1551
|
Abstract
Surgical resection is the mainstay of therapy for patients with resectable and operable early stage non-small cell lung cancer (NSCLC). Surgery alone yields an unacceptably high rate of lung cancer recurrence. The addition of chemotherapy to surgery as adjuvant or neoadjuvant treatment can improve survival rates by roughly 5% at 5 years. Recently, major advances in cancer immunotherapy have led to better outcomes for many patients with lung cancer. Monoclonal antibodies to programmed death 1 and its ligand are now approved for both first and second line treatment patients with metastatic lung cancer. In this review, we will outline the rationale and current research strategies investigating the role of immunotherapy in resectable NSCLC.
Collapse
Affiliation(s)
- Dwight Owen
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Jamie E Chaft
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
1552
|
Affiliation(s)
- Jun Qian
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Pierre P Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| |
Collapse
|
1553
|
Hench IB, Hench J, Tolnay M. Liquid Biopsy in Clinical Management of Breast, Lung, and Colorectal Cancer. Front Med (Lausanne) 2018; 5:9. [PMID: 29441349 PMCID: PMC5797586 DOI: 10.3389/fmed.2018.00009] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022] Open
Abstract
Examination of tumor molecular characteristics by liquid biopsy is likely to greatly influence personalized cancer patient management. Analysis of circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and tumor-derived exosomes, all collectively referred to as “liquid biopsies,” are not only a modality to monitor treatment efficacy, disease progression, and emerging therapy resistance mechanisms, but they also assess tumor heterogeneity and evolution in real time. We review the literature concerning the examination of ctDNA and CTC in a diagnostic setting, evaluating their prognostic, predictive, and monitoring capabilities. We discuss the advantages and limitations of various leading ctDNA/CTC analysis technologies. Finally, guided by the results of clinical trials, we discuss the readiness of cell-free DNA and CTC as routine biomarkers in the context of various common types of neoplastic disease. At this moment, one cannot conclude whether or not liquid biopsy will become a mainstay in oncology practice.
Collapse
Affiliation(s)
- Ivana Bratić Hench
- Institute for Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Jürgen Hench
- Institute for Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Markus Tolnay
- Institute for Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
1554
|
Bakhoum SF, Ngo B, Laughney AM, Cavallo JA, Murphy CJ, Ly P, Shah P, Sriram RK, Watkins TBK, Taunk NK, Duran M, Pauli C, Shaw C, Chadalavada K, Rajasekhar VK, Genovese G, Venkatesan S, Birkbak NJ, McGranahan N, Lundquist M, LaPlant Q, Healey JH, Elemento O, Chung CH, Lee NY, Imielenski M, Nanjangud G, Pe’er D, Cleveland DW, Powell SN, Lammerding J, Swanton C, Cantley LC. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 2018; 553:467-472. [PMID: 29342134 PMCID: PMC5785464 DOI: 10.1038/nature25432] [Citation(s) in RCA: 1054] [Impact Index Per Article: 150.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022]
Abstract
Chromosomal instability is a hallmark of cancer that results from ongoing errors in chromosome segregation during mitosis. Although chromosomal instability is a major driver of tumour evolution, its role in metastasis has not been established. Here we show that chromosomal instability promotes metastasis by sustaining a tumour cell-autonomous response to cytosolic DNA. Errors in chromosome segregation create a preponderance of micronuclei whose rupture spills genomic DNA into the cytosol. This leads to the activation of the cGAS-STING (cyclic GMP-AMP synthase-stimulator of interferon genes) cytosolic DNA-sensing pathway and downstream noncanonical NF-κB signalling. Genetic suppression of chromosomal instability markedly delays metastasis even in highly aneuploid tumour models, whereas continuous chromosome segregation errors promote cellular invasion and metastasis in a STING-dependent manner. By subverting lethal epithelial responses to cytosolic DNA, chromosomally unstable tumour cells co-opt chronic activation of innate immune pathways to spread to distant organs.
Collapse
Affiliation(s)
- Samuel F. Bakhoum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Bryan Ngo
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Ashley M. Laughney
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Julie-Ann Cavallo
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Charles J. Murphy
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Peter Ly
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, California 92093, USA
| | - Pragya Shah
- Nancy E. and Peter C. Meinig School of Biomedical Engineering & Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14850, USA
| | - Roshan K Sriram
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | | | - Neil K. Taunk
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Mercedes Duran
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Chantal Pauli
- Institute for Pathology and Molecular Pathology, University Hospital Zurich, Zurich 8091, Switzerland
| | - Christine Shaw
- Molecular Cytogenetics Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Kalyani Chadalavada
- Molecular Cytogenetics Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Vinagolu K. Rajasekhar
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Giulio Genovese
- The Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | | | - Nicolai J. Birkbak
- The Francis Crick Institute, London NW1 1AT, UK
- UCL Cancer Institute, London WC1E 6BT, UK
| | - Nicholas McGranahan
- The Francis Crick Institute, London NW1 1AT, UK
- UCL Cancer Institute, London WC1E 6BT, UK
| | - Mark Lundquist
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Quincey LaPlant
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - John H. Healey
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Olivier Elemento
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | | | - Nancy Y. Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Marcin Imielenski
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Gouri Nanjangud
- Molecular Cytogenetics Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Dana Pe’er
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Don W. Cleveland
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, California 92093, USA
| | - Simon N. Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Jan Lammerding
- Nancy E. and Peter C. Meinig School of Biomedical Engineering & Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14850, USA
| | - Charles Swanton
- The Francis Crick Institute, London NW1 1AT, UK
- UCL Cancer Institute, London WC1E 6BT, UK
| | - Lewis C. Cantley
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| |
Collapse
|
1555
|
Lim LC, Lim YM. Proteome Heterogeneity in Colorectal Cancer. Proteomics 2018; 18. [PMID: 29316255 DOI: 10.1002/pmic.201700169] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/17/2017] [Indexed: 01/26/2023]
Abstract
Tumor heterogeneity is an important feature of colorectal cancer (CRC) manifested by dynamic changes in gene expression, protein expression, and availability of different tumor subtypes. Recent publications in the past 10 years have revealed proteome heterogeneity between different colorectal tumors and within the same tumor site. This paper reviews recent research works on the proteome heterogeneity in CRC, which includes the heterogeneity within a single tumor (intratumor heterogeneity), between different anatomical sites at the same organ, and between primary and metastatic sites (intertumor heterogeneity). The potential use of proteome heterogeneity in precision medicine and its implications in biomarker discovery and therapeutic outcomes will be discussed. Identification of the unique proteome landscape between and within individual tumors is imperative for understanding cancer biology and the management of CRC patients.
Collapse
Affiliation(s)
- Lay Cheng Lim
- Centre for Cancer Research, Faculty of Medicine and Health Sciences, University of Tunku Abdul Rahman, Selangor, Malaysia
| | - Yang Mooi Lim
- Centre for Cancer Research, Faculty of Medicine and Health Sciences, University of Tunku Abdul Rahman, Selangor, Malaysia
| |
Collapse
|
1556
|
Barris DM, Weiner SB, Dubin RA, Fremed M, Zhang X, Piperdi S, Zhang W, Maqbool S, Gill J, Roth M, Hoang B, Geller D, Gorlick R, Weiser DA. Detection of circulating tumor DNA in patients with osteosarcoma. Oncotarget 2018; 9:12695-12704. [PMID: 29560102 PMCID: PMC5849166 DOI: 10.18632/oncotarget.24268] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/09/2018] [Indexed: 12/30/2022] Open
Abstract
Identification and quantification of somatic alterations in plasma-derived, circulating tumor DNA (ctDNA) is gaining traction as a non-invasive and cost effective method of disease monitoring in cancer patients, particularly to evaluate response to treatment and monitor for disease recurrence. To our knowledge, genetic analysis of ctDNA in osteosarcoma has not yet been studied. To determine whether somatic alterations can be detected in ctDNA and perhaps applied to patient management in this disease, we collected germline, tumor, and serial plasma samples from pediatric, adolescent, and young adult patients with osteosarcoma and used targeted Next Generation Sequencing (NGS) to identify somatic single nucleotide variants (SNV), insertions and deletions (INDELS), and structural variants (SV) in 7 genes commonly mutated in osteosarcoma. We demonstrate that patient-specific somatic alterations identified through comparison of tumor-germline pairs can be detected and quantified in cell-free DNA of osteosarcoma patients.
