of Arizona, Tucson, AZ
| | - Hani M Babiker
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Alejandro Recio-Boiles
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| |
Collapse
|
213
|
Seguin L, Odouard S, Corlazzoli F, Haddad SA, Moindrot L, Calvo Tardón M, Yebra M, Koval A, Marinari E, Bes V, Guérin A, Allard M, Ilmjärv S, Katanaev VL, Walker PR, Krause KH, Dutoit V, Sarkaria JN, Dietrich PY, Cosset É. Macropinocytosis requires Gal-3 in a subset of patient-derived glioblastoma stem cells. Commun Biol 2021; 4:718. [PMID: 34112916 PMCID: PMC8192788 DOI: 10.1038/s42003-021-02258-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Recently, we involved the carbohydrate-binding protein Galectin-3 (Gal-3) as a druggable target for KRAS-mutant-addicted lung and pancreatic cancers. Here, using glioblastoma patient-derived stem cells (GSCs), we identify and characterize a subset of Gal-3high glioblastoma (GBM) tumors mainly within the mesenchymal subtype that are addicted to Gal-3-mediated macropinocytosis. Using both genetic and pharmacologic inhibition of Gal-3, we showed a significant decrease of GSC macropinocytosis activity, cell survival and invasion, in vitro and in vivo. Mechanistically, we demonstrate that Gal-3 binds to RAB10, a member of the RAS superfamily of small GTPases, and β1 integrin, which are both required for macropinocytosis activity and cell survival. Finally, by defining a Gal-3/macropinocytosis molecular signature, we could predict sensitivity to this dependency pathway and provide proof-of-principle for innovative therapeutic strategies to exploit this Achilles' heel for a significant and unique subset of GBM patients.
Collapse
Affiliation(s)
- Laetitia Seguin
- University Côte d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging (IRCAN), Nice, France
| | - Soline Odouard
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Francesca Corlazzoli
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Sarah Al Haddad
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Laurine Moindrot
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Marta Calvo Tardón
- Laboratory of Immunobiology of brain tumors, Center for Translational Research in Onco-Hematology, Geneva University Hospitals, and University of Geneva, Geneva, Switzerland
| | - Mayra Yebra
- Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Alexey Koval
- Department of Cell Physiology and Metabolism, Medical School, University of Geneva, Geneva, Switzerland
| | - Eliana Marinari
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Viviane Bes
- Laboratory of Immunobiology of brain tumors, Center for Translational Research in Onco-Hematology, Geneva University Hospitals, and University of Geneva, Geneva, Switzerland
| | - Alexandre Guérin
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Geneva, Switzerland
| | - Mathilde Allard
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Sten Ilmjärv
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Geneva, Switzerland
| | - Vladimir L Katanaev
- Department of Cell Physiology and Metabolism, Medical School, University of Geneva, Geneva, Switzerland
| | - Paul R Walker
- Laboratory of Immunobiology of brain tumors, Center for Translational Research in Onco-Hematology, Geneva University Hospitals, and University of Geneva, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Geneva, Switzerland
| | - Valérie Dutoit
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Pierre-Yves Dietrich
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Érika Cosset
- Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
214
|
Horak P, Heining C, Kreutzfeldt S, Hutter B, Mock A, Hullein J, Frohlich M, Uhrig S, Jahn A, Rump A, Gieldon L, Mohrmann L, Hanf D, Teleanu V, Heilig CE, Lipka DB, Allgauer M, Ruhnke L, Lassmann A, Endris V, Neumann O, Penzel R, Beck K, Richter D, Winter U, Wolf S, Pfutze K, Georg C, Meissburger B, Buchhalter I, Augustin M, Aulitzky WE, Hohenberger P, Kroiss M, Schirmacher P, Schlenk RF, Keilholz U, Klauschen F, Folprecht G, Bauer S, Siveke JT, Brandts CH, Kindler T, Boerries M, Illert AL, von Bubnoff N, Jost PJ, Spiekermann K, Bitzer M, Schulze-Osthoff K, von Kalle C, Klink B, Brors B, Stenzinger A, Schrock E, Hubschmann D, Weichert W, Glimm H, Frohling S. Comprehensive Genomic and Transcriptomic Analysis for Guiding Therapeutic Decisions in Patients with Rare Cancers. Cancer Discov 2021; 11:2780-2795. [PMID: 34112699 DOI: 10.1158/2159-8290.cd-21-0126] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/03/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022]
Abstract
The clinical relevance of comprehensive molecular analysis in rare cancers is not established. We analyzed the molecular profiles and clinical outcomes of 1,310 patients (rare cancers, 75.5%) enrolled in a prospective observational study by the German Cancer Consortium that applies whole-genome/exome and RNA sequencing to inform the care of adults with incurable cancers. Based on 472 single and six composite biomarkers, a cross-institutional molecular tumor board provided evidence-based management recommendations, including diagnostic reevaluation, genetic counseling, and experimental treatment, in 88% of cases. Recommended therapies were administered in 362 of 1,138 patients (31.8%) and resulted in significantly improved overall response and disease control rates (23.9% and 55.3%) compared to previous therapies, translating into a progression-free survival ratio >1.3 in 35.7% of patients. These data demonstrate the benefit of molecular stratification in rare cancers and represent a resource that may promote clinical trial access and drug approvals in this underserved patient population.
Collapse
Affiliation(s)
- Peter Horak
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg
| | - Christoph Heining
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden
| | | | - Barbara Hutter
- Division of Applied Bioinformatics, German Cancer Research Center
| | | | | | - Martina Frohlich
- Computational Oncology, Molecular Diagnostics Program, German Cancer Research Center
| | - Sebastian Uhrig
- Division of Applied Bioinformatics, German Cancer Research Center
| | - Arne Jahn
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technical University Dresden
| | - Andreas Rump
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus
| | - Laura Gieldon
- Heidelberg University Hospital, Institute of Human Genetics
| | - Lino Mohrmann
- Translational Medical Oncology, National Center for Tumor Diseases Dresden
| | - Dorothea Hanf
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden
| | - Veronica Teleanu
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg
| | - Christoph E Heilig
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg
| | - Daniel B Lipka
- Section Translational Cancer Epigenomics; Division Translational Medical Oncology, German Cancer Research Center
| | | | - Leo Ruhnke
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden
| | - Andreas Lassmann
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg
| | | | - Olaf Neumann
- Department of General Pathology, University Hospital Heidelberg
| | | | - Katja Beck
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg
| | | | - Ulrike Winter
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg
| | - Stephan Wolf
- Genomics and Proteomics Core Facility, German Cancer Research Center
| | - Katrin Pfutze
- Center for Personalized Medicine, National Center for Tumor Diseases
| | - Christina Georg
- Department of Translational Oncology, National Center for Tumor Diseases
| | - Bettina Meissburger
- Sample Processing Laboratory, Molecular Diagnostics Program, German Cancer Research Center
| | - Ivo Buchhalter
- Omics IT and Data Management Core Facility, German Cancer Research Center
| | - Marinela Augustin
- Department of Hematology and Oncology, Paracelsus Medical University, Nuremberg
| | | | | | - Matthias Kroiss
- Comprehensive Cancer Center Mainfranken, University of Würzburg
| | | | - Richard F Schlenk
- NCT Clinical Trials Center, Heidelberg University Hospital and German Cancer Research Center
| | | | | | - Gunnar Folprecht
- University Cancer Center / Medical Department I, University Hospital Carl Gustav Carus
| | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, Essen, Germany; DKTK partner site Essen and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Jens Thomas Siveke
- West German Cancer Center, University Hospital Essen, Bridge Institute of Experimental Tumor Therapy
| | - Christian H Brandts
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University
| | - Thomas Kindler
- Third Department of Medicine, University Medical Center of the Johannes Gutenberg University
| | - Melanie Boerries
- Medical Center - University Freiburg, Institute of Medical Bioinformatics and Systems Medicine
| | - Anna L Illert
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, Medical Center, University of Schleswig-Holstein, Campus Lübeck
| | | | | | | | | | | | - Barbara Klink
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, TU Dresden
| | - Benedikt Brors
- Department of Applied Bioinformatics, German Cancer Research Center
| | | | - Evelin Schrock
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technical University Dresden
| | | | - Wilko Weichert
- Institute of General Pathology and Pathological Anatomy, Technical University of Munich
| | - Hanno Glimm
- Department of Translational Oncology, NCT National Center for Tumor Diseases
| | - Stefan Frohling
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg
| |
Collapse
|
215
|
du Rusquec P, Le Tourneau C. Drug Development in Tissue-Agnostic Indications. Cancers (Basel) 2021; 13:2758. [PMID: 34199382 PMCID: PMC8199632 DOI: 10.3390/cancers13112758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/14/2022] Open
Abstract
A better understanding of cancer biology has led to the development of targeted therapies specifically designed to modulate an altered molecular pathway in the cancer cells or their microenvironment. Despite the identification of molecular targets across cancer types, most of targeted therapies were developed per cancer type. In this ancestral paradigm, randomization was the gold-standard approach for market access. Randomization of large patient populations was feasible for drugs developed in common cancer types but more challenging in rare cancer types. The traditional paradigm of drug development in oncology was further challenged by the ever-expanding molecular segmentation of cancer with ever-smaller subgroups of patients who might benefit from specific targeted therapies or immunotherapies and the identification of molecular alterations against which drugs may be effective across cancer types. In this novel drug development paradigm, novel ways of evaluating the efficacy of drugs are highly needed in these small patient populations. One approach is to use each patient as his/her own control by comparing the efficacy of a drug to the efficacy of prior treatments received. This approach allows to overcome patient heterogeneity, especially in a tissue-agnostic drug development paradigm.
Collapse
Affiliation(s)
- Pauline du Rusquec
- Department of Drug Development and Innovation (D3i), Institut Curie, 75005 Paris, France;
- INSERM U900, 92210 Saint-Cloud, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, 75005 Paris, France;
- INSERM U900, 92210 Saint-Cloud, France
- Faculty of Medicine, Paris-Saclay University, 78180 Montigny le Bretonneux, France
| |
Collapse
|
216
|
de Guillebon E, Jimenez M, Mazzarella L, Betsou F, Stadler P, Peták I, Jeannot E, Chanas L, Servant N, Marret G, Duso BA, Legrand F, Kornerup KN, Bernhart SH, Balogh G, Dóczi R, Filotás P, Curigliano G, Bièche I, Guérin J, Dirner A, Neuzillet C, Girard N, Borcoman E, Larbi Chérif L, Tresca P, Roufai DB, Dupain C, Scholl S, André F, Fernandez X, Filleron T, Kamal M, Le Tourneau C. Combining immunotherapy with an epidrug in squamous cell carcinomas of different locations: rationale and design of the PEVO basket trial. ESMO Open 2021; 6:100106. [PMID: 33865192 PMCID: PMC8066350 DOI: 10.1016/j.esmoop.2021.100106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/12/2021] [Accepted: 03/06/2021] [Indexed: 12/25/2022] Open
Abstract
Squamous cell carcinomas (SCCs) are among the most frequent solid tumors in humans. SCCs, related or not to the human papillomavirus, share common molecular features. Immunotherapies, and specifically immune checkpoint inhibitors, have been shown to improve overall survival in multiple cancer types, including SCCs. However, only a minority of patients experience a durable response with immunotherapy. Epigenetic modulation plays a major role in escaping tumor immunosurveillance and confers resistance to immune checkpoint inhibitors. Preclinical evidence suggests that modulating the epigenome might improve the efficacy of immunotherapy. We herein review the preclinical and the clinical rationale for combining immunotherapy with an epidrug, and detail the design of PEVOsq, a basket clinical trial combining pembrolizumab with vorinostat, a histone deacetylase inhibitor, in patients with SCCs of different locations. Sequential blood and tumor sampling will be collected in order to identify predictive and pharmacodynamics biomarkers of efficacy of the combination. We also present how clinical and biological data will be managed with the aim to enable the development of a prospective integrative platform to allow secure and controlled access to the project data as well as further exploitations.
Collapse
Affiliation(s)
- E de Guillebon
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France; Inserm U932 Research Unit - Immunite et cancer, Paris, France
| | | | - L Mazzarella
- Department of Experimental Oncology, European Institute of Oncology - IRCCS, Milan, Italy; Division of Innovative Therapies, European Institute of Oncology - IRCCS, Milan, Italy
| | - F Betsou
- Integrated Biobank of Luxembourg, Dudelange, Luxembourg
| | - P Stadler
- Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany
| | - I Peták
- Oncompass Medicine Ltd, Budapest, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, USA
| | - E Jeannot
- Department of Genetics, Institut Curie, Paris, France; Department of Pathology, Institut Curie, Paris, France
| | - L Chanas
- Data Direction, Institut Curie, Paris, France
| | - N Servant
- Inserm U900 Research Unit, Saint Cloud, France
| | - G Marret
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - B A Duso
- Department of Experimental Oncology, European Institute of Oncology - IRCCS, Milan, Italy
| | | | - K N Kornerup
- Integrated Biobank of Luxembourg, Dudelange, Luxembourg
| | - S H Bernhart
- Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany
| | - G Balogh
- Bioinformatics Group, Department of Computer, University of Leipzig, Leipzig, Germany
| | - R Dóczi
- Oncompass Medicine Ltd, Budapest, Hungary
| | - P Filotás
- Oncompass Medicine Ltd, Budapest, Hungary
| | - G Curigliano
- Division of Innovative Therapies, European Institute of Oncology - IRCCS, Milan, Italy; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; University of Milano, Milan, Italy
| | - I Bièche
- Department of Genetics, Institut Curie, Paris, France
| | - J Guérin
- Data Direction, Institut Curie, Paris, France
| | - A Dirner
- Oncompass Medicine Ltd, Budapest, Hungary
| | - C Neuzillet
- Department of Medical Oncology, Institut Curie, Paris, France; Paris-Saclay University, Paris, France
| | - N Girard
- Department of Medical Oncology, Institut Curie, Paris, France; Paris-Saclay University, Paris, France
| | - E Borcoman
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - L Larbi Chérif
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - P Tresca
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - D B Roufai
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - C Dupain
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - S Scholl
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - F André
- Department of Medical Oncology, Gustave Roussy, Villejuif; INSERM, Gustave Roussy Cancer Campus, UMR981, Villejuif; University of Paris-Sud, Orsay, France
| | - X Fernandez
- Data Direction, Institut Curie, Paris, France
| | - T Filleron
- Biostatistics Unit, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - M Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France.
| | - C Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France; Inserm U900 Research Unit, Saint Cloud, France; Paris-Saclay University, Paris, France.
| |
Collapse
|
217
|
Jiang Z, Li N, Zhu D, Ren L, Shao Q, Yu K, Yang G. Genetically modified cell sheets in regenerative medicine and tissue engineering. Biomaterials 2021; 275:120908. [PMID: 34119885 DOI: 10.1016/j.biomaterials.2021.120908] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/16/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
Genetically modified cell sheet technology is emerging as a promising biomedical tool to deliver therapeutic genes for regenerative medicine and tissue engineering. Virus-based gene transfection and non-viral gene transfection have been used to fabricate genetically modified cell sheets. Preclinical and clinical studies have shown various beneficial effects of genetically modified cell sheets in the regeneration of bone, periodontal tissue, cartilage and nerves, as well as the amelioration of dental implant osseointegration, myocardial infarction, skeletal muscle ischemia and kidney injury. Furthermore, this technology provides a potential treatment option for various hereditary diseases. However, the method has several limitations, such as safety concerns and difficulties in controlling transgene expression. Therefore, recent studies explored efficient and safe gene transfection methods, prolonged and controllable transgene expression and their potential application in personalized and precision medicine. This review summarizes various types of genetically modified cell sheets, preparation procedures, therapeutic applications and possible improvements.
Collapse
Affiliation(s)
- Zhiwei Jiang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Na Li
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Danji Zhu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Lingfei Ren
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Qin Shao
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Ke Yu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Guoli Yang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
218
|
Seet AOL, Tan AC, Tan TJ, Ng MCH, Tai DWM, Lam JYC, Tan GS, Gogna A, Too CW, Tan BS, Takano A, Lim A, Lim TH, Lim ST, Dent RA, Ang MK, Yap YS, Tan IBH, Choo SP, Toh CK, Lim EH, Farid M, Skanderup AJ, Iyer NG, Lim WT, Tan EH, Lim TKH, Tan DSW. Individualized Molecular Profiling for Allocation to Clinical Trials Singapore Study-An Asian Tertiary Cancer Center Experience. JCO Precis Oncol 2021; 5:PO.20.00261. [PMID: 34250396 DOI: 10.1200/po.20.00261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/02/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Precision oncology has transformed the management of advanced cancers through implementation of advanced molecular profiling technologies to identify increasingly defined subsets of patients and match them to appropriate therapy. We report outcomes of a prospective molecular profiling study in a high-volume Asian tertiary cancer center. PATIENTS AND METHODS Patients with advanced cancer were enrolled onto a prospective protocol for genomic profiling, the Individualized Molecular Profiling for Allocation to Clinical Trials Singapore study, at the National Cancer Center Singapore. Primary objective was to identify molecular biomarkers in patient's tumors for allocation to clinical trials. The study commenced in February 2012 and is ongoing, with the results of all patients who underwent multiplex next-generation sequencing (NGS) testing until December 2018 presented here. The results were discussed at a molecular tumor board where recommendations for allocation to biomarker-directed trials or targeted therapies were made. RESULTS One thousand fifteen patients were enrolled with a median age of 58 years (range 20-83 years). Most common tumor types were lung adenocarcinoma (26%), colorectal cancer (15%), and breast cancer (12%). A total of 1,064 NGS assays were performed, on fresh tumor tissue for 369 (35%) and archival tumor tissue for 687 (65%) assays. TP53 (39%) alterations were most common, followed by EGFR (21%), KRAS (14%), and PIK3CA (10%). Of 405 NGS assays with potentially actionable alterations, 111 (27%) were allocated to a clinical trial after molecular tumor board and 20 (4.9%) were enrolled on a molecularly matched clinical trial. Gene fusions were detected in 23 of 311 (7%) patients tested, including rare fusions in new tumor types and known fusions in rare tumors. CONCLUSION Individualized Molecular Profiling for Allocation to Clinical Trials Singapore demonstrates the feasibility of a prospective broad molecular profiling program in an Asian tertiary cancer center, with the ability to develop and adapt to a dynamic landscape of precision oncology.
Collapse
Affiliation(s)
- Amanda O L Seet
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Aaron C Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Tira J Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Matthew C H Ng
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - David W M Tai
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Justina Y C Lam
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Gek San Tan
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Apoorva Gogna
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Chow Wei Too
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Bien Soo Tan
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Angela Takano
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Alvin Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Tse Hui Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Soon Thye Lim
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | | | - Mei Kim Ang
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Yoon-Sim Yap
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Iain B H Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Chee Keong Toh
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Elaine H Lim
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Mohamad Farid
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | | | - N Gopalakrishna Iyer
- Division of Surgical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Wan Teck Lim
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Eng Huat Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Tony K H Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
219
|
Bertucci F, Gonçalves A, Guille A, Adelaïde J, Garnier S, Carbuccia N, Billon E, Finetti P, Sfumato P, Monneur A, Pécheux C, Khran M, Brunelle S, Mescam L, Thomassin-Piana J, Poizat F, Charafe-Jauffret E, Turrini O, Lambaudie E, Provansal M, Extra JM, Madroszyk A, Gilabert M, Sabatier R, Vicier C, Mamessier E, Chabannon C, Pakradouni J, Viens P, André F, Gravis G, Popovici C, Birnbaum D, Chaffanet M. Prospective high-throughput genome profiling of advanced cancers: results of the PERMED-01 clinical trial. Genome Med 2021; 13:87. [PMID: 34006291 PMCID: PMC8132379 DOI: 10.1186/s13073-021-00897-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The benefit of precision medicine based on relatively limited gene sets and often-archived samples remains unproven. PERMED-01 (NCT02342158) was a prospective monocentric clinical trial assessing, in adults with advanced solid cancer, the feasibility and impact of extensive molecular profiling applied to newly biopsied tumor sample and based on targeted NGS (t-NGS) of the largest gene panel to date and whole-genome array-comparative genomic hybridization (aCGH) with assessment of single-gene alterations and clinically relevant genomic scores. METHODS Eligible patients with refractory cancer had one tumor lesion accessible to biopsy. Extracted tumor DNA was profiled by t-NGS and aCGH. We assessed alterations of 802 "candidate cancer" genes and global genomic scores, such as homologous recombination deficiency (HRD) score and tumor mutational burden. The primary endpoint was the number of patients with actionable genetic alterations (AGAs). Secondary endpoints herein reported included a description of patients with AGA who received a "matched therapy" and their clinical outcome, and a comparison of AGA identification with t-NGS and aCGH versus whole-exome sequencing (WES). RESULTS Between November 2014 and September 2019, we enrolled 550 patients heavily pretreated. An exploitable complete molecular profile was obtained in 441/550 patients (80%). At least one AGA, defined in real time by our molecular tumor board, was found in 393/550 patients (71%, two-sided 90%CI 68-75%). Only 94/550 patients (17%, 95%CI 14-21) received an "AGA-matched therapy" on progression. The most frequent AGAs leading to "matched therapy" included PIK3CA mutations, KRAS mutations/amplifications, PTEN deletions/mutations, ERBB2 amplifications/mutations, and BRCA1/2 mutations. Such "matched therapy" improved by at least 1.3-fold the progression-free survival on matched therapy (PFS2) compared to PFS on prior therapy (PFS1) in 36% of cases, representing 6% of the enrolled patients. Within patients with AGA treated on progression, the use of "matched therapy" was the sole variable associated with an improved PFS2/PFS1 ratio. Objective responses were observed in 19% of patients treated with "matched therapy," and 6-month overall survival (OS) was 62% (95%CI 52-73). In a subset of 112 metastatic breast cancers, WES did not provide benefit in term of AGA identification when compared with t-NGS/aCGH. CONCLUSIONS Extensive molecular profiling of a newly biopsied tumor sample identified AGA in most of cases, leading to delivery of a "matched therapy" in 17% of screened patients, of which 36% derived clinical benefit. WES did not seem to improve these results. TRIAL REGISTRATION ID-RCB identifier: 2014-A00966-41; ClinicalTrials.gov identifier: NCT02342158 .
Collapse
Affiliation(s)
- François Bertucci
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France.
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France.