Collapse
Affiliation(s)
- David M Barris
- Department of Genetics and Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shoshana B Weiner
- Department of Genetics and Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert A Dubin
- Computational Genomics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael Fremed
- Department of Genetics and Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xusheng Zhang
- Computational Genomics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sajida Piperdi
- Department of Pediatrics, Montefiore Medical Center, Bronx, NY, USA
| | - Wendong Zhang
- Department of Pediatrics, Montefiore Medical Center, Bronx, NY, USA
| | - Shahina Maqbool
- Department of Genetics and Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jonathan Gill
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael Roth
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bang Hoang
- Department of Orthopedic Surgery, Montefiore Medical Center, Bronx, NY, USA
| | - David Geller
- Department of Orthopedic Surgery, Montefiore Medical Center, Bronx, NY, USA
| | - Richard Gorlick
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel A Weiser
- Division of Hematology/Oncology, Children's Hospital at Montefiore, Bronx, NY, USA.,Departments of Pediatrics and Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
1557
|
Abstract
Despite advances in targeted treatments, lung cancer remains a common and deadly malignancy, in part owing to its typical late presentation. Recent developments in lung cancer screening and ongoing efforts aimed at early detection, treatment, and prevention are promising areas to impact the mortality from lung cancer. In the past several years, lung cancer screening with low-dose chest computed tomography (CT) was shown to have mortality benefit, and lung cancer screening programs have been implemented in some clinical settings. Biomarkers for screening, diagnosis, and monitoring of response to therapy are under development. Prevention efforts aimed at smoking cessation are as crucial as ever, and there have been encouraging findings in recent clinical trials of lung cancer chemoprevention. Here we review advancements in the field of lung cancer prevention and early malignancy and discuss future directions that we believe will result in a reduction in the mortality from lung cancer.
Collapse
Affiliation(s)
- Melissa New
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Robert Keith
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA.,VA Eastern Colorado Health Care System, Denver, Colorado, USA.,University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
1558
|
Nahar R, Zhai W, Zhang T, Takano A, Khng AJ, Lee YY, Liu X, Lim CH, Koh TPT, Aung ZW, Lim TKH, Veeravalli L, Yuan J, Teo ASM, Chan CX, Poh HM, Chua IML, Liew AA, Lau DPX, Kwang XL, Toh CK, Lim WT, Lim B, Tam WL, Tan EH, Hillmer AM, Tan DSW. Elucidating the genomic architecture of Asian EGFR-mutant lung adenocarcinoma through multi-region exome sequencing. Nat Commun 2018; 9:216. [PMID: 29335443 PMCID: PMC5768770 DOI: 10.1038/s41467-017-02584-z] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 12/11/2017] [Indexed: 12/14/2022] Open
Abstract
EGFR-mutant lung adenocarcinomas (LUAD) display diverse clinical trajectories and are characterized by rapid but short-lived responses to EGFR tyrosine kinase inhibitors (TKIs). Through sequencing of 79 spatially distinct regions from 16 early stage tumors, we show that despite low mutation burdens, EGFR-mutant Asian LUADs unexpectedly exhibit a complex genomic landscape with frequent and early whole-genome doubling, aneuploidy, and high clonal diversity. Multiple truncal alterations, including TP53 mutations and loss of CDKN2A and RB1, converge on cell cycle dysregulation, with late sector-specific high-amplitude amplifications and deletions that potentially beget drug resistant clones. We highlight the association between genomic architecture and clinical phenotypes, such as co-occurring truncal drivers and primary TKI resistance. Through comparative analysis with published smoking-related LUAD, we postulate that the high intra-tumor heterogeneity observed in Asian EGFR-mutant LUAD may be contributed by an early dominant driver, genomic instability, and low background mutation rates. EGFR mutant lung adenocarcinoma (LUAD) exhibit diverse clinical outcomes in response to targeted therapies. Here the authors show that these LUADs involve a complex genomic landscape with high intratumor heterogeneity, providing insights into the evolutionary trajectory of oncogene-driven LUAD and potential mediators of EGFR TKI resistance.
Collapse
Affiliation(s)
- Rahul Nahar
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Weiwei Zhai
- Human Genetics, Genome Institute of Singapore, Singapore, 138672, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Tong Zhang
- Human Genetics, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Angela Takano
- Department of Pathology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Alexis J Khng
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Yin Yeng Lee
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Xingliang Liu
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Chong Hee Lim
- Department of Cardiothoracic Surgery, National Heart Centre Singapore, Singapore, 169609, Singapore
| | - Tina P T Koh
- Department of Cardiothoracic Surgery, National Heart Centre Singapore, Singapore, 169609, Singapore
| | - Zaw Win Aung
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Tony Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore, 169608, Singapore
| | - Lavanya Veeravalli
- Research Pipeline Development, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Ju Yuan
- Cancer Stem Cell Biology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Audrey S M Teo
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Cheryl X Chan
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Huay Mei Poh
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Ivan M L Chua
- Next Generation Sequencing Platform, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Audrey Ann Liew
- Cancer Stem Cell Biology, Genome Institute of Singapore, Singapore, 138672, Singapore.,Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.,Cancer Therapeutics Research Laboratory, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Dawn Ping Xi Lau
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.,Cancer Therapeutics Research Laboratory, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Xue Lin Kwang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.,Cancer Therapeutics Research Laboratory, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Chee Keong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Bing Lim
- Cancer Stem Cell Biology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Wai Leong Tam
- Cancer Stem Cell Biology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Eng-Huat Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Axel M Hillmer
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore, 138672, Singapore. .,Institute of Pathology, University Hospital Cologne, 50937, Cologne, Germany.
| | - Daniel S W Tan
- Cancer Stem Cell Biology, Genome Institute of Singapore, Singapore, 138672, Singapore. .,Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore. .,Cancer Therapeutics Research Laboratory, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, 169610, Singapore.
| |
Collapse
|
1559
|
|
1560
|
Pihlak R, Weaver JMJ, Valle JW, McNamara MG. Advances in Molecular Profiling and Categorisation of Pancreatic Adenocarcinoma and the Implications for Therapy. Cancers (Basel) 2018; 10:E17. [PMID: 29329208 PMCID: PMC5789367 DOI: 10.3390/cancers10010017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to be a disease with poor outcomes and short-lived treatment responses. New information is emerging from genome sequencing identifying potential subgroups based on somatic and germline mutations. A variety of different mutations and mutational signatures have been identified; the driver mutation in around 93% of PDAC is KRAS, with other recorded alterations being SMAD4 and CDKN2A. Mutations in the deoxyribonucleic acid (DNA) damage repair pathway have also been investigated in PDAC and multiple clinical trials are ongoing with DNA-damaging agents. Rare mutations in BRAF and microsatellite instability (MSI) have been reported in about 1-3% of patients with PDAC, and agents used in other cancers to target these have also shown some promise. Immunotherapy is a developing field, but has failed to demonstrate benefits in PDAC to date. While many trials have failed to improve outcomes in this deadly disease, there is optimism that by developing a better understanding of the translational aspects of this cancer, future informed therapeutic strategies may prove more successful.
Collapse
Affiliation(s)
- Rille Pihlak
- Division of Cancer Sciences, University of Manchester, Manchester M13 9NT, UK.
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK.
| | - Jamie M J Weaver
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK.
| | - Juan W Valle
- Division of Cancer Sciences, University of Manchester, Manchester M13 9NT, UK.