| | - Anthony Gonçalves
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Arnaud Guille
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France
| | - José Adelaïde
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France
| | - Séverine Garnier
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France
| | - Nadine Carbuccia
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France
| | - Emilien Billon
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Pascal Finetti
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France
| | - Patrick Sfumato
- Biostatistics Unit, Institut Paoli-Calmettes, Marseille, France
| | - Audrey Monneur
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Christophe Pécheux
- Department of Medical genetics, Hôpital Timone Enfants, AP-HM, Marseille, France
| | - Martin Khran
- Department of Medical genetics, Hôpital Timone Enfants, AP-HM, Marseille, France
- Aix-Marseille University, Inserm, U1251-MMG, Marseille Medical Genetics, Marseille, France
| | - Serge Brunelle
- Department of Imaging, Institut Paoli-Calmettes, Marseille, France
| | - Lenaïg Mescam
- Department of Biopathology, Institut Paoli-Calmettes, Marseille, France
| | | | - Flora Poizat
- Department of Biopathology, Institut Paoli-Calmettes, Marseille, France
| | | | - Olivier Turrini
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Eric Lambaudie
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Magali Provansal
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Jean-Marc Extra
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Anne Madroszyk
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Marine Gilabert
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Renaud Sabatier
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Cécile Vicier
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Emilie Mamessier
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France
| | - Christian Chabannon
- Biobank, Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Jihane Pakradouni
- Department of Clinical Research and Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Patrice Viens
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Fabrice André
- Department of Medical Oncology, Gustave Roussy Cancer Campus, UMR981 Inserm, Villejuif, France
- Paris Sud University, Orsay, France
| | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Cornel Popovici
- Department of Oncogenetics, Institut Paoli-Calmettes, Marseille, France
| | - Daniel Birnbaum
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France
| | - Max Chaffanet
- Laboratory of Predictive Oncology, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Aix-Marseille University, 232 Boulevard Sainte-Marguerite, 13009, Marseille, France
| |
Collapse
|
220
|
Kikuchi J, Ohhara Y, Takada K, Tanabe H, Hatanaka K, Amano T, C Hatanaka K, Hatanaka Y, Mitamura T, Kato M, Shibata Y, Yabe I, Endoh A, Komatsu Y, Matsuno Y, Sugiyama M, Manabe A, Sakurai A, Takahashi M, Naruse H, Torimoto Y, Dosaka-Akita H, Kinoshita I. Clinical significance of comprehensive genomic profiling tests covered by public insurance in patients with advanced solid cancers in Hokkaido, Japan. Jpn J Clin Oncol 2021; 51:753-761. [PMID: 33532831 DOI: 10.1093/jjco/hyaa277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/30/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Comprehensive cancer genomic profiling has been used recently for patients with advanced solid cancers. Two cancer genomic profiling tests for patients with no standard treatment are covered by Japanese public health insurance since June 2019. METHODS We prospectively analyzed data of 189 patients with solid cancers who underwent either of the two-cancer genomic profiling tests at Hokkaido University Hospital and its liaison hospitals and whose results were discussed in molecular tumor board at Hokkaido University Hospital between August 2019 and July 2020. RESULTS All 189 patients had appropriate results. Actionable gene alterations were identified in 93 patients (49%). Frequent mutations included PIK3CA (12%) mutation, BRCA1/2 alteration (7%), ERBB2 amplification (6%) and tumor mutation burden-High (4%). The median turnaround time from sample shipping to acquisition by the expert panel was 26 days. Although 115 patients (61%) were provided with information for genotype-matched therapies, only 21 (11%) received them. Notably, four of eight patients below the age of 20 years were provided information for genotype-matched therapies, and three received them. Their response rates and disease control rates were 29% and 67%, respectively. Most patients who did not undergo the genotype-matched therapies were provided information for only investigational drugs in phases I and II at distant clinical trial sites in central Japan. Twenty-six patients were informed of suspected germline findings, while 11 patients (42%) received genetic counseling. CONCLUSIONS The publicly reimbursed cancer genomic profilings may lead to the modest but favorable therapeutic efficacy of genotype-matched therapy for solid cancer patients with no standard therapy. However, poor access to genotype-matched therapy needs to be resolved.
Collapse
Affiliation(s)
- Junko Kikuchi
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshihito Ohhara
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Japan
| | - Kohichi Takada
- Department of Medical Oncology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Hiroki Tanabe
- Genetic Oncology Department, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Kazuteru Hatanaka
- Department of Gastroenterology, Hakodate Municipal Hospital, Hakodate, Japan
| | - Toraji Amano
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Japan
| | - Kanako C Hatanaka
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan.,Clinical Biobank, Clinical Research and Medical Innovation Center, Hokkaido University Hospital, Sapporo, Japan
| | - Yutaka Hatanaka
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan.,Clinical Biobank, Clinical Research and Medical Innovation Center, Hokkaido University Hospital, Sapporo, Japan
| | - Takashi Mitamura
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Momoko Kato
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Yuka Shibata
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Ichiro Yabe
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Akira Endoh
- Division of Medical Information Planning, Hokkaido University Hospital, Sapporo, Japan
| | - Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital, Cancer Center, Sapporo, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Minako Sugiyama
- Department of Pediatrics, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Sakurai
- Department of Medical Genetics and Genomics, Sapporo Medical University, Sapporo, Japan
| | - Masato Takahashi
- Department of Cancer Genome Medical Center, NHO Hokkaido Cancer Center, Sapporo, Japan
| | - Hirohito Naruse
- Department of Gastroenterology, Hakodate Municipal Hospital, Hakodate, Japan
| | - Yoshihiro Torimoto
- Genetic Oncology Department, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Hirotoshi Dosaka-Akita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Japan
| | - Ichiro Kinoshita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
221
|
Kato S, Adashek JJ, Shaya J, Okamura R, Jimenez RE, Lee S, Sicklick JK, Kurzrock R. Concomitant MEK and Cyclin Gene Alterations: Implications for Response to Targeted Therapeutics. Clin Cancer Res 2021; 27:2792-2797. [PMID: 33472910 PMCID: PMC11005753 DOI: 10.1158/1078-0432.ccr-20-3761] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/07/2020] [Accepted: 01/13/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Cyclin and MAPK/MEK-related gene alterations are implicated in cell-cycle progression and cancer growth. Yet, monotherapy to target the cyclin (CDK4/6) or the MEK pathway has often yielded disappointing results. Because coalterations in cyclin and MEK pathway genes frequently cooccur, we hypothesized that resistance to CDK4/6 or MEK inhibitor monotherapy might be mediated via activation of oncogenic codrivers, and that combination therapy might be useful. EXPERIMENTAL DESIGN Herein, we describe 9 patients with advanced malignancies harboring concomitant CDKN2A and/or CDKN2B alterations (upregulate CDK4/6) along with KRAS or BRAF alterations (activate the MEK pathway) who were treated with palbociclib (CDK4/6 inhibitor) and trametinib (MEK inhibitor) combination-based regimens. RESULTS Two patients (with pancreatic cancer) achieved a partial remission (PR) and, overall, 5 patients (56%) had clinical benefit (stable disease ≥ 6 months/PR) with progression-free survival of approximately 7, 9, 9, 11, and 17.5+ months. Interestingly, 1 of these patients whose cancer (gastrointestinal stromal tumor) had progressed on MEK targeting regimen, did well for about 1 year after palbociclib was added. CONCLUSIONS These observations suggest that cotargeting cyclin and MEK signaling can be successful when tumors bear genomic coalterations that activate both of these pathways. Further prospective studies using this matching precision strategy to overcome resistance are warranted.See related commentary by Groisberg and Subbiah, p. 2672.
Collapse
Affiliation(s)
- Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California.
| | - Jacob J Adashek
- Department of Internal Medicine, University of South Florida, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Justin Shaya
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Ryosuke Okamura
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Rebecca E Jimenez
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Jason K Sicklick
- Division of Surgical Oncology, Department of Surgery, and Center for Personalized Cancer Therapy, University of California, San Diego, La Jolla, California
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| |
Collapse
|
222
|
Lazar V, Magidi S, Girard N, Savignoni A, Martini JF, Massimini G, Bresson C, Berger R, Onn A, Raynaud J, Wunder F, Berindan-Neagoe I, Sekacheva M, Braña I, Tabernero J, Felip E, Porgador A, Kleinman C, Batist G, Solomon B, Tsimberidou AM, Soria JC, Rubin E, Kurzrock R, Schilsky RL. Digital Display Precision Predictor: the prototype of a global biomarker model to guide treatments with targeted therapy and predict progression-free survival. NPJ Precis Oncol 2021; 5:33. [PMID: 33911192 PMCID: PMC8080819 DOI: 10.1038/s41698-021-00171-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 03/26/2021] [Indexed: 12/28/2022] Open
Abstract
The expanding targeted therapy landscape requires combinatorial biomarkers for patient stratification and treatment selection. This requires simultaneous exploration of multiple genes of relevant networks to account for the complexity of mechanisms that govern drug sensitivity and predict clinical outcomes. We present the algorithm, Digital Display Precision Predictor (DDPP), aiming to identify transcriptomic predictors of treatment outcome. For example, 17 and 13 key genes were derived from the literature by their association with MTOR and angiogenesis pathways, respectively, and their expression in tumor versus normal tissues was associated with the progression-free survival (PFS) of patients treated with everolimus or axitinib (respectively) using DDPP. A specific eight-gene set best correlated with PFS in six patients treated with everolimus: AKT2, TSC1, FKB-12, TSC2, RPTOR, RHEB, PIK3CA, and PIK3CB (r = 0.99, p = 5.67E-05). A two-gene set best correlated with PFS in five patients treated with axitinib: KIT and KITLG (r = 0.99, p = 4.68E-04). Leave-one-out experiments demonstrated significant concordance between observed and DDPP-predicted PFS (r = 0.9, p = 0.015) for patients treated with everolimus. Notwithstanding the small cohort and pending further prospective validation, the prototype of DDPP offers the potential to transform patients' treatment selection with a tumor- and treatment-agnostic predictor of outcomes (duration of PFS).
Collapse
Affiliation(s)
- Vladimir Lazar
- Worldwide Innovative Network (WIN) Association - WIN Consortium, Villejuif, France.
| | - Shai Magidi
- Worldwide Innovative Network (WIN) Association - WIN Consortium, Villejuif, France
| | | | | | | | | | - Catherine Bresson
- Worldwide Innovative Network (WIN) Association - WIN Consortium, Villejuif, France
| | | | - Amir Onn
- Sheba Medical Center, Tel-Hashomer, Israel
| | | | - Fanny Wunder
- Worldwide Innovative Network (WIN) Association - WIN Consortium, Villejuif, France
| | - Ioana Berindan-Neagoe
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| | - Marina Sekacheva
- I.M Sechenov First Medical State University, Moscow, Russian Federation
| | - Irene Braña
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Josep Tabernero
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Enriqueta Felip
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | | | - Claudia Kleinman
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, and NCE Exactis Innovations, Montreal, QC, Canada
| | - Gerald Batist
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, and NCE Exactis Innovations, Montreal, QC, Canada
| | | | | | | | - Eitan Rubin
- Ben-Gurion University of the Negev, Beer-Sheeva, Israel
| | - Razelle Kurzrock
- University of California San Diego, Moores Cancer Center, San Diego, CA, USA
| | | |
Collapse
|
223
|
Freidlin B, Allegra CJ, Korn EL. Moving Molecular Profiling to Routine Clinical Practice: A Way Forward? J Natl Cancer Inst 2021; 112:773-778. [PMID: 31868907 DOI: 10.1093/jnci/djz240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/09/2019] [Accepted: 12/18/2019] [Indexed: 01/09/2023] Open
Abstract
Molecular profiling of a patient's tumor to guide targeted treatment selection offers the potential to advance patient care by improving outcomes and minimizing toxicity (by avoiding ineffective treatments). However, current development of molecular profile (MP) panels is often based on applying institution-specific or subjective algorithms to nonrandomized patient cohorts. Consequently, obtaining reliable evidence that molecular profiling is offering clinical benefit and is ready for routine clinical practice is challenging. In particular, we discuss here the problems with interpreting for clinical utility nonrandomized studies that compare outcomes in patients treated based on their MP vs those treated with standard of care, studies that compare the progression-free survival (PFS) seen on a MP-directed treatment to the PFS seen for the same patient on a previous standard treatment (PFS ratio), and multibasket trials that evaluate the response rates of targeted therapies in specific molecularly defined subpopulations (regardless of histology). We also consider some limitations of randomized trial designs. A two-step strategy is proposed in which multiple mutation-agent pairs are tested for activity in one or more multibasket trials in the first step. The results of the first step are then used to identify promising mutation-agent pairs that are combined in a molecular panel that is then tested in the step-two strategy-design randomized clinical trial (the molecular panel-guided treatment for the selected mutations vs standard of care). This two-step strategy should allow rigorous evidence-driven identification of mutation-agent pairs that can be moved into routine clinical practice.
Collapse
Affiliation(s)
- Boris Freidlin
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr, Bethesda, MD 20892, USA
| | - Carmen J Allegra
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr, Bethesda, MD 20892, USA.,Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32608, USA
| | - Edward L Korn
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr, Bethesda, MD 20892, USA
| |
Collapse
|
224
|
Li Y, Zheng Y, Wu L, Li J, Ji J, Yu Q, Dai W, Feng J, Wu J, Guo C. Current status of ctDNA in precision oncology for hepatocellular carcinoma. J Exp Clin Cancer Res 2021; 40:140. [PMID: 33902698 PMCID: PMC8074474 DOI: 10.1186/s13046-021-01940-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/06/2021] [Indexed: 01/12/2023] Open
Abstract
The conventional method used to obtain a tumor biopsy for hepatocellular carcinoma (HCC) is invasive and does not evaluate dynamic cancer progression or assess tumor heterogeneity. It is thus imperative to create a novel non-invasive diagnostic technique for improvement in cancer screening, diagnosis, treatment selection, response assessment, and predicting prognosis for HCC. Circulating tumor DNA (ctDNA) is a non-invasive liquid biopsy method that reveals cancer-specific genetic and epigenetic aberrations. Owing to the development of technology in next-generation sequencing and PCR-based assays, the detection and quantification of ctDNA have greatly improved. In this publication, we provide an overview of current technologies used to detect ctDNA, the ctDNA markers utilized, and recent advances regarding the multiple clinical applications in the field of precision medicine for HCC.
Collapse
Affiliation(s)
- Yan Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
| |
Collapse
|
225
|
Hlevnjak M, Schulze M, Elgaafary S, Fremd C, Michel L, Beck K, Pfütze K, Richter D, Wolf S, Horak P, Kreutzfeldt S, Pixberg C, Hutter B, Ishaque N, Hirsch S, Gieldon L, Stenzinger A, Springfeld C, Smetanay K, Seitz J, Mavratzas A, Brors B, Kirsten R, Schuetz F, Fröhling S, Sinn HP, Jäger D, Thewes V, Zapatka M, Lichter P, Schneeweiss A. CATCH: A Prospective Precision Oncology Trial in Metastatic Breast Cancer. JCO Precis Oncol 2021; 5:PO.20.00248. [PMID: 34036222 PMCID: PMC8140780 DOI: 10.1200/po.20.00248] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 01/13/2021] [Accepted: 03/10/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE CATCH (Comprehensive Assessment of clinical feaTures and biomarkers to identify patients with advanced or metastatic breast Cancer for marker driven trials in Humans) is a prospective precision oncology program that uses genomics and transcriptomics to guide therapeutic decisions in the clinical management of metastatic breast cancer. Herein, we report our single-center experience and results on the basis of the first 200 enrolled patients of an ongoing trial. METHODS From June 2017 to March 2019, 200 patients who had either primary metastatic or progressive disease, with any number of previous treatment lines and at least one metastatic site accessible to biopsy, were enrolled. DNA and RNA from tumor tissue and corresponding blood-derived nontumor DNA were profiled using whole-genome and transcriptome sequencing. Identified actionable alterations were brought into clinical context in a multidisciplinary molecular tumor board (MTB) with the aim of prioritizing personalized treatment recommendations. RESULTS Among the first 200 enrolled patients, 128 (64%) were discussed in the MTB, of which 64 (50%) were subsequently treated according to MTB recommendation. Of 53 evaluable patients, 21 (40%) achieved either stable disease (n = 13, 25%) or partial response (n = 8, 15%). Furthermore, 16 (30%) of those patients showed improvement in progression-free survival of at least 30% while on MTB-recommended treatment compared with the progression-free survival of the previous treatment line. CONCLUSION The initial phase of this study demonstrates that precision oncology on the basis of whole-genome and RNA sequencing is feasible when applied in the clinical management of patients with metastatic breast cancer and provides clinical benefit to a substantial proportion of patients.
Collapse
Affiliation(s)
- Mario Hlevnjak
- Division of Molecular Genetics, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Markus Schulze
- Division of Molecular Genetics, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Shaymaa Elgaafary
- Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carlo Fremd
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Michel
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katja Beck
- Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katrin Pfütze
- Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Daniela Richter
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Wolf
- Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Horak
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simon Kreutzfeldt
- Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Constantin Pixberg
- Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Hutter
- Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.,Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Naveed Ishaque
- Heidelberg Center for Personalized Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steffen Hirsch
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | - Laura Gieldon
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Katharina Smetanay
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia Seitz
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Athanasios Mavratzas
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Romy Kirsten
- National Center for Tumor Diseases (NCT), Liquid Biobank, Heidelberg, Germany
| | - Florian Schuetz
- Department of Gynecology and Obstetrics, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Peter Sinn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Verena Thewes
- Division of Molecular Genetics, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc Zapatka
- Division of Molecular Genetics, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Center for Personalized Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Schneeweiss
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
226
|
Sultova E, Westphalen CB, Jung A, Kumbrink J, Kirchner T, Mayr D, Rudelius M, Ormanns S, Heinemann V, Metzeler KH, Greif PA, Hester A, Mahner S, Harbeck N, Wuerstlein R. Implementation of Precision Oncology for Patients with Metastatic Breast Cancer in an Interdisciplinary MTB Setting. Diagnostics (Basel) 2021; 11:733. [PMID: 33924134 PMCID: PMC8074310 DOI: 10.3390/diagnostics11040733] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
The advent of molecular diagnostics and the rising number of targeted therapies have facilitated development of precision oncology for cancer patients. In order to demonstrate its impact for patients with metastatic breast cancer (mBC), we initiated a Molecular Tumor Board (MTB) to provide treatment recommendations for mBC patients who had disease progression under standard treatment. NGS (next generation sequencing) was carried out using the Oncomine multi-gene panel testing system (Ion Torrent). The MTB reviewed molecular diagnostics' results, relevant tumor characteristics, patient's course of disease and made personalized treatment and/or diagnostic recommendations for each patient. From May 2017 to December 2019, 100 mBC patients were discussed by the local MTB. A total 72% of the mBC tumors had at least one molecular alteration (median 2 per case, range: 1 to 6). The most frequent genetic changes were found in the following genes: PIK3CA (19%) and TP53 (17%). The MTB rated 53% of these alterations as actionable and treatment recommendations were made accordingly for 49 (49%) patients. Sixteen patients (16%) underwent the suggested therapy. Nine out of sixteen patients (56%; 9% of all) experienced a clinical benefit with a progression-free survival ratio ≥ 1.3. Personalized targeted therapy recommendations resulting from MTB case discussions could provide substantial benefits for patients with mBC and should be implemented for all suitable patients.
Collapse
Affiliation(s)
- Elena Sultova
- Department of Obstetrics and Gynecology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (E.S.); (A.H.); (S.M.); (N.H.)
| | - C. Benedikt Westphalen
- Department of Internal Medicine III and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (C.B.W.); (V.H.); (K.H.M.); (P.A.G.)
| | - Andreas Jung
- Institute of Pathology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (A.J.); (J.K.); (T.K.); (D.M.); (M.R.); (S.O.)
| | - Joerg Kumbrink
- Institute of Pathology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (A.J.); (J.K.); (T.K.); (D.M.); (M.R.); (S.O.)
| | - Thomas Kirchner
- Institute of Pathology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (A.J.); (J.K.); (T.K.); (D.M.); (M.R.); (S.O.)
| | - Doris Mayr
- Institute of Pathology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (A.J.); (J.K.); (T.K.); (D.M.); (M.R.); (S.O.)
| | - Martina Rudelius
- Institute of Pathology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (A.J.); (J.K.); (T.K.); (D.M.); (M.R.); (S.O.)
| | - Steffen Ormanns
- Institute of Pathology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (A.J.); (J.K.); (T.K.); (D.M.); (M.R.); (S.O.)
| | - Volker Heinemann
- Department of Internal Medicine III and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (C.B.W.); (V.H.); (K.H.M.); (P.A.G.)
| | - Klaus H. Metzeler
- Department of Internal Medicine III and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (C.B.W.); (V.H.); (K.H.M.); (P.A.G.)
| | - Philipp A. Greif
- Department of Internal Medicine III and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (C.B.W.); (V.H.); (K.H.M.); (P.A.G.)
| | - Anna Hester
- Department of Obstetrics and Gynecology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (E.S.); (A.H.); (S.M.); (N.H.)
| | - Sven Mahner
- Department of Obstetrics and Gynecology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (E.S.); (A.H.); (S.M.); (N.H.)
- Gynecologic Oncology Center and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
| | - Nadia Harbeck
- Department of Obstetrics and Gynecology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (E.S.); (A.H.); (S.M.); (N.H.)
- Breast Center and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
| | - Rachel Wuerstlein
- Department of Obstetrics and Gynecology and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany; (E.S.); (A.H.); (S.M.); (N.H.)
- Gynecologic Oncology Center and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
- Breast Center and CCC Munich LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
| |
Collapse
|
227
|
Pfohl U, Pflaume A, Regenbrecht M, Finkler S, Graf Adelmann Q, Reinhard C, Regenbrecht CRA, Wedeken L. Precision Oncology Beyond Genomics: The Future Is Here-It Is Just Not Evenly Distributed. Cells 2021; 10:928. [PMID: 33920536 PMCID: PMC8072767 DOI: 10.3390/cells10040928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is a multifactorial disease with increasing incidence. There are more than 100 different cancer types, defined by location, cell of origin, and genomic alterations that influence oncogenesis and therapeutic response. This heterogeneity between tumors of different patients and also the heterogeneity within the same patient's tumor pose an enormous challenge to cancer treatment. In this review, we explore tumor heterogeneity on the longitudinal and the latitudinal axis, reviewing current and future approaches to study this heterogeneity and their potential to support oncologists in tailoring a patient's treatment regimen. We highlight how the ideal of precision oncology is reaching far beyond the knowledge of genetic variants to inform clinical practice and discuss the technologies and strategies already available to improve our understanding and management of heterogeneity in cancer treatment. We will focus on integrating multi-omics technologies with suitable in vitro models and their proficiency in mimicking endogenous tumor heterogeneity.
Collapse
Affiliation(s)
- Ulrike Pfohl
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
- Institut für Molekulare Biowissenschaften, Goethe Universität Frankfurt am Main, Theodor-W.-Adorno-Platz 1, 60323 Frankfurt am Main, Germany
| | - Alina Pflaume
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Manuela Regenbrecht
- Helios Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125 Berlin, Germany;
| | - Sabine Finkler
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Quirin Graf Adelmann
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Christoph Reinhard
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Christian R. A. Regenbrecht
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
- Institut für Pathologie, Universitätsklinikum Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Lena Wedeken
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| |
Collapse
|
228
|
Wenzel C, Herold S, Wermke M, E. Aust D, B. Baretton G. Routine Molecular Pathology Diagnostics in Precision Oncology. DEUTSCHES ARZTEBLATT INTERNATIONAL 2021; 118:arztebl.m2021.0025. [PMID: 33536117 PMCID: PMC8287073 DOI: 10.3238/arztebl.m2021.0025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 12/01/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Technical advances in the field of molecular genetics permit precise genomic characterization of malignant tumors. This has not only improved our understanding of tumor biology but also paved the way for molecularly stratified treatment strategies in routine clinical practice. METHODS A selective search of PubMed to identify literature on molecular pathology methods, their indications, the challenges associated with molecular findings, and future developments. RESULTS Tumors can be characterized with the aid of immunohistochemistry, in-situ hybridization, and sequencing of DNA or RNA. The benefits of molecularly stratified tumor treatment have been demonstrated by randomized clinical trials on numerous tumor entities, e.g., non-small-cell lung cancer, colorectal cancer, and breast cancer. Therefore, initiation of specific treatment for these entities should be preceded by molecular pathology biomarker analyses, generally carried out on tumor tissue. Randomized controlled trials and non-controlled studies show that enhanced progression-free survival ensues if the pharmacological treatment is oriented on the findings of molecular pathology diagnostics. In next-generation sequencing, numerous relevant gene sequences or even whole genes can be sequenced in parallel, dispensing with complex staged diagnostics and reducing the use of biomaterials. These new methods also complement the currently relevant predictive biomarkers by permitting the investigation of genetic alterations presently of interest in the context of clinical studies. Prior to widespread routine clinical application, however, sequencing of large gene panels or whole genomes or exomes need to be even more stringently validated. CONCLUSION Quality-assured molecular pathology assays are universally available for the determination of currently relevant predictive biomarkers. However, the integration of extensive genomic analyses into routine molecular pathology diagnostics represents a future challenge in precision oncology.
Collapse
Affiliation(s)
- Carina Wenzel
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden
| | - Sylvia Herold
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden
| | - Martin Wermke
- Medical Department I, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden: Dr. med. Martin Wermke
| | - Daniela E. Aust
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden
| | - Gustavo B. Baretton
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden
| |
Collapse
|
229
|
Kuksin M, Morel D, Aglave M, Danlos FX, Marabelle A, Zinovyev A, Gautheret D, Verlingue L. Applications of single-cell and bulk RNA sequencing in onco-immunology. Eur J Cancer 2021; 149:193-210. [PMID: 33866228 DOI: 10.1016/j.ejca.2021.03.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 02/08/2023]
Abstract
The rising interest for precise characterization of the tumour immune contexture has recently brought forward the high potential of RNA sequencing (RNA-seq) in identifying molecular mechanisms engaged in the response to immunotherapy. In this review, we provide an overview of the major principles of single-cell and conventional (bulk) RNA-seq applied to onco-immunology. We describe standard preprocessing and statistical analyses of data obtained from such techniques and highlight some computational challenges relative to the sequencing of individual cells. We notably provide examples of gene expression analyses such as differential expression analysis, dimensionality reduction, clustering and enrichment analysis. Additionally, we used public data sets to exemplify how deconvolution algorithms can identify and quantify multiple immune subpopulations from either bulk or single-cell RNA-seq. We give examples of machine and deep learning models used to predict patient outcomes and treatment effect from high-dimensional data. Finally, we balance the strengths and weaknesses of single-cell and bulk RNA-seq regarding their applications in the clinic.