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK.
| | - Mairéad G McNamara
- Division of Cancer Sciences, University of Manchester, Manchester M13 9NT, UK.
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK.
| |
Collapse
|
1561
|
Affiliation(s)
- Christian Rolfo
- Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, Edegem, Belgium.
- Center for Oncological Research, University of Antwerp, Edegem, Belgium.
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, VU University medical center, Amsterdam, The Netherlands.
- Cancer Pharmacology Lab, AIRC-Start-Up Unit, Pisa, Italy.
| |
Collapse
|
1562
|
Chen K, Chen W, Cai J, Yang F, Lou F, Wang X, Zhang J, Zhao M, Zhang J, Wang J. Favorable prognosis and high discrepancy of genetic features in surgical patients with multiple primary lung cancers. J Thorac Cardiovasc Surg 2018; 155:371-379.e1. [DOI: 10.1016/j.jtcvs.2017.08.141] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/16/2017] [Accepted: 08/09/2017] [Indexed: 12/17/2022]
|
1563
|
McDonald F, Hanna GG. Oligoprogressive Oncogene-addicted Lung Tumours: Does Stereotactic Body Radiotherapy Have a Role? Introducing the HALT Trial. Clin Oncol (R Coll Radiol) 2018; 30:1-4. [PMID: 29153859 DOI: 10.1016/j.clon.2017.10.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/01/2017] [Accepted: 10/09/2017] [Indexed: 11/19/2022]
Affiliation(s)
| | - G G Hanna
- The Royal Marsden Hospital, London, UK
| |
Collapse
|
1564
|
Goldman JW, Noor ZS, Remon J, Besse B, Rosenfeld N. Are liquid biopsies a surrogate for tissue EGFR testing? Ann Oncol 2018; 29:i38-i46. [PMID: 29462257 DOI: 10.1093/annonc/mdx706] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Molecular profiling has changed the treatment landscape in advanced non-small-cell lung cancer. Accurately identifying the tumours that harbour sensitizing EGFR mutations, the most common targetable molecular alteration, as well as those with acquired resistance mutations (e.g. T790M) on treatment is a high clinical priority. The current clinical gold standard is genotyping of tumour specimens. However, the practical utility of this approach is limited by the lack of available tissue and the potential complications associated with biopsies. With the advent of newer sequencing assays, it has become feasible to assess tumour genomics via a blood sample, termed a 'liquid biopsy'. In this review, we summarize the available techniques for liquid biopsies and their applicability for detecting sensitizing and resistance EGFR mutations and how these results may be used for making treatment decisions.
Collapse
Affiliation(s)
- J W Goldman
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, USA
| | - Z S Noor
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, USA
| | - J Remon
- Department of Oncology Medicine, Gustave Roussy, Villejuif
| | - B Besse
- Department of Oncology Medicine, Gustave Roussy, Villejuif
- University Paris-Sud, Orsay, France
| | - N Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge
- Cancer Research UK Major Centre - Cambridge, Cambridge, UK
| |
Collapse
|
1565
|
Dreyer SB, Jamieson NB, Upstill-Goddard R, Bailey PJ, McKay CJ, Biankin AV, Chang DK. Defining the molecular pathology of pancreatic body and tail adenocarcinoma. Br J Surg 2018; 105:e183-e191. [PMID: 29341146 PMCID: PMC5817249 DOI: 10.1002/bjs.10772] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/14/2017] [Accepted: 10/30/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a dismal disease, with very little improvement in survival over the past 50 years. Recent large-scale genomic studies have improved understanding of the genomic and transcriptomic landscape of the disease, yet very little is known about molecular heterogeneity according to tumour location in the pancreas; body and tail PDACs especially tend to have a significantly worse prognosis. The aim was to investigate the molecular differences between PDAC of the head and those of the body and tail of the pancreas. METHODS Detailed correlative analysis of clinicopathological variables, including tumour location, genomic and transcriptomic data, was performed using the Australian Pancreatic Cancer Genome Initiative (APGI) cohort, part of the International Cancer Genome Consortium study. RESULTS Clinicopathological data were available for 518 patients recruited to the APGI, of whom 421 underwent genomic analyses; 179 of these patients underwent whole-genome and 96 RNA sequencing. Patients with tumours of the body and tail had significantly worse survival than those with pancreatic head tumours (12·1 versus 22·0 months; P = 0·001). Location in the body and tail was associated with the squamous subtype of PDAC. Body and tail PDACs enriched for gene programmes involved in tumour invasion and epithelial-to-mesenchymal transition, as well as features of poor antitumour immune response. Whether this is due to a molecular predisposition from the outset, or reflects a later time point on the tumour molecular clock, requires further investigation using well designed prospective studies in pancreatic cancer. CONCLUSION PDACs of the body and tail demonstrate aggressive tumour biology that may explain worse clinical outcomes.
Collapse
Affiliation(s)
- S B Dreyer
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - N B Jamieson
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - P J Bailey
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - C J McKay
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - A V Biankin
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - D K Chang
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
1566
|
Ziogas DE, Kyrochristos ID, Roukos DH. Next-generation sequencing: from conventional applications to breakthrough genomic analyses and precision oncology. Expert Rev Med Devices 2018; 15:1-3. [PMID: 29284319 DOI: 10.1080/17434440.2017.1419059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Demosthenes E Ziogas
- a Centre for Biosystems and Genome Network Medicine , Ioannina University , Ioannina , Greece
- d Department of Surgery , 'G. Hatzikosta' General Hospital , Ioannina , Greece
| | - Ioannis D Kyrochristos
- a Centre for Biosystems and Genome Network Medicine , Ioannina University , Ioannina , Greece
- b Department of Surgery , Ioannina University Hospital , Ioannina , Greece
| | - Dimitrios H Roukos
- a Centre for Biosystems and Genome Network Medicine , Ioannina University , Ioannina , Greece
- b Department of Surgery , Ioannina University Hospital , Ioannina , Greece
- c Department of Systems Biology , Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens , Greece
| |
Collapse
|
1567
|
Lee H, Choi M, Lim J, Jo M, Han JY, Kim TM, Cho Y. Magnetic Nanowire Networks for Dual-Isolation and Detection of Tumor-Associated Circulating Biomarkers. Theranostics 2018; 8:505-517. [PMID: 29290823 PMCID: PMC5743563 DOI: 10.7150/thno.21967] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/21/2017] [Indexed: 11/05/2022] Open
Abstract
Purpose: Recent developments in genomic and molecular methods have revolutionized the range of utilities of tumor-associated circulating biomarkers in both basic and clinical research. Herein, we present a novel approach for ultrasensitive extraction of cfDNA and CTCs, at high yield and purity, via the formation of magnetic nanowire networks. Materials and Methods: We fabricated and characterized biotinylated cationic polyethylenimine and biotinylated antibody cocktail-conjugated magnetic polypyrrole NWs (PEI/mPpy NW and Ab cocktail/mPpy NW, respectively). We applied these NWs to the extraction of cfDNA and CTC from the blood of 14 patients with lung cancer. We demonstrated reliable detection of EGFR mutations based on digital droplet PCR analysis of cfDNA and CTC DNA from patients with lung cancer. Results: The NW networks confined with a high density of magnetic nanoparticles exhibited superior saturation magnetization, which enabled rapid and high-yield capture whilst avoiding or minimizing damage and loss. The NW networks enabled the co-isolation of CTCs and cfDNA of high quality and sufficient quantities, thus allowing the amplification of rare and low-prevalence cancer-related mutations. Conclusion: The simple, versatile, and highly efficient nanowire network tool allows sensitive and robust assessment of clinical samples.
Collapse
|
1568
|
Vargas-Rondón N, Villegas VE, Rondón-Lagos M. The Role of Chromosomal Instability in Cancer and Therapeutic Responses. Cancers (Basel) 2017; 10:cancers10010004. [PMID: 29283387 PMCID: PMC5789354 DOI: 10.3390/cancers10010004] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/22/2017] [Accepted: 12/25/2017] [Indexed: 12/31/2022] Open
Abstract
Cancer is one of the leading causes of death, and despite increased research in recent years, control of advanced-stage disease and optimal therapeutic responses remain elusive. Recent technological improvements have increased our understanding of human cancer as a heterogeneous disease. For instance, four hallmarks of cancer have recently been included, which in addition to being involved in cancer development, could be involved in therapeutic responses and resistance. One of these hallmarks is chromosome instability (CIN), a source of genetic variation in either altered chromosome number or structure. CIN has become a hot topic in recent years, not only for its implications in cancer diagnostics and prognostics, but also for its role in therapeutic responses. Chromosomal alterations are mainly used to determine genetic heterogeneity in tumors, but CIN could also reveal treatment efficacy, as many therapies are based on increasing CIN, which causes aberrant cells to undergo apoptosis. However, it should be noted that contradictory findings on the implications of CIN for the therapeutic response have been reported, with some studies associating high CIN with a better therapeutic response and others associating it with therapeutic resistance. Considering these observations, it is necessary to increase our understanding of the role CIN plays not only in tumor development, but also in therapeutic responses. This review focuses on recent studies that suggest possible mechanisms and consequences of CIN in different disease types, with a primary focus on cancer outcomes and therapeutic responses.
Collapse
Affiliation(s)
- Natalia Vargas-Rondón
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia.
| | - Victoria E Villegas
- Biology Program, Faculty of Natural Sciences and Mathematics, Universidad del Rosario, Bogotá 111221, Colombia.
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia.
| |
Collapse
|
1569
|
Volckmar AL, Sültmann H, Riediger A, Fioretos T, Schirmacher P, Endris V, Stenzinger A, Dietz S. A field guide for cancer diagnostics using cell-free DNA: From principles to practice and clinical applications. Genes Chromosomes Cancer 2017; 57:123-139. [DOI: 10.1002/gcc.22517] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 12/16/2022] Open
Affiliation(s)
- Anna-Lena Volckmar
- Institute of Pathology, University Hospital Heidelberg; Heidelberg Germany
| | - Holger Sültmann
- Division of Cancer Genome Research; German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK); Heidelberg Germany
| | - Anja Riediger
- Division of Cancer Genome Research; German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK); Heidelberg Germany
| | - Thoas Fioretos
- Department of Clinical Genetics; Lund University; Lund Sweden
- Department of Clinical Genetics; University and Regional Laboratories; Region Skåne Lund Sweden
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg; Heidelberg Germany
| | - Volker Endris
- Institute of Pathology, University Hospital Heidelberg; Heidelberg Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg; Heidelberg Germany
- German Cancer Consortium (DKTK), Partner Site Heidelberg, and German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Steffen Dietz
- Division of Cancer Genome Research; German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK); Heidelberg Germany
| |
Collapse
|
1570
|
Liu D, Abbosh P, Keliher D, Reardon B, Miao D, Mouw K, Weiner-Taylor A, Wankowicz S, Han G, Teo MY, Cipolla C, Kim J, Iyer G, Al-Ahmadie H, Dulaimi E, Chen DYT, Alpaugh RK, Hoffman-Censits J, Garraway LA, Getz G, Carter SL, Bellmunt J, Plimack ER, Rosenberg JE, Van Allen EM. Mutational patterns in chemotherapy resistant muscle-invasive bladder cancer. Nat Commun 2017; 8:2193. [PMID: 29259186 PMCID: PMC5736752 DOI: 10.1038/s41467-017-02320-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022] Open
Abstract
Despite continued widespread use, the genomic effects of cisplatin-based chemotherapy and implications for subsequent treatment are incompletely characterized. Here, we analyze whole exome sequencing of matched pre- and post-neoadjuvant cisplatin-based chemotherapy primary bladder tumor samples from 30 muscle-invasive bladder cancer patients. We observe no overall increase in tumor mutational burden post-chemotherapy, though a significant proportion of subclonal mutations are unique to the matched pre- or post-treatment tumor, suggesting chemotherapy-induced and/or spatial heterogeneity. We subsequently identify and validate a novel mutational signature in post-treatment tumors consistent with known characteristics of cisplatin damage and repair. We find that post-treatment tumor heterogeneity predicts worse overall survival, and further observe alterations in cell-cycle and immune checkpoint regulation genes in post-treatment tumors. These results provide insight into the clinical and genomic dynamics of tumor evolution with cisplatin-based chemotherapy, suggest mechanisms of clinical resistance, and inform development of clinically relevant biomarkers and trials of combination therapies. The impact of cisplatin-based chemotherapy on tumor genomes is complex. Here, the authors study matched pre- and post-chemotherapy primary samples in muscle-invasive bladder cancer, finding a cisplatin-based mutational signature, and highlighting the impact of intratumor heterogeneity on survival.
Collapse
Affiliation(s)
- David Liu
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Philip Abbosh
- Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Daniel Keliher
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Brendan Reardon
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Diana Miao
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Kent Mouw
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | | | - Stephanie Wankowicz
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Garam Han
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Min Yuen Teo
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Jaegil Kim
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Gopa Iyer
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Essel Dulaimi
- Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | | | | | - Levi A Garraway
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Gad Getz
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Scott L Carter
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Joaquim Bellmunt
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | | | | | - Eliezer M Van Allen
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
| |
Collapse
|
1571
|
Kyrochristos ID, Ziogas DE, Glantzounis GK, Roukos DH. Prediction of pancreatic cancer risk and therapeutic response with next-generation sequencing. Biomark Med 2017; 12:5-8. [PMID: 29235881 DOI: 10.2217/bmm-2017-0315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Ioannis D Kyrochristos
- Centre for Biosystems & Genome Network Medicine, Ioannina University, Ioannina, 45110, Greece.,Department of Surgery, Ioannina University Hospital, Ioannina, 45110, Greece
| | - Demosthenes E Ziogas
- Centre for Biosystems & Genome Network Medicine, Ioannina University, Ioannina, 45110, Greece.,Department of Surgery, 'G. Hatzikosta' General Hospital, Ioannina, 45001, Greece
| | | | - Dimitrios H Roukos
- Centre for Biosystems & Genome Network Medicine, Ioannina University, Ioannina, 45110, Greece.,Department of Surgery, Ioannina University Hospital, Ioannina, 45110, Greece.,Department of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, 11527, Greece
| |
Collapse
|
1572
|
Comunanza V, Bussolino F. Therapy for Cancer: Strategy of Combining Anti-Angiogenic and Target Therapies. Front Cell Dev Biol 2017; 5:101. [PMID: 29270405 PMCID: PMC5725406 DOI: 10.3389/fcell.2017.00101] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022] Open
Abstract
The concept that blood supply is required and necessary for cancer growth and spreading is intuitive and was firstly formalized by Judah Folkman in 1971, when he demonstrated that cancer cells release molecules able to promote the proliferation of endothelial cells and the formation of new vessels. This seminal result has initiated one of the most fascinating story of the medicine, which is offering a window of opportunity for cancer treatment based on the use of molecules inhibiting tumor angiogenesis and in particular vascular-endothelial growth factor (VEGF), which is the master gene in vasculature formation and is the commonest target of anti-angiogenic regimens. However, the clinical results are far from the remarkable successes obtained in pre-clinical models. The reasons of this discrepancy have been partially understood and well addressed in many reviews (Bergers and Hanahan, 2008; Bottsford-Miller et al., 2012; El-Kenawi and El-Remessy, 2013; Wang et al., 2015; Jayson et al., 2016). At present anti-angiogenic regimens are not used as single treatments but associated with standard chemotherapies. Based on emerging knowledge of the biology of VEGF, here we sustain the hypothesis of the efficacy of a dual approach based on targeting pro-angiogenic pathways and other druggable targets such as mutated oncogenes or the immune system.