Collapse
Affiliation(s)
- Maria Kuksin
- ENS de Lyon, 15 Parvis René Descartes, 69007, Lyon, France; Département d'Innovations Thérapeutiques et Essais Précoces (DITEP), Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France
| | - Daphné Morel
- Département d'Innovations Thérapeutiques et Essais Précoces (DITEP), Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France; Département de Radiothérapie, Gustave Roussy Cancer Campus, Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France; INSERM UMR1030, Molecular Radiotherapy and Therapeutic Innovations, Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
| | - Marine Aglave
- INSERM US23, CNRS UMS 3655, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France
| | | | - Aurélien Marabelle
- Département d'Innovations Thérapeutiques et Essais Précoces (DITEP), Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France; INSERM U1015, Gustave Roussy, Université Paris Saclay, France
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, F-75005, Paris, France; INSERM, U900, F-75005, Paris, France; MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, F-75006, Paris, France; Laboratory of Advanced Methods for High-dimensional Data Analysis, Lobachevsky University, 603000, Nizhny Novgorod, Russia
| | - Daniel Gautheret
- Institute for Integrative Biology of the Cell, UMR 9198, CEA, CNRS, Université Paris-Saclay, Gif-Sur-Yvette, France; IHU PRISM, Gustave Roussy Cancer Campus, Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France; Université Paris-Saclay, France
| | - Loïc Verlingue
- Département d'Innovations Thérapeutiques et Essais Précoces (DITEP), Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94800, Villejuif, France; INSERM UMR1030, Molecular Radiotherapy and Therapeutic Innovations, Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France; Institut Curie, PSL Research University, F-75005, Paris, France; Université Paris-Saclay, France.
| |
Collapse
|
230
|
Synthetic lethality-mediated precision oncology via the tumor transcriptome. Cell 2021; 184:2487-2502.e13. [PMID: 33857424 DOI: 10.1016/j.cell.2021.03.030] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/29/2020] [Accepted: 03/12/2021] [Indexed: 01/27/2023]
Abstract
Precision oncology has made significant advances, mainly by targeting actionable mutations in cancer driver genes. Aiming to expand treatment opportunities, recent studies have begun to explore the utility of tumor transcriptome to guide patient treatment. Here, we introduce SELECT (synthetic lethality and rescue-mediated precision oncology via the transcriptome), a precision oncology framework harnessing genetic interactions to predict patient response to cancer therapy from the tumor transcriptome. SELECT is tested on a broad collection of 35 published targeted and immunotherapy clinical trials from 10 different cancer types. It is predictive of patients' response in 80% of these clinical trials and in the recent multi-arm WINTHER trial. The predictive signatures and the code are made publicly available for academic use, laying a basis for future prospective clinical studies.
Collapse
|
231
|
Yu Z, Bian C, Liu G, Zhang S, Wong KC, Li X. Elucidating transcriptomic profiles from single-cell RNA sequencing data using nature-inspired compressed sensing. Brief Bioinform 2021; 22:6225863. [PMID: 33855366 DOI: 10.1093/bib/bbab125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/07/2021] [Accepted: 03/16/2021] [Indexed: 11/12/2022] Open
Abstract
Gene-expression profiling can define the cell state and gene-expression pattern of cells at the genetic level in a high-throughput manner. With the development of transcriptome techniques, processing high-dimensional genetic data has become a major challenge in expression profiling. Thanks to the recent widespread use of matrix decomposition methods in bioinformatics, a computational framework based on compressed sensing was adopted to reduce dimensionality. However, compressed sensing requires an optimization strategy to learn the modular dictionaries and activity levels from the low-dimensional random composite measurements to reconstruct the high-dimensional gene-expression data. Considering this, here we introduce and compare four compressed sensing frameworks coming from nature-inspired optimization algorithms (CSCS, ABCCS, BACS and FACS) to improve the quality of the decompression process. Several experiments establish that the three proposed methods outperform benchmark methods on nine different datasets, especially the FACS method. We illustrate therefore, the robustness and convergence of FACS in various aspects; notably, time complexity and parameter analyses highlight properties of our proposed FACS. Furthermore, differential gene-expression analysis, cell-type clustering, gene ontology enrichment and pathology analysis are conducted, which bring novel insights into cell-type identification and characterization mechanisms from different perspectives. All algorithms are written in Python and available at https://github.com/Philyzh8/Nature-inspired-CS.
Collapse
Affiliation(s)
- Zhuohan Yu
- school of Artificial Intelligence, Jilin University, Jilin, China
| | - Chuang Bian
- school of Artificial Intelligence, Jilin University, Jilin, China
| | - Genggeng Liu
- Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, China
| | - Shixiong Zhang
- School of Computer Science and Technology, Xidian University, Xi'an, China 710000
| | | | - Xiangtao Li
- School of Artificial Intelligence, Jilin University, Jilin, China
| |
Collapse
|
232
|
Identifying potential germline variants from sequencing hematopoietic malignancies. Blood 2021; 136:2498-2506. [PMID: 33236764 DOI: 10.1182/blood.2020006910] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/23/2020] [Indexed: 12/12/2022] Open
Abstract
Next-generation sequencing (NGS) of bone marrow and peripheral blood increasingly guides clinical care in hematological malignancies. NGS data may help to identify single nucleotide variants, insertions/deletions, copy number variations, and translocations at a single time point, and repeated NGS testing allows tracking of dynamic changes in variants during the course of a patient's disease. Tumor cells used for NGS may contain germline, somatic, and clonal hematopoietic DNA alterations, and distinguishing the etiology of a variant may be challenging. We describe an approach using patient history, individual variant characteristics, and sequential NGS assays to identify potential germline variants. Our current criteria for identifying an individual likely to have a deleterious germline variant include a strong family history or multiple cancers in a single patient, diagnosis of a hematopoietic malignancy at a younger age than seen in the general population, variant allele frequency > 0.3 of a deleterious allele in a known germline predisposition gene, and variant persistence identified on clinical NGS panels, despite a change in disease state. Sequential molecular testing of hematopoietic specimens may provide insight into disease pathology, impact patient and family members' care, and potentially identify new cancer-predisposing risk alleles. Ideally, individuals should give consent at the time of NGS testing to receive information about potential germline variants and to allow future contact as research advances.
Collapse
|
233
|
Rodriguez H, Zenklusen JC, Staudt LM, Doroshow JH, Lowy DR. The next horizon in precision oncology: Proteogenomics to inform cancer diagnosis and treatment. Cell 2021; 184:1661-1670. [PMID: 33798439 PMCID: PMC8459793 DOI: 10.1016/j.cell.2021.02.055] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/13/2021] [Accepted: 02/26/2021] [Indexed: 12/18/2022]
Abstract
When it comes to precision oncology, proteogenomics may provide better prospects to the clinical characterization of tumors, help make a more accurate diagnosis of cancer, and improve treatment for patients with cancer. This perspective describes the significant contributions of The Cancer Genome Atlas and the Clinical Proteomic Tumor Analysis Consortium to precision oncology and makes the case that proteogenomics needs to be fully integrated into clinical trials and patient care in order for precision oncology to deliver the right cancer treatment to the right patient at the right dose and at the right time.
Collapse
Affiliation(s)
- Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Jean Claude Zenklusen
- Center for Cancer Genomics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Louis M Staudt
- Center for Cancer Genomics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Office of the Director, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Douglas R Lowy
- Office of the Director, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
234
|
Ebata K, Yamashiro S, Iida K, Okada M. Building patient-specific models for receptor tyrosine kinase signaling networks. FEBS J 2021; 289:90-101. [PMID: 33755310 DOI: 10.1111/febs.15831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/26/2021] [Accepted: 03/19/2021] [Indexed: 12/16/2022]
Abstract
Cancer progresses due to changes in the dynamic interactions of multidimensional factors associated with gene mutations. Cancer research has actively adopted computational methods, including data-driven and mathematical model-driven approaches, to identify causative factors and regulatory rules that can explain the complexity and diversity of cancers. A data-driven, statistics-based approach revealed correlations between gene alterations and clinical outcomes in many types of cancers. A model-driven mathematical approach has elucidated the dynamic features of cancer networks and identified the mechanisms of drug efficacy and resistance. More recently, machine learning methods have emerged that can be used for mining omics data and classifying patient. However, as the strengths and weaknesses of each method becoming apparent, new analytical tools are emerging to combine and improve the methodologies and maximize their predictive power for classifying cancer subtypes and prognosis. Here, we introduce recent advances in cancer systems biology aimed at personalized medicine, with focus on the receptor tyrosine kinase signaling network.
Collapse
Affiliation(s)
- Kyoichi Ebata
- Institute for Protein Research, Osaka University, Suita, Japan
| | - Sawa Yamashiro
- Institute for Protein Research, Osaka University, Suita, Japan
| | - Keita Iida
- Institute for Protein Research, Osaka University, Suita, Japan
| | - Mariko Okada
- Institute for Protein Research, Osaka University, Suita, Japan.,Center for Drug Design and Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan.,Institute for Chemical Research, Kyoto University, Japan
| |
Collapse
|
235
|
Nikanjam M, Tinajero J, Barkauskas DA, Kurzrock R. BRAF V600E/V600K Mutations versus Nonstandard Alterations: Prognostic Implications and Therapeutic Outcomes. Mol Cancer Ther 2021; 20:1072-1079. [PMID: 33722853 DOI: 10.1158/1535-7163.mct-20-0861] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/05/2020] [Accepted: 02/24/2021] [Indexed: 11/16/2022]
Abstract
BRAF and MEK inhibitors are standard of care for BRAF V600E/K-mutated melanoma, but the benefit of BRAF and/or MEK inhibitors for nonstandard BRAF alterations for melanoma and other cancers is unclear. Patients with diverse malignancies whose cancers had undergone next-generation sequencing were screened for BRAF alterations. Demographics, treatment with BRAF and/or MEK inhibitors, clinical response, progression-free survival (PFS), and overall survival (OS) were determined from review of the electronic medical records for patients with standard BRAF V600E/K versus nonstandard BRAF alterations. A total of 213 patients with BRAF alterations (87 with nonstandard alterations) were identified; OS from diagnosis was significantly worse with nonstandard BRAF versus standard alterations, regardless of therapy [HR (95% confidence interval), 0.58 (0.38-0.88); P = 0.01]. Overall, 45 patients received BRAF/MEK-directed therapy (eight with nonstandard alterations); there were no significant differences in clinical benefit rate [stable disease ≥6 months/partial/complete response (74% vs. 63%; P = 0.39) or PFS (P = 0.24; BRAF V600E/K vs. others)]. In conclusion, patients with nonstandard versus standard BRAF alterations (BRAF V600E/K) have a worse prognosis with shorter survival from diagnosis. Even so, 63% of patients with nonstandard BRAF alterations achieved clinical benefit with BRAF/MEK inhibitors. Larger prospective studies are warranted to better understand the prognostic versus predictive implication of standard versus nonstandard BRAF alterations.
Collapse
Affiliation(s)
- Mina Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, California.
| | - Jose Tinajero
- Deparatment of Pharmacy, UC San Diego Health, San Diego, California
| | - Donald A Barkauskas
- Division of Biostatistics, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, California
| |
Collapse
|
236
|
Irmisch A, Bonilla X, Chevrier S, Lehmann KV, Singer F, Toussaint NC, Esposito C, Mena J, Milani ES, Casanova R, Stekhoven DJ, Wegmann R, Jacob F, Sobottka B, Goetze S, Kuipers J, Sarabia Del Castillo J, Prummer M, Tuncel MA, Menzel U, Jacobs A, Engler S, Sivapatham S, Frei AL, Gut G, Ficek J, Miglino N, Aebersold R, Bacac M, Beerenwinkel N, Beisel C, Bodenmiller B, Dummer R, Heinzelmann-Schwarz V, Koelzer VH, Manz MG, Moch H, Pelkmans L, Snijder B, Theocharides APA, Tolnay M, Wicki A, Wollscheid B, Rätsch G, Levesque MP. The Tumor Profiler Study: integrated, multi-omic, functional tumor profiling for clinical decision support. Cancer Cell 2021; 39:288-293. [PMID: 33482122 DOI: 10.1016/j.ccell.2021.01.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The application and integration of molecular profiling technologies create novel opportunities for personalized medicine. Here, we introduce the Tumor Profiler Study, an observational trial combining a prospective diagnostic approach to assess the relevance of in-depth tumor profiling to support clinical decision-making with an exploratory approach to improve the biological understanding of the disease.
Collapse
Affiliation(s)
- Anja Irmisch
- University Hospital Zurich, Department of Dermatology, University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Ximena Bonilla
- ETH Zurich, Department of Computer Science, Institute of Machine Learning, Universitätstrasse 6, 8092 Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Hospital Zurich, Biomedical Informatics, Schmelzbergstrasse 26, 8006 Zurich, Switzerland
| | - Stéphane Chevrier
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Kjong-Van Lehmann
- ETH Zurich, Department of Computer Science, Institute of Machine Learning, Universitätstrasse 6, 8092 Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Hospital Zurich, Biomedical Informatics, Schmelzbergstrasse 26, 8006 Zurich, Switzerland
| | - Franziska Singer
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, NEXUS Personalized Health Technologies, John-von-Neumann-Weg 9, 8093 Zurich, Switzerland
| | - Nora C Toussaint
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, NEXUS Personalized Health Technologies, John-von-Neumann-Weg 9, 8093 Zurich, Switzerland
| | - Cinzia Esposito
- University of Zurich, Department of Molecular Life Sciences, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Julien Mena
- ETH Zurich, Department of Biology, Institute of Molecular Systems Biology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Emanuela S Milani
- ETH Zurich, Department of Health Sciences and Technology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Ruben Casanova
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Daniel J Stekhoven
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, NEXUS Personalized Health Technologies, John-von-Neumann-Weg 9, 8093 Zurich, Switzerland
| | - Rebekka Wegmann
- ETH Zurich, Department of Biology, Institute of Molecular Systems Biology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Francis Jacob
- University Hospital Basel and University of Basel, Department of Biomedicine, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Bettina Sobottka
- University Hospital Zurich, Department of Pathology and Molecular Pathology, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Sandra Goetze
- ETH Zurich, Department of Health Sciences and Technology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Jack Kuipers
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Jacobo Sarabia Del Castillo
- University of Zurich, Department of Molecular Life Sciences, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Michael Prummer
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, NEXUS Personalized Health Technologies, John-von-Neumann-Weg 9, 8093 Zurich, Switzerland
| | - Mustafa A Tuncel
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Ulrike Menzel
- ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Andrea Jacobs
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Stefanie Engler
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Sujana Sivapatham
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Anja L Frei
- University Hospital Zurich, Department of Pathology and Molecular Pathology, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Gabriele Gut
- University of Zurich, Department of Molecular Life Sciences, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Joanna Ficek
- ETH Zurich, Department of Computer Science, Institute of Machine Learning, Universitätstrasse 6, 8092 Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Hospital Zurich, Biomedical Informatics, Schmelzbergstrasse 26, 8006 Zurich, Switzerland
| | - Nicola Miglino
- University Hospital Zurich, Department of Medical Oncology and Hematology, Rämistrasse 100, 8091 Zurich, Switzerland
| | | | - Rudolf Aebersold
- ETH Zurich, Department of Biology, Institute of Molecular Systems Biology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Marina Bacac
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Christian Beisel
- ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Bernd Bodenmiller
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland; ETH Zurich, Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Reinhard Dummer
- University Hospital Zurich, Department of Dermatology, University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Viola Heinzelmann-Schwarz
- University Hospital Basel and University of Basel, Department of Biomedicine, Hebelstrasse 20, 4031 Basel, Switzerland; University Hospital Basel, Gynecological Cancer Center, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Viktor H Koelzer
- University Hospital Zurich, Department of Pathology and Molecular Pathology, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Markus G Manz
- University Hospital Zurich, Department of Medical Oncology and Hematology, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Holger Moch
- University Hospital Zurich, Department of Pathology and Molecular Pathology, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Lucas Pelkmans
- University of Zurich, Department of Molecular Life Sciences, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Berend Snijder
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, Department of Biology, Institute of Molecular Systems Biology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Alexandre P A Theocharides
- University Hospital Zurich, Department of Medical Oncology and Hematology, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Markus Tolnay
- University Hospital Basel, Institute of Medical Genetics and Pathology, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Andreas Wicki
- University Hospital Zurich, Department of Medical Oncology and Hematology, Rämistrasse 100, 8091 Zurich, Switzerland; University of Zurich, Faculty of Medicine, Zurich, Switzerland
| | - Bernd Wollscheid
- ETH Zurich, Department of Health Sciences and Technology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Gunnar Rätsch
- ETH Zurich, Department of Computer Science, Institute of Machine Learning, Universitätstrasse 6, 8092 Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Hospital Zurich, Biomedical Informatics, Schmelzbergstrasse 26, 8006 Zurich, Switzerland; ETH Zurich, Department of Biology, Wolfgang-Pauli-Strasse 27, 8093 Zurich, Switzerland.
| | - Mitchell P Levesque
- University Hospital Zurich, Department of Dermatology, University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland.
| |
Collapse
|
237
|
Krebs MG, Blay JY, Le Tourneau C, Hong D, Veronese L, Antoniou M, Bennett I. Intrapatient comparisons of efficacy in a single-arm trial of entrectinib in tumour-agnostic indications. ESMO Open 2021; 6:100072. [PMID: 33676294 PMCID: PMC8103537 DOI: 10.1016/j.esmoop.2021.100072] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/03/2021] [Accepted: 02/03/2021] [Indexed: 12/29/2022] Open
Abstract
Background Entrectinib is a tropomyosin receptor kinase inhibitor approved for the treatment of neurotrophic tyrosine receptor kinase (NTRK) fusion-positive solid tumours based on single-arm trials. Traditional randomised clinical trials in rare cancers are not feasible; we conducted an intrapatient analysis to evaluate the clinical benefit of entrectinib versus prior standard-of-care systemic therapies. Methods Patients with locally advanced/metastatic NTRK fusion-positive tumours enrolled in the global phase II, single-arm STARTRK-2 trial were grouped according to prior systemic therapy and response. The key analysis used growth modulation index [GMI; ratio of progression-free survival (PFS) on entrectinib to time to discontinuation (TTD) on the most recent prior therapy]; ratio ≥1.3 indicated clinically meaningful efficacy. Additional analyses investigated TTD and objective response rate (ORR) for entrectinib and prior therapies. Results Seventy-one patients were included; 51 received prior systemic therapy. In 38 patients who progressed on prior therapy, ORR was 60.5% (23/38) with entrectinib and 15.8% (6/38) with the most recent prior therapy. Median PFS [11.2 months; 95% confidence interval (CI) 6.7–not estimable] for entrectinib exceeded median TTD (2.9 months; 95% CI 2.0-4.9) for most recent prior therapy. From the intrapatient analysis of GMI, 65.8% had a ratio ≥1.3 and median GMI was 2.53. Consistent results were observed at more stringent GMI thresholds; 60.5% of patients had GMI ≥1.5 or ≥1.8 and 57.9% had GMI ≥2.0. Conclusions ORR was high and PFS was longer on entrectinib versus TTD on prior therapy. Furthermore, 65.8% of patients experienced clinically meaningful benefit based on GMI. This intrapatient analysis demonstrates comparative effectiveness of entrectinib in a rare, heterogeneous adult population. Randomised trials are unfeasible for molecular targeted agents in rare indications because of low patient numbers. Intrapatient comparison with prior therapies can be used to evaluate relative treatment efficacy in rare tumours. Entrectinib is a potent tropomyosin receptor kinase (TRK) inhibitor with proven efficacy in neurotrophic tyrosine receptor kinase (NTRK) fusion-positive tumours from the global STARTRK-2 trial. Median progression-free survival on entrectinib was longer than time to discontinuation on prior therapy (11.2 months versus 2.9 months). About 61% of patients with prior therapy progression responded to entrectinib; 66% had growth modulation index ≥1.3 (clinically meaningful threshold).
Collapse
Affiliation(s)
- M G Krebs
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| | - J-Y Blay
- Centre Léon Bérard, UNICANCER, Université Claude Bernard Lyon, Lyon, France
| | - C Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, Paris & Saint-Cloud, France
| | - D Hong
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - L Veronese
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - M Antoniou
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - I Bennett
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
238
|
Kamashev D, Sorokin M, Kochergina I, Drobyshev A, Vladimirova U, Zolotovskaia M, Vorotnikov I, Shaban N, Raevskiy M, Kuzmin D, Buzdin A. Human blood serum can donor-specifically antagonize effects of EGFR-targeted drugs on squamous carcinoma cell growth. Heliyon 2021; 7:e06394. [PMID: 33748471 PMCID: PMC7966997 DOI: 10.1016/j.heliyon.2021.e06394] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/29/2020] [Accepted: 02/25/2021] [Indexed: 02/09/2023] Open
Abstract
Many patients fail to respond to EGFR-targeted therapeutics, and personalized diagnostics is needed to identify putative responders. We investigated 1630 colorectal and lung squamous carcinomas and 1357 normal lung and colon samples and observed huge variation in EGFR pathway activation in both cancerous and healthy tissues, irrespectively on EGFR gene mutation status. We investigated whether human blood serum can affect squamous carcinoma cell growth and EGFR drug response. We demonstrate that human serum antagonizes the effects of EGFR-targeted drugs erlotinib and cetuximab on A431 squamous carcinoma cells by increasing IC50 by about 2- and 20-fold, respectively. The effects on clonogenicity varied significantly across the individual serum samples in every experiment, with up to 100% differences. EGF concentration could explain many effects of blood serum samples, and EGFR ligands-depleted serum showed lesser effect on drug sensitivity.
Collapse
Affiliation(s)
- Dmitry Kamashev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
| | - Maksim Sorokin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Irina Kochergina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
| | - Aleksey Drobyshev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Uliana Vladimirova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
| | - Marianna Zolotovskaia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Igor Vorotnikov
- Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russia
| | - Nina Shaban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Mikhail Raevskiy
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
- OmicsWay Corp., Walnut, CA, USA
| | - Denis Kuzmin
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- Moscow Institute of Physics and Technology (National Research University), Moscow Region 141700, Russia
| |
Collapse
|
239
|
Cobain EF, Wu YM, Vats P, Chugh R, Worden F, Smith DC, Schuetze SM, Zalupski MM, Sahai V, Alva A, Schott AF, Caram MEV, Hayes DF, Stoffel EM, Jacobs MF, Kumar-Sinha C, Cao X, Wang R, Lucas D, Ning Y, Rabban E, Bell J, Camelo-Piragua S, Udager AM, Cieslik M, Lonigro RJ, Kunju LP, Robinson DR, Talpaz M, Chinnaiyan AM. Assessment of Clinical Benefit of Integrative Genomic Profiling in Advanced Solid Tumors. JAMA Oncol 2021; 7:525-533. [PMID: 33630025 PMCID: PMC7907987 DOI: 10.1001/jamaoncol.2020.7987] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Question What is the clinical utility of genomic profiling for patients with advanced solid tumors? Findings In this cohort study of 1015 patients who underwent integrative genomic profiling, a high rate of pathogenic germline variants and a subset of patients who derive substantial clinical benefit from sequencing information were identified. Meaning These findings support (1) directed germline testing for inherited cancer predisposition in all patients with advanced cancer and (2) use of integrative genomic profiling as a component of standard of care for patients with cancer of unknown origin and other rare malignant neoplasms. Importance Use of next-generation sequencing (NGS) to identify clinically actionable genomic targets has been incorporated into routine clinical practice in the management of advanced solid tumors; however, the clinical utility of this testing remains uncertain. Objective To determine which patients derived the greatest degree of clinical benefit from NGS profiling. Design, Setting, and Participants Patients in this cohort study underwent fresh tumor biopsy and blood sample collection for genomic profiling of paired tumor and normal DNA (whole-exome or targeted-exome capture with analysis of 1700 genes) and tumor transcriptome (RNA) sequencing. Somatic and germline genomic alterations were annotated and classified according to degree of clinical actionability. Results were returned to treating oncologists. Data were collected from May 1, 2011, to February 28, 2018, and analyzed from May 1, 2011, to April 30, 2020. Main Outcomes and Measures Patients’ subsequent therapy and treatment response were extracted from the medical record to determine clinical benefit rate from NGS-directed therapy at 6 months and exceptional responses lasting 12 months or longer. Results During the study period, NGS was attempted on tumors from 1138 patients and was successful in 1015 (89.2%) (MET1000 cohort) (538 men [53.0%]; mean [SD] age, 57.7 [13.3] years). Potentially clinically actionable genomic alterations were discovered in 817 patients (80.5%). Of these, 132 patients (16.2%) received sequencing-directed therapy, and 49 had clinical benefit (37.1%). Exceptional responses were observed in 26 patients (19.7% of treated patients). Pathogenic germline variants (PGVs) were identified in 160 patients (15.8% of cohort), including 49 PGVs (4.8% of cohort) with therapeutic relevance. For 55 patients with carcinoma of unknown primary origin, NGS identified the primary site in 28 (50.9%), and sequencing-directed therapy in 13 patients resulted in clinical benefit in 7 instances (53.8%), including 5 exceptional responses. Conclusions and Relevance The high rate of therapeutically relevant PGVs identified across diverse cancer types supports a recommendation for directed germline testing in all patients with advanced cancer. The high frequency of therapeutically relevant somatic and germline findings in patients with carcinoma of unknown primary origin and other rare cancers supports the use of comprehensive NGS profiling as a component of standard of care for these disease entities.