Collapse
Affiliation(s)
- Valentina Comunanza
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute FPO-IRCCS, Candiolo, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute FPO-IRCCS, Candiolo, Italy
| |
Collapse
|
1573
|
González S, Volkova N, Beer P, Gerstung M. Immuno-oncology from the perspective of somatic evolution. Semin Cancer Biol 2017; 52:75-85. [PMID: 29223477 DOI: 10.1016/j.semcancer.2017.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/29/2017] [Accepted: 12/05/2017] [Indexed: 12/30/2022]
Abstract
The past years have witnessed significant success for cancer immunotherapies that activate a patient's immune system against their cancer cells. At the same time our understanding of the genetic changes driving tumor evolution have progressed dramatically. The study of cancer genomes has shown that tumors are best understood as cell populations governed by the rules of evolution, leading to the emergence and spread of cell lineages with pathogenic mutations. Moreover, somatic evolution can explain the acquisition of mutations conferring drug resistance in the ever-lasting battle for reaching even fitter cell states. Here, we review the current state of the art of somatic cancer evolution and mechanisms of immune control and escape. We also revisit the principles of immunotherapy from the perspective of somatic evolution and discuss the basic rules of resistance to immunotherapies as dictated by evolution.
Collapse
Affiliation(s)
- Santiago González
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK
| | - Nadezda Volkova
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK
| | - Philip Beer
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK.
| | - Moritz Gerstung
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK.
| |
Collapse
|
1574
|
Affiliation(s)
- Erik Sahai
- Tumour Cell Biology Laboratory, Francis Crick Institute
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
1575
|
Mason J, Blyth B, MacManus MP, Martin OA. Treatment for non-small-cell lung cancer and circulating tumor cells. Lung Cancer Manag 2017; 6:129-139. [PMID: 30643579 PMCID: PMC6310303 DOI: 10.2217/lmt-2017-0019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/02/2018] [Indexed: 12/25/2022] Open
Abstract
Surgery is the main curative therapy for patients with localized non-small-cell lung cancer while radiotherapy (RT), alone or with concurrent platinum-based chemotherapy, remains the primary curative modality for locoregionally advanced non-small-cell lung cancer. The risk of distant metastasis is high after curative-intent treatment, largely attributable to the presence of undetected micrometastases, but which could also be related to treatment-related increases in circulating tumor cells (CTCs). CTC mobilization by RT or systemic therapies might either reflect efficient tumor destruction with improved prognosis, or might promote metastasis and thus represent a potential therapeutic target. RT may induce prometastatic biological alterations in CTC at the cellular level, which are detectable by 'liquid biopsies', though their rarity represents a major challenge. Improved methods of isolation and ex vivo propagation will be essential for the future of CTC research.
Collapse
Affiliation(s)
- Joel Mason
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Research Division, Peter MacCallum Cancer Center, Melbourne, Australia
- Department of Pathology, The University of Melbourne, Melbourne, Australia
| | - Benjamin Blyth
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Research Division, Peter MacCallum Cancer Center, Melbourne, Australia
| | - Michael P MacManus
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Olga A Martin
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Research Division, Peter MacCallum Cancer Center, Melbourne, Australia
- Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
1576
|
Zou Z, Qi P, Qing Z, Zheng J, Yang S, Chen W, Yang R. Technologies for analysis of circulating tumour DNA: Progress and promise. Trends Analyt Chem 2017. [DOI: 10.1016/j.trac.2017.08.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
1577
|
Blakely CM, Watkins TB, Wu W, Gini B, Chabon JJ, McCoach CE, McGranahan N, Wilson GA, Birkbak NJ, Olivas VR, Rotow J, Maynard A, Wang V, Gubens MA, Banks KC, Lanman RB, Caulin AF, John JS, Cordero AR, Giannikopoulos P, Simmons AD, Mack PC, Gandara DR, Husain H, Doebele RC, Riess JW, Diehn M, Swanton C, Bivona TG. Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat Genet 2017; 49:1693-1704. [PMID: 29106415 PMCID: PMC5709185 DOI: 10.1038/ng.3990] [Citation(s) in RCA: 408] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/12/2017] [Indexed: 12/12/2022]
Abstract
A widespread approach to modern cancer therapy is to identify a single oncogenic driver gene and target its mutant-protein product (for example, EGFR-inhibitor treatment in EGFR-mutant lung cancers). However, genetically driven resistance to targeted therapy limits patient survival. Through genomic analysis of 1,122 EGFR-mutant lung cancer cell-free DNA samples and whole-exome analysis of seven longitudinally collected tumor samples from a patient with EGFR-mutant lung cancer, we identified critical co-occurring oncogenic events present in most advanced-stage EGFR-mutant lung cancers. We defined new pathways limiting EGFR-inhibitor response, including WNT/β-catenin alterations and cell-cycle-gene (CDK4 and CDK6) mutations. Tumor genomic complexity increases with EGFR-inhibitor treatment, and co-occurring alterations in CTNNB1 and PIK3CA exhibit nonredundant functions that cooperatively promote tumor metastasis or limit EGFR-inhibitor response. This study calls for revisiting the prevailing single-gene driver-oncogene view and links clinical outcomes to co-occurring genetic alterations in patients with advanced-stage EGFR-mutant lung cancer.
Collapse
Affiliation(s)
- Collin M. Blakely
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Thomas B.K. Watkins
- The Francis Crick Institute, London WC2A 3LY, UK. Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, London WC1E 6BT, UK
| | - Wei Wu
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Beatrice Gini
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jacob J. Chabon
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Caroline E. McCoach
- Division of Medical Oncology, Department of Medicine, University of Colorado, Denver, Aurora, CO, USA
| | - Nicholas McGranahan
- The Francis Crick Institute, London WC2A 3LY, UK. Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, London WC1E 6BT, UK
| | - Gareth A. Wilson
- The Francis Crick Institute, London WC2A 3LY, UK. Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, London WC1E 6BT, UK
| | - Nicolai J. Birkbak
- The Francis Crick Institute, London WC2A 3LY, UK. Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, London WC1E 6BT, UK
| | - Victor R. Olivas
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Julia Rotow
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ashley Maynard
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Victoria Wang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Matthew A. Gubens
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | | - Philip C. Mack
- University of California Davis Cancer Center, Sacramento, CA, USA
| | - David R. Gandara
- University of California Davis Cancer Center, Sacramento, CA, USA
| | | | - Robert C. Doebele
- Division of Medical Oncology, Department of Medicine, University of Colorado, Denver, Aurora, CO, USA
| | | | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Charles Swanton
- The Francis Crick Institute, London WC2A 3LY, UK. Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, London WC1E 6BT, UK
| | - Trever G. Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
1578
|
Bräunlein E, Krackhardt AM. Identification and Characterization of Neoantigens As Well As Respective Immune Responses in Cancer Patients. Front Immunol 2017; 8:1702. [PMID: 29250075 PMCID: PMC5714868 DOI: 10.3389/fimmu.2017.01702] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/17/2017] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy has recently emerged as a powerful tool for the treatment of diverse advanced malignancies. In particular, therapeutic application of immune checkpoint modulators, such as anti-CTLA4 or anti-PD-1/PD-L1 antibodies, have shown efficacy in a broad range of malignant diseases. Although pharmacodynamics of these immune modulators are complex, recent studies strongly support the notion that altered peptide ligands presented on tumor cells representing neoantigens may play an essential role in tumor rejection by T cells activated by anti-CTLA4 and anti-PD-1 antibodies. Neoantigens may have diverse sources as viral and mutated proteins. Moreover, posttranslational modifications and altered antigen processing may also contribute to the neoantigenic peptide ligand landscape. Different approaches of target identification are currently applied in combination with subsequent characterization of autologous and non-self T-cell responses against such neoantigens. Additional efforts are required to elucidate key characteristics and interdependences of neoantigens, immunodominance, respective T-cell responses, and the tumor microenvironment in order to define decisive determinants involved in effective T-cell-mediated tumor rejection. This review focuses on our current knowledge of identification and characterization of such neoantigens as well as respective T-cell responses. It closes with challenges to be addressed in future relevant for further improvement of immunotherapeutic strategies in malignant diseases.