Collapse
Affiliation(s)
- Erin F Cobain
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor.,Department of Pathology, University of Michigan, Ann Arbor
| | - Pankaj Vats
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor
| | - Rashmi Chugh
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Francis Worden
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - David C Smith
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Scott M Schuetze
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Mark M Zalupski
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Vaibhav Sahai
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Ajjai Alva
- Department of Internal Medicine, University of Michigan, Ann Arbor.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor
| | - Anne F Schott
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Megan E V Caram
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Daniel F Hayes
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Elena M Stoffel
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | | | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor.,Department of Pathology, University of Michigan, Ann Arbor
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor
| | - David Lucas
- Department of Pathology, University of Michigan, Ann Arbor
| | - Yu Ning
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor
| | - Erica Rabban
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor
| | - Janice Bell
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor
| | | | - Aaron M Udager
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor.,Department of Pathology, University of Michigan, Ann Arbor
| | - Marcin Cieslik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor.,Department of Pathology, University of Michigan, Ann Arbor
| | - Robert J Lonigro
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor
| | - Lakshmi P Kunju
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor.,Department of Pathology, University of Michigan, Ann Arbor
| | - Dan R Robinson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor.,Department of Pathology, University of Michigan, Ann Arbor
| | - Moshe Talpaz
- Department of Internal Medicine, University of Michigan, Ann Arbor.,Rogel Cancer Center, University of Michigan, Ann Arbor
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor.,Department of Pathology, University of Michigan, Ann Arbor.,Rogel Cancer Center, University of Michigan, Ann Arbor.,Department of Urology, University of Michigan, Ann Arbor.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor
| |
Collapse
|
240
|
Abstract
Glioblastoma remains incurable despite advances in surgery, radiation, and chemotherapy, underscoring the need for new therapies. The genetic heterogenicity, presence of redundant molecular pathways, and the blood-brain barrier have limited the applicability of molecularly targeted agents. The therapeutic benefit seen with a small subset of patients suggests, however, that patient selection is critical. Recent investigations show that molecularly targeted synthetic lethality is a promising complementary approach. The article provides an overview of the challenges of molecularly targeted therapy in adults with glioblastoma, including current trials and future therapeutic directions.
Collapse
Affiliation(s)
- Matthew A Smith-Cohn
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Building 37, Room 1016, Bethesda, MD 20892, USA; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Orieta Celiku
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Building 37, Room 1142, Bethesda, MD 20892, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
241
|
Patient Derived Xenografts for Genome-Driven Therapy of Osteosarcoma. Cells 2021; 10:cells10020416. [PMID: 33671173 PMCID: PMC7922432 DOI: 10.3390/cells10020416] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is a rare malignant primary tumor of mesenchymal origin affecting bone. It is characterized by a complex genotype, mainly due to the high frequency of chromothripsis, which leads to multiple somatic copy number alterations and structural rearrangements. Any effort to design genome-driven therapies must therefore consider such high inter- and intra-tumor heterogeneity. Therefore, many laboratories and international networks are developing and sharing OS patient-derived xenografts (OS PDX) to broaden the availability of models that reproduce OS complex clinical heterogeneity. OS PDXs, and new cell lines derived from PDXs, faithfully preserve tumor heterogeneity, genetic, and epigenetic features and are thus valuable tools for predicting drug responses. Here, we review recent achievements concerning OS PDXs, summarizing the methods used to obtain ectopic and orthotopic xenografts and to fully characterize these models. The availability of OS PDXs across the many international PDX platforms and their possible use in PDX clinical trials are also described. We recommend the coupling of next-generation sequencing (NGS) data analysis with functional studies in OS PDXs, as well as the setup of OS PDX clinical trials and co-clinical trials, to enhance the predictive power of experimental evidence and to accelerate the clinical translation of effective genome-guided therapies for this aggressive disease.
Collapse
|
242
|
Kato S, McFall T, Takahashi K, Bamel K, Ikeda S, Eskander RN, Plaxe S, Parker B, Stites E, Kurzrock R. KRAS-Mutated, Estrogen Receptor-Positive Low-Grade Serous Ovarian Cancer: Unraveling an Exceptional Response Mystery. Oncologist 2021; 26:e530-e536. [PMID: 33528846 PMCID: PMC8018312 DOI: 10.1002/onco.13702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/22/2021] [Indexed: 12/31/2022] Open
Abstract
We report on a woman with aggressive estrogen receptor‐positive, KRAS‐mutated ovarian cancer who achieved a remarkable response to combination therapy with the MEK inhibitor (trametinib) and the aromatase inhibitor (letrozole), even though the disease had failed to respond to a combination of a PI3K inhibitor and different MEK inhibitor, as well as to trametinib and the estrogen modulator, tamoxifen, and to letrozole by itself. The mechanism of action for exceptional response was elucidated by in vitro experiments that demonstrated that the fact that tamoxifen can have an agonistic effect in addition to antagonist activity, whereas letrozole results only in estrogen depletion was crucial to the response achieved when letrozole was combined with an MEK inhibitor. Our current observations indicate that subtle variations in mechanisms of action of outwardly similar regimens may have a major impact on outcome and that such translational knowledge is critical for optimizing a precision medicine strategy. Key Points This report describes the remarkable response of a patient with KRAS‐mutated, estrogen receptor‐positive low‐grade serous ovarian cancer treated with trametinib (MEK inhibitor) and letrozole (aromatase inhibitor), despite prior progression on similar agents including tamoxifen (estrogen modulator). In vitro investigation revealed that tamoxifen can have agonistic in addition to antagonistic effects, which could be the reason for the patient not responding to the combination of trametinib and tamoxifen. The current observations suggest that drugs with different mechanisms of action targeting the same receptor may have markedly different anticancer activity when used in combinations.
This article reports the case of a patient with aggressive estrogen receptor‐positive, KRAS‐mutated ovarian cancer who achieved a remarkable response to combination therapy with the MEK inhibitor trametinib and the aromatase inhibitor letrozole, despite earlier failures of treatment with other combination inhibitor treatment. This article focuses on the possibility that subtle variations in mechanisms of action of outwardly similar regimens may have major effects on patient outcomes.
Collapse
Affiliation(s)
- Shumei Kato
- Center for Personalized Cancer Therapy and Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Thomas McFall
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Kenta Takahashi
- Cancer Center, Tokyo Medical and Dental University, Medical Hospital Bunkyo-ku, Tokyo, Japan
| | - Kasey Bamel
- Center for Personalized Cancer Therapy and Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Sadakatsu Ikeda
- Cancer Center, Tokyo Medical and Dental University, Medical Hospital Bunkyo-ku, Tokyo, Japan
| | - Ramez N Eskander
- Center for Personalized Cancer Therapy and Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Steven Plaxe
- Center for Personalized Cancer Therapy and Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Barbara Parker
- Center for Personalized Cancer Therapy and Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Edward Stites
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
243
|
Okamura R, Kurzrock R, Mallory RJ, Fanta PT, Burgoyne AM, Clary BM, Kato S, Sicklick JK. Comprehensive genomic landscape and precision therapeutic approach in biliary tract cancers. Int J Cancer 2021; 148:702-712. [PMID: 32700810 PMCID: PMC7739197 DOI: 10.1002/ijc.33230] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/21/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Biliary tract cancers have dismal prognoses even when cytotoxic chemotherapy is administered. There is an unmet need to develop precision treatment approaches using comprehensive genomic profiling. A total of 121 patients with biliary tract cancers were analyzed for circulating-tumor DNA (ctDNA) and/or tissue-based tumor DNA (tissue-DNA) using clinical-grade next-generation sequencing: 71 patients (59%) had ctDNA; 90 (74%), tissue-DNA; and 40 (33%), both. Efficacy of targeted therapeutic approaches was assessed based upon ctDNA and tissue-DNA. At least one characterized alteration was detected in 76% of patients (54/71) for ctDNA [median, 2 (range, 0-9)] and 100% (90/90) for tissue-DNA [median, 4 (range, 1-9)]. Most common alterations occurred in TP53 (38%), KRAS (28%), and PIK3CA (14%) for ctDNA vs TP53 (44%), CDKN2A/B (33%) and KRAS (29%) for tissue-DNA. In 40 patients who had both ctDNA and tissue-DNA sequencing, overall concordance was higher between ctDNA and metastatic site tissue-DNA than between ctDNA and primary tumor DNA (78% vs 65% for TP53, 100% vs 74% for KRAS and 100% vs 87% for PIK3CA [But not statistical significance]). Among 80 patients who received systemic treatment, the molecularly matched therapeutic regimens based on genomic profiling showed a significantly longer progression-free survival (hazard ratio [95%confidence interval], 0.60 [0.37-0.99]. P = .047 [multivariate]) and higher disease control rate (61% vs 35%, P = .04) than unmatched regimens. Evaluation of ctDNA and tissue-DNA is feasible in biliary tract cancers.
Collapse
Affiliation(s)
- Ryosuke Okamura
- Center for Personalized Cancer TherapyUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
- Division of Hematology‐OncologyUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | - Razelle Kurzrock
- Center for Personalized Cancer TherapyUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
- Division of Hematology‐OncologyUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | - Robert J. Mallory
- Division of Surgical Oncology, Department of SurgeryUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | - Paul T. Fanta
- Division of Hematology‐OncologyUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | - Adam M. Burgoyne
- Division of Hematology‐OncologyUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | - Bryan M. Clary
- Division of Surgical Oncology, Department of SurgeryUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | - Shumei Kato
- Center for Personalized Cancer TherapyUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
- Division of Hematology‐OncologyUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| | - Jason K. Sicklick
- Center for Personalized Cancer TherapyUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
- Division of Surgical Oncology, Department of SurgeryUC San Diego Moores Cancer CenterLa JollaCaliforniaUSA
| |
Collapse
|
244
|
Abstract
Progress in genomic analytical technologies has improved our possibilities to obtain information regarding DNA, RNA, and their dynamic changes that occur over time or in response to specific challenges. This information describes the blueprint for cells, tissues, and organisms and has fundamental importance for all living organisms. This review focuses on the technological challenges to analyze the transcriptome and what is the impact of transcriptomics on precision medicine. The transcriptome is a term that covers all RNA present in cells and a substantial part of it will never be translated into protein but is nevertheless functional in determining cell phenotype. Recent developments in transcriptomics have challenged the fundamentals of the central dogma of biology by providing evidence of pervasive transcription of the genome. Such massive transcriptional activity is challenging the definition of a gene and especially the term "pseudogene" that has now been demonstrated in many examples to be both transcribed and translated. We also review the common sources of biomaterials for transcriptomics and justify the suitability of whole blood RNA as the current optimal analyte for clinical transcriptomics. At the end of the review, a brief overview of the clinical implications of transcriptomics in clinical trial design and clinical diagnosis is given. Finally, we introduce the transcriptome as a target for modern drug development as a tool for extending our capacity for precision medicine in multiple diseases.