Collapse
Affiliation(s)
- Eva Bräunlein
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Angela M Krackhardt
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,German Cancer Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
1579
|
McGranahan N, Rosenthal R, Hiley CT, Rowan AJ, Watkins TBK, Wilson GA, Birkbak NJ, Veeriah S, Van Loo P, Herrero J, Swanton C. Allele-Specific HLA Loss and Immune Escape in Lung Cancer Evolution. Cell 2017; 171:1259-1271.e11. [PMID: 29107330 PMCID: PMC5720478 DOI: 10.1016/j.cell.2017.10.001] [Citation(s) in RCA: 902] [Impact Index Per Article: 112.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/06/2017] [Accepted: 09/28/2017] [Indexed: 12/16/2022]
Abstract
Immune evasion is a hallmark of cancer. Losing the ability to present neoantigens through human leukocyte antigen (HLA) loss may facilitate immune evasion. However, the polymorphic nature of the locus has precluded accurate HLA copy-number analysis. Here, we present loss of heterozygosity in human leukocyte antigen (LOHHLA), a computational tool to determine HLA allele-specific copy number from sequencing data. Using LOHHLA, we find that HLA LOH occurs in 40% of non-small-cell lung cancers (NSCLCs) and is associated with a high subclonal neoantigen burden, APOBEC-mediated mutagenesis, upregulation of cytolytic activity, and PD-L1 positivity. The focal nature of HLA LOH alterations, their subclonal frequencies, enrichment in metastatic sites, and occurrence as parallel events suggests that HLA LOH is an immune escape mechanism that is subject to strong microenvironmental selection pressures later in tumor evolution. Characterizing HLA LOH with LOHHLA refines neoantigen prediction and may have implications for our understanding of resistance mechanisms and immunotherapeutic approaches targeting neoantigens. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK.
| | - Rachel Rosenthal
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Crispin T Hiley
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK; Division of Cancer Studies, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Andrew J Rowan
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Thomas B K Watkins
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Gareth A Wilson
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK; Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Nicolai J Birkbak
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK; Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Peter Van Loo
- Cancer Genomics Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK; Department of Human Genetics, University of Leuven, 3000 BE Leuven, Belgium
| | - Javier Herrero
- Bill Lyons Informatics Centre, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK; Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK.
| |
Collapse
|
1580
|
El-Deiry WS, Taylor B, Neal JW. Tumor Evolution, Heterogeneity, and Therapy for Our Patients With Advanced Cancer: How Far Have We Come? Am Soc Clin Oncol Educ Book 2017; 37:e8-e15. [PMID: 28746017 DOI: 10.1200/edbk_175524] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The clinical and molecular heterogeneity of various cancer types is well documented. In the era of precision oncology whereby molecular profiling of tumors is incorporated into clinical care, both intra- and intertumoral molecular and genetic heterogeneity have been described. Together, they impact patient treatment and outcomes. Host genetics and the tumor microenvironment impact on tumor evolution and heterogeneity through variations in immune cell infiltration, stromal variations, and selection pressures from hypoxia or nutrient stress, among others. Tumor progression and exposure to therapeutic agents lead to further molecular evolution and heterogeneity that is clinically relevant. Moreover, tumors that evolve after diagnosis and as a function of therapy generally become more aggressive and refractory to available therapeutics, including targeted agents and immunotherapy. The evolving clinical and molecular heterogeneity of patient tumors can be explored with various clinical and research-based specimens and testing such as pre- and post-treatment biopsies; serial liquid biopsies; single cell analysis; PDX and organoid models; anatomic, functional, and molecular imaging; and rapid postmortem studies. Other factors that influence tumor heterogeneity include immune checkpoints, cancer stem cells, therapy-acquired resistance mechanisms that may occur through secondary mutations, and adaptive responses. Modern technologic advances for tumor characterization provide opportunities to understand tumor evolution and its impact on clinical outcomes to improve therapeutic regimens. Characterization of novel targets and development of effective therapeutics are needed to target heterogeneity and the evolution of resistance mechanisms.
Collapse
Affiliation(s)
- Wafik S El-Deiry
- From the Fox Chase Cancer Center, Philadelphia, PA; Memorial Sloan Kettering Cancer Center, New York, NY; Stanford University, Palo Alto, CA
| | - Barry Taylor
- From the Fox Chase Cancer Center, Philadelphia, PA; Memorial Sloan Kettering Cancer Center, New York, NY; Stanford University, Palo Alto, CA
| | - Joel W Neal
- From the Fox Chase Cancer Center, Philadelphia, PA; Memorial Sloan Kettering Cancer Center, New York, NY; Stanford University, Palo Alto, CA
| |
Collapse
|
1581
|
Itchins M, Chia PL, Hayes SA, Howell VM, Gill AJ, Cooper WA, John T, Mitchell P, Millward M, Clarke SJ, Solomon B, Pavlakis N. Treatment of ALK-rearranged non-small cell lung cancer: A review of the landscape and approach to emerging patterns of treatment resistance in the Australian context. Asia Pac J Clin Oncol 2017; 13 Suppl 3:3-13. [PMID: 28795492 DOI: 10.1111/ajco.12754] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Since the identification of anaplastic lymphoma kinase (ALK) gene rearrangements in non-small cell lung cancer (NSCLC) in 2005, the treatment of ALK-rearranged NSCLC (ALK+ NSCLC) has evolved at a rapid pace. This molecularly distinct subset of NSCLC has uniquely important biology, clinicopathologic features and mechanisms of drug resistance which impact on the choice of treatment for a patient with this disease. There are multiple ALK tyrosine kinase inhibitors now available in clinical practice with efficacy data continuing to emerge and guide the optimal treatment algorithm. A detailed search of medical databases and clinical trial registries was conducted to capture all relevant articles on this topic enabling an updated detailed overview of the landscape of management of ALK-rearranged NSCLC.
Collapse
Affiliation(s)
- M Itchins
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Northern Cancer Institute, St Leonards, New South Wales, Australia
| | - P L Chia
- Medical Oncology Unit, Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne.,Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Department of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - S A Hayes
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - V M Howell
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - A J Gill
- Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - W A Cooper
- Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.,School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
| | - T John
- Medical Oncology Unit, Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne.,Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Department of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - P Mitchell
- Medical Oncology Unit, Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne.,Olivia Newton-John Cancer Research Institute, Melbourne, Australia.,Department of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - M Millward
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Linear Clinical Research, Nedlands, Western Australia, Australia
| | - S J Clarke
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Northern Cancer Institute, St Leonards, New South Wales, Australia.,Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - B Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - N Pavlakis
- Bill Walsh Translational Research Laboratory, Kolling Institute Medical Institute of Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Sydney Medical School, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Northern Cancer Institute, St Leonards, New South Wales, Australia.,Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
1582
|
Metcalfe JZ, Streicher E, Theron G, Colman RE, Allender C, Lemmer D, Warren R, Engelthaler DM. Cryptic Microheteroresistance Explains Mycobacterium tuberculosis Phenotypic Resistance. Am J Respir Crit Care Med 2017; 196:1191-1201. [PMID: 28614668 DOI: 10.1164/rccm.201703-0556oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
RATIONALE Minority drug-resistant Mycobacterium tuberculosis subpopulations can be associated with phenotypic resistance but are poorly detected by Sanger sequencing or commercial molecular diagnostic assays. OBJECTIVES To determine the role of targeted next-generation sequencing in resolving these minor variant subpopulations. METHODS We used single molecule overlapping reads (SMOR), a targeted next-generation sequencing approach that dramatically reduces sequencing error, to analyze primary cultured isolates phenotypically resistant to rifampin, fluoroquinolones, or aminoglycosides, but for which Sanger sequencing found no resistance-associated variants (RAVs) within respective resistance-determining regions (study group). Isolates also underwent single-colony selection on antibiotic-containing agar, blinded to sequencing results. As a positive control, isolates with multiple colocalizing chromatogram peaks were also analyzed (control group). MEASUREMENTS AND MAIN RESULTS Among 61 primary culture isolates (25 study group and 36 control group), SMOR described 66 (49%) and 45 (33%) of 135 total heteroresistant RAVs at frequencies less than 5% and less than 1% of the total mycobacterial population, respectively. In the study group, SMOR detected minor resistant variant subpopulations in 80% (n = 20/25) of isolates with no Sanger-identified RAVs (median subpopulation size, 1.0%; interquartile range, 0.2-3.9%). Single-colony selection on drug-containing media corroborated SMOR results for 90% (n = 18/20) of RAV-containing specimens, and the absence of RAVs in 60% (n = 3/5) of isolates. Overall, Sanger sequencing was concordant with SMOR for 77% (n = 53/69) of macroheteroresistant (5-95% total population), but only 5% of microheteroresistant (<5%) subpopulations (n = 3/66) across both groups. CONCLUSIONS Cryptic minor variant mycobacterial subpopulations exist below the resolving capability of current drug susceptibility testing methodologies, and may explain an important proportion of false-negative resistance determinations.
Collapse
Affiliation(s)
- John Z Metcalfe
- 1 Division of Pulmonary and Critical Care Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, California
| | - Elizabeth Streicher
- 2 DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Grant Theron
- 2 DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Rebecca E Colman
- 3 Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, San Diego, San Diego, California; and
| | | | - Darrin Lemmer
- 4 Translational Genomics Research Institute, Flagstaff, Arizona
| | - Rob Warren
- 2 DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | | |
Collapse
|
1583
|
Progress and challenges of predictive biomarkers of anti PD-1/PD-L1 immunotherapy: A systematic review. Cancer Lett 2017; 414:166-173. [PMID: 29155348 DOI: 10.1016/j.canlet.2017.11.014] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/26/2017] [Accepted: 11/11/2017] [Indexed: 12/22/2022]
Abstract
Despite the marked success of applications of PD-1/PD-L1 checkpoint blockades in clinical, the efficacy and responsiveness of these agents varies greatly among different tumor types and across individual patients. Therefore, establishment of predictive biomarkers for checkpoint blockades is of the most importance to maximize the therapeutic benefits. In this review, we discuss the current progress and challenges of developing predictive biomarkers of immunotherapy responsiveness, aiming to provide some directions for future studies. PD-L1 expression is a logical biomarker for the prediction of response to anti-PD-(L)1 immunotherapies. However, the predictive values of PD-L1 expressions for immunotherapy are currently debating and challenging. Multiplex detecting methods and combined biomarkers may provide new strategies. For example, tumor mutation and neoantigens burden, some oncogene mutations, like EGFR, ALK, KRAS and STK11. In addition, with development of new probes and tracers, immuno-PET provide a new, non-invasive and quantitative strategy to monitor treatment response. As current evidence of those potential predictors, a consensus and standardization is needed to establish to widely applied in future studies. Multiplex detecting methods and combined biomarkers may provide new strategies.
Collapse
|
1584
|
TRACERx: Tracking tumor evolution to impact the course of lung cancer. J Thorac Cardiovasc Surg 2017; 155:1199-1202. [PMID: 29331175 DOI: 10.1016/j.jtcvs.2017.10.134] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/27/2017] [Accepted: 10/11/2017] [Indexed: 11/21/2022]
|
1585
|
Abstract
Cancer is a dynamic disease. During the course of disease, cancers generally become more heterogeneous. As a result of this heterogeneity, the bulk tumour might include a diverse collection of cells harbouring distinct molecular signatures with differential levels of sensitivity to treatment. This heterogeneity might result in a non-uniform distribution of genetically distinct tumour-cell subpopulations across and within disease sites (spatial heterogeneity) or temporal variations in the molecular makeup of cancer cells (temporal heterogeneity). Heterogeneity provides the fuel for resistance; therefore, an accurate assessment of tumour heterogeneity is essential for the development of effective therapies. Multiregion sequencing, single-cell sequencing, analysis of autopsy samples, and longitudinal analysis of liquid biopsy samples are all emerging technologies with considerable potential to dissect the complex clonal architecture of cancers. In this Review, we discuss the driving forces behind intratumoural heterogeneity and the current approaches used to combat this heterogeneity and its consequences. We also explore how clinical assessments of tumour heterogeneity might facilitate the development of more-effective personalized therapies.
Collapse
|
1586
|
Moore DA, Young CA, Morris HT, Oien KA, Lee JL, Jones JL, Salto-Tellez M. Time for change: a new training programme for morpho-molecular pathologists? J Clin Pathol 2017; 71:285-290. [PMID: 29113995 PMCID: PMC5868526 DOI: 10.1136/jclinpath-2017-204821] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 12/20/2022]
Abstract
The evolution of cellular pathology as a specialty has always been driven by technological developments and the clinical relevance of incorporating novel investigations into diagnostic practice. In recent years, the molecular characterisation of cancer has become of crucial relevance in patient treatment both for predictive testing and subclassification of certain tumours. Much of this has become possible due to the availability of next-generation sequencing technologies and the whole-genome sequencing of tumours is now being rolled out into clinical practice in England via the 100 000 Genome Project. The effective integration of cellular pathology reporting and genomic characterisation is crucial to ensure the morphological and genomic data are interpreted in the relevant context, though despite this, in many UK centres molecular testing is entirely detached from cellular pathology departments. The CM-Path initiative recognises there is a genomics knowledge and skills gap within cellular pathology that needs to be bridged through an upskilling of the current workforce and a redesign of pathology training. Bridging this gap will allow the development of an integrated ‘morphomolecular pathology’ specialty, which can maintain the relevance of cellular pathology at the centre of cancer patient management and allow the pathology community to continue to be a major influence in cancer discovery as well as playing a driving role in the delivery of precision medicine approaches. Here, several alternative models of pathology training, designed to address this challenge, are presented and appraised.
Collapse
Affiliation(s)
- David A Moore
- Department of Cancer Studies, University of Leicester, Leicester, UK
| | | | - Hayley T Morris
- Institute of Cancer Sciences - Pathology, University of Glasgow, Glasgow, UK
| | - Karin A Oien
- Institute of Cancer Sciences - Pathology, University of Glasgow, Glasgow, UK
| | - Jessica L Lee
- Strategy and Initiatives, National Cancer Research Institute, London, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, London, UK
| | - Manuel Salto-Tellez
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| |
Collapse
|
1587
|
Lee H, Um SW. Intratumoral heterogeneity and biomarkers for transformation into small cell lung carcinomas from lung adenocarcinomas. J Thorac Dis 2017; 9:4248-4250. [PMID: 29268484 DOI: 10.21037/jtd.2017.10.91] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Hyun Lee
- Department of Medicine, Samsung Medical Center, Division of Pulmonary and Critical Care Medicine, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sang-Won Um
- Department of Medicine, Samsung Medical Center, Division of Pulmonary and Critical Care Medicine, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
1588
|
Abstract
The expanding spectrum of both established and candidate oncogenic driver mutations identified in non-small-cell lung cancer (NSCLC), coupled with the increasing number of clinically available signal transduction pathway inhibitors targeting these driver mutations, offers a tremendous opportunity to enhance patient outcomes. Despite these molecular advances, advanced-stage NSCLC remains largely incurable due to therapeutic resistance. In this Review, we discuss alterations in the targeted oncogene ('on-target' resistance) and in other downstream and parallel pathways ('off-target' resistance) leading to resistance to targeted therapies in NSCLC, and we provide an overview of the current understanding of the bidirectional interactions with the tumour microenvironment that promote therapeutic resistance. We highlight common mechanistic themes underpinning resistance to targeted therapies that are shared by NSCLC subtypes, including those with oncogenic alterations in epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), ROS1 proto-oncogene receptor tyrosine kinase (ROS1), serine/threonine-protein kinase b-raf (BRAF) and other less established oncoproteins. Finally, we discuss how understanding these themes can inform therapeutic strategies, including combination therapy approaches, and overcome the challenge of tumour heterogeneity.