Collapse
Affiliation(s)
| | - Abigail L Pfaff
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch 6150, Australia
- Perron Institute for Neurological and Translational Science, Nedlands 6009, Australia
| | - Vivien J Bubb
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3BX, UK
| | - John P Quinn
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3BX, UK
| | - Sulev Koks
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch 6150, Australia
- Perron Institute for Neurological and Translational Science, Nedlands 6009, Australia
| |
Collapse
|
245
|
Buzdin A, Skvortsova II, Li X, Wang Y. Editorial: Next Generation Sequencing Based Diagnostic Approaches in Clinical Oncology. Front Oncol 2021; 10:635555. [PMID: 33585258 PMCID: PMC7876435 DOI: 10.3389/fonc.2020.635555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 01/26/2023] Open
Affiliation(s)
- Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia.,Translational Genome Bioinformatics Laboratory, Moscow Institute of Physics and Technology (National Research University), Moscow, Russia.,Research Department, OmicsWay Corp., Walnut, CA, United States
| | - Ira Ida Skvortsova
- Therapeutic Radiology and Oncology, Medical University of Innsbruck, Innsbruck, Austria.,Group for Experimental and Translational Radiooncology, Tyrolean Cancer Research Institute, Innsbruck, Austria.,PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Xinmin Li
- Department of Pathology & Laboratory Medicine, University of California Los Angeles (UCLA) Technology Center for Genomics & Bioinformatics, Los Angeles, CA, United States
| | - Ye Wang
- Clinical Laboratory, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, China
| |
Collapse
|
246
|
Gilhooley MJ, Owen N, Moosajee M, Yu Wai Man P. From Transcriptomics to Treatment in Inherited Optic Neuropathies. Genes (Basel) 2021; 12:147. [PMID: 33499292 PMCID: PMC7912133 DOI: 10.3390/genes12020147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Inherited optic neuropathies, including Leber Hereditary Optic Neuropathy (LHON) and Dominant Optic Atrophy (DOA), are monogenetic diseases with a final common pathway of mitochondrial dysfunction leading to retinal ganglion cell (RGC) death and ultimately loss of vision. They are, therefore, excellent models with which to investigate this ubiquitous disease process-implicated in both common polygenetic ocular diseases (e.g., Glaucoma) and late-onset central nervous system neurodegenerative diseases (e.g., Parkinson disease). In recent years, cellular and animal models of LHON and DOA have matured in parallel with techniques (such as RNA-seq) to determine and analyze the transcriptomes of affected cells. This confluence leaves us at a particularly exciting time with the potential for the identification of novel pathogenic players and therapeutic targets. Here, we present a discussion of the importance of inherited optic neuropathies and how transcriptomic techniques can be exploited in the development of novel mutation-independent, neuroprotective therapies.
Collapse
Affiliation(s)
- Michael James Gilhooley
- Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK; (N.O.); (M.M.); (P.Y.W.M.)
- Moorfields Eye Hospital NHS Foundation Trust, 162 City Road, London EC1V 2PD, UK
| | - Nicholas Owen
- Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK; (N.O.); (M.M.); (P.Y.W.M.)
| | - Mariya Moosajee
- Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK; (N.O.); (M.M.); (P.Y.W.M.)
- Moorfields Eye Hospital NHS Foundation Trust, 162 City Road, London EC1V 2PD, UK
- The Francis Crick Institute, 1 Midland Road, Somers Town, London NW1 1AT, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Patrick Yu Wai Man
- Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK; (N.O.); (M.M.); (P.Y.W.M.)
- Moorfields Eye Hospital NHS Foundation Trust, 162 City Road, London EC1V 2PD, UK
- Department of Clinical Neurosciences, University of Cambridge, Robinson Way, Cambridge CB2 0PY, UK
- MRC Mitochondrial Biology Unit, University of Cambridge, Robinson Way, Cambridge CB2 0PY, UK
- Cambridge Eye Unit, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
247
|
Hildebrand LA, Pierce CJ, Dennis M, Paracha M, Maoz A. Artificial Intelligence for Histology-Based Detection of Microsatellite Instability and Prediction of Response to Immunotherapy in Colorectal Cancer. Cancers (Basel) 2021; 13:391. [PMID: 33494280 PMCID: PMC7864494 DOI: 10.3390/cancers13030391] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Microsatellite instability (MSI) is a molecular marker of deficient DNA mismatch repair (dMMR) that is found in approximately 15% of colorectal cancer (CRC) patients. Testing all CRC patients for MSI/dMMR is recommended as screening for Lynch Syndrome and, more recently, to determine eligibility for immune checkpoint inhibitors in advanced disease. However, universal testing for MSI/dMMR has not been uniformly implemented because of cost and resource limitations. Artificial intelligence has been used to predict MSI/dMMR directly from hematoxylin and eosin (H&E) stained tissue slides. We review the emerging data regarding the utility of machine learning for MSI classification, focusing on CRC. We also provide the clinician with an introduction to image analysis with machine learning and convolutional neural networks. Machine learning can predict MSI/dMMR with high accuracy in high quality, curated datasets. Accuracy can be significantly decreased when applied to cohorts with different ethnic and/or clinical characteristics, or different tissue preparation protocols. Research is ongoing to determine the optimal machine learning methods for predicting MSI, which will need to be compared to current clinical practices, including next-generation sequencing. Predicting response to immunotherapy remains an unmet need.
Collapse
Affiliation(s)
- Lindsey A. Hildebrand
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA; (L.A.H.); (C.J.P.); (M.D.); (M.P.)
| | - Colin J. Pierce
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA; (L.A.H.); (C.J.P.); (M.D.); (M.P.)
| | - Michael Dennis
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA; (L.A.H.); (C.J.P.); (M.D.); (M.P.)
- Division of Hematology Oncology, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Munizay Paracha
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA; (L.A.H.); (C.J.P.); (M.D.); (M.P.)
| | - Asaf Maoz
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA; (L.A.H.); (C.J.P.); (M.D.); (M.P.)
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
248
|
Chen AP, Kummar S, Moore N, Rubinstein LV, Zhao Y, Williams PM, Palmisano A, Sims D, O'Sullivan Coyne G, Rosenberger CL, Simpson M, Raghav KPS, Meric-Bernstam F, Leong S, Waqar S, Foster JC, Konaté MM, Das B, Karlovich C, Lih CJ, Polley E, Simon R, Li MC, Piekarz R, Doroshow JH. Molecular Profiling-Based Assignment of Cancer Therapy (NCI-MPACT): A Randomized Multicenter Phase II Trial. JCO Precis Oncol 2021; 5:PO.20.00372. [PMID: 33928209 PMCID: PMC8078898 DOI: 10.1200/po.20.00372] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
This trial assessed the utility of applying tumor DNA sequencing to treatment selection for patients with advanced, refractory cancer and somatic mutations in one of four signaling pathways by comparing the efficacy of four study regimens that were either matched to the patient's aberrant pathway (experimental arm) or not matched to that pathway (control arm). MATERIALS AND METHODS Adult patients with an actionable mutation of interest were randomly assigned 2:1 to receive either (1) a study regimen identified to target the aberrant pathway found in their tumor (veliparib with temozolomide or adavosertib with carboplatin [DNA repair pathway], everolimus [PI3K pathway], or trametinib [RAS/RAF/MEK pathway]), or (2) one of the same four regimens, but chosen from among those not targeting that pathway. RESULTS Among 49 patients treated in the experimental arm, the objective response rate was 2% (95% CI, 0% to 10.9%). One of 20 patients (5%) in the experimental trametinib cohort had a partial response. There were no responses in the other cohorts. Although patients and physicians were blinded to the sequencing and random assignment results, a higher pretreatment dropout rate was observed in the control arm (22%) compared with the experimental arm (6%; P = .038), suggesting that some patients may have had prior tumor mutation profiling performed that led to a lack of participation in the control arm. CONCLUSION Further investigation, better annotation of predictive biomarkers, and the development of more effective agents are necessary to inform treatment decisions in an era of precision cancer medicine. Increasing prevalence of tumor mutation profiling and preference for targeted therapy make it difficult to use a randomized phase II design to evaluate targeted therapy efficacy in an advanced disease setting.
Collapse
Affiliation(s)
- Alice P. Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Nancy Moore
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Yingdong Zhao
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - P. Mickey Williams
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Alida Palmisano
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- General Dynamics Information Technology (GDIT), Falls Church, VA
| | - David Sims
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Mel Simpson
- Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Kanwal P. S. Raghav
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Funda Meric-Bernstam
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Saiama Waqar
- Department of Medical Oncology, Washington University School of Medicine, St Louis, MO
| | - Jared C. Foster
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Mariam M. Konaté
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Biswajit Das
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chris Karlovich
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chih-Jian Lih
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Eric Polley
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Richard Simon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Ming-Chung Li
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Richard Piekarz
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
249
|
Kato S, Okamura R, Adashek JJ, Khalid N, Lee S, Nguyen V, Sicklick JK, Kurzrock R. Targeting G1/S phase cell-cycle genomic alterations and accompanying co-alterations with individualized CDK4/6 inhibitor-based regimens. JCI Insight 2021; 6:142547. [PMID: 33427211 PMCID: PMC7821594 DOI: 10.1172/jci.insight.142547] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/18/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUNDAlthough CDK4/6 inhibitors are an established treatment for hormone receptor-positive, HER2-negative metastatic breast cancers, their benefit in other malignancies remains limited.METHODSWe investigated factors associated with clinical outcomes from CDK4/6 inhibitor-based therapy among patients with G1/S phase cell-cycle alterations (CDK4/6 amplifications, CCND1/2/3 amplifications, or CDKN2A/B alterations).RESULTSOverall, 2457 patients with diverse solid tumors that underwent clinical-grade, next-generation sequencing (182-465 genes) and therapy outcome of (non-breast cancer) patients treated with matched CDK4/6 inhibitors were analyzed. G1/S phase cell-cycle alterations occurred in 20.6% (507 of 2457) of patients; 99% of those patients (n = 501) harbored ≥1 characterized co-alteration (median, 4; range, 0-24). In 40 patients with G1/S phase cell-cycle alterations given CDK4/6 inhibitors as part of their regimen, significantly longer median progression-free survival (PFS) was observed when CDK4/6 inhibitor-based therapies matched a larger proportion of tumor alterations, often because CDK4/6 inhibitors were administered together with other drugs that were matched to genomic co-alterations, hence achieving a high matching score (high vs. low [≥50% vs. <50%] matching score, PFS, 6.2 vs. 2.0 months, P < 0.001 [n = 40] [multivariate]) and higher rate of stable disease ≥6 months or an objective response (57% vs. 21%, P = 0.048).CONCLUSIONIn summary, in cell-cycle-altered cancers, matched CDK4/6 inhibitors, as part of an individualized regimen targeting a majority of genomic alterations, was independently associated with longer PFS.TRIAL REGISTRATIONClinicalTrials.gov NCT02478931.FUNDINGJoan and Irwin Jacobs Fund, National Cancer Institute (P30 CA023100, R01 CA226803), and the FDA (R01 FD006334).
Collapse
Affiliation(s)
- Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, Moores Cancer Center at UC San Diego Health, La Jolla, California, USA
| | - Ryosuke Okamura
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, Moores Cancer Center at UC San Diego Health, La Jolla, California, USA
| | - Jacob J Adashek
- Department of Internal Medicine, University of South Florida, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Noor Khalid
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, Moores Cancer Center at UC San Diego Health, La Jolla, California, USA
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, Moores Cancer Center at UC San Diego Health, La Jolla, California, USA
| | - Van Nguyen
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, Moores Cancer Center at UC San Diego Health, La Jolla, California, USA
| | - Jason K Sicklick
- Center for Personalized Cancer Therapy and Division of Surgical Oncology, Department of Surgery, Moores Cancer Center at UC San Diego Health, La Jolla, California, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, Moores Cancer Center at UC San Diego Health, La Jolla, California, USA
| |
Collapse
|
250
|
Homicsko K. Deep Tumor Profiling for Molecular Tumor Boards. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11680-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|