Collapse
Affiliation(s)
- Julia Rotow
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, 505 Parnassus Avenue, Box 1270, San Francisco, California 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, Box 0981, San Francisco, California 94143, USA
| | - Trever G Bivona
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco, 505 Parnassus Avenue, Box 1270, San Francisco, California 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, Box 0981, San Francisco, California 94143, USA
- Cellular and Molecular Pharmacology, University of California San Francisco, Box 2140, San Francisco, California 94158, USA
| |
Collapse
|
1589
|
Scher HI, Graf RP, Schreiber NA, McLaughlin B, Jendrisak A, Wang Y, Lee J, Greene S, Krupa R, Lu D, Bamford P, Louw JE, Dugan L, Vargas HA, Fleisher M, Landers M, Heller G, Dittamore R. Phenotypic Heterogeneity of Circulating Tumor Cells Informs Clinical Decisions between AR Signaling Inhibitors and Taxanes in Metastatic Prostate Cancer. Cancer Res 2017; 77:5687-5698. [PMID: 28819021 PMCID: PMC5666339 DOI: 10.1158/0008-5472.can-17-1353] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/07/2017] [Accepted: 08/09/2017] [Indexed: 01/25/2023]
Abstract
The heterogeneity of an individual patient's tumor has been linked to treatment resistance, but quantitative biomarkers to rapidly and reproducibly evaluate heterogeneity in a clinical setting are currently lacking. Using established tools available in a College of American Pathologists-accredited and Clinical Laboratory Improvement Amendments-certified clinical laboratory, we quantified digital pathology features on 9,225 individual circulating tumor cells (CTC) from 179 unique metastatic castration-resistant prostate cancer (mCRPC) patients to define phenotypically distinct cell types. Heterogeneity was quantified on the basis of the diversity of cell types in individual patient samples using the Shannon index and associated with overall survival (OS) in the 145 specimens collected prior to initiation of the second or later lines of therapy. Low CTC phenotypic heterogeneity was associated with better OS in patients treated with androgen receptor signaling inhibitors (ARSI), whereas high heterogeneity was associated with better OS in patients treated with taxane chemotherapy. Overall, the results show that quantifying CTC phenotypic heterogeneity can help inform the choice between ARSI and taxanes in mCRPC patients. Cancer Res; 77(20); 5687-98. ©2017 AACR.
Collapse
Affiliation(s)
- Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | | | - Nicole A Schreiber
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brigit McLaughlin
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | | | | | - David Lu
- Epic Sciences, La Jolla, California
| | | | | | | | - Hebert A Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Martin Fleisher
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Glenn Heller
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
1590
|
Duréndez-Sáez E, Azkárate A, Meri M, Calabuig-Fariñas S, Aguilar-Gallardo C, Blasco A, Jantus-Lewintre E, Camps C. New insights in non-small-cell lung cancer: circulating tumor cells and cell-free DNA. J Thorac Dis 2017; 9:S1332-S1345. [PMID: 29184672 DOI: 10.21037/jtd.2017.06.112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Lung cancer is the second most frequent tumor and the leading cause of death by cancer in both men and women. Increasing knowledge about the cancer genome and tumor environment has led to a new setting in which morphological and molecular characterization is needed to treat patients in the most personalized way in order to achieve better outcomes. Since tumor products can be detected in body fluids, the liquid biopsy, particularly, peripheral blood, has emerged as a new source for lung cancer biomarker's analysis. A variety of tumor components can be used for this purpose. Among them, circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) should be especially considered. Different detection methods for both CTCs and ctDNA have been and are being developed to improve the sensitivity and specificity of these tests. This would lead to better characterization and would solve some clinical doubts at different disease evolution times, e.g., intratumoral or temporal heterogeneity, difficulty in the obtaining a tumor sample, etc., and would also avoid the side effects of very expensive and complicated tumor obtaining interventions. CTCs and ctDNA are useful in different lung cancer settings. Their value has been shown for the early diagnosis, prognosis, prediction of treatment efficacy, monitoring responses and early detection of lung cancer relapse. CTCs have still not been validated for use in clinical settings in non-small-cell lung cancer (NSCLC), while ctDNA has been approved by the Food and Drug Administration (FDA) and European Medical Association (EMA), and the main clinical guidelines used for detect different epidermal growth factor receptor (EGFR) mutations and the monitoring and treatment choice of mutated patients with tyrosine kinase inhibitors (TKIs). This review, describes how ctDNA seem to be winning the race against CTCs from the laboratory bench to clinical practice due to easier obtaining methods, manipulation and its implementation into clinical practice.
Collapse
Affiliation(s)
- Elena Duréndez-Sáez
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain
| | - Aitor Azkárate
- Department of Oncology, University Hospital Son Espases, Palma de Mallorca, Spain
| | - Marina Meri
- Department of Medical Oncology, Hospital General Universitario de Valencia, Valencia, Spain
| | - Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain.,Department of Pathology, Universitat de València, Valencia, Spain.,CIBERONC, Valencia, Spain
| | | | - Ana Blasco
- Department of Medical Oncology, Hospital General Universitario de Valencia, Valencia, Spain.,CIBERONC, Valencia, Spain
| | - Eloisa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain.,CIBERONC, Valencia, Spain.,Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain
| | - Carlos Camps
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain.,Department of Medical Oncology, Hospital General Universitario de Valencia, Valencia, Spain.,CIBERONC, Valencia, Spain.,Department of Medicine, Universitat de València, Valencia, Spain
| |
Collapse
|
1591
|
Li J, Yan S, Liu Z, Zhou Y, Pan Y, Yuan W, Liu M, Tan Q, Tian G, Dong B, Cai H, Wu N, Ke Y. Multiregional Sequencing Reveals Genomic Alterations and Clonal Dynamics in Primary Malignant Melanoma of the Esophagus. Cancer Res 2017; 78:338-347. [PMID: 28972077 DOI: 10.1158/0008-5472.can-17-0938] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/28/2017] [Accepted: 09/18/2017] [Indexed: 11/16/2022]
Abstract
Primary malignant melanoma of the esophagus (PMME) is a rare and aggressive disease with high tendency of metastasis. To characterize the genetic basis and intratumor heterogeneity of PMME, we performed multiregion exome sequencing and whole genome SNP array genotyping of 12 samples obtained from a patient with PMME. High intratumor heterogeneity was observed in both somatic mutation and copy-number alteration levels. Nine geographically separate samples including two normal samples were clonally related and followed a branched evolution model. Most putative oncogenic drivers such as BRAF and KRAS mutations as well as CDKN2A biallelic inactivation were observed in trunk clones, whereas clinically actionable mutations such as PIK3CA and JAK1 mutations were detected in branch clones. Ancestor tumor clones evolved into three subclonal clades: clade1 fostered metastatic subclones that carried metastatic features of PIK3CA and ARHGAP26 point mutations as well as chr13 arm-level deletion, clade2 owned branch-specific JAK1 mutations and PTEN deletion, and clade3 was found in two vertical distribution samples below the primary tumor area, highlighting the fact that it is possible for PMME to disseminate by the submucosal longitudinal lymphatic route at an early stage of metastasis. These findings facilitate interpretation of the genetic essence of this rare melanoma subtype as well as the pattern of cancer evolution, thus reinforcing the therapeutic challenges associated with PMME.Significance: This study highlights the use of multiregion exome sequencing and whole genome SNP array genotyping to comprehensively characterize the genetic landscape of a rare type of esophogeal melanoma. Cancer Res; 78(2); 338-47. ©2017 AACR.
Collapse
Affiliation(s)
- Jingjing Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yaqi Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - WenQin Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Mengfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Qin Tan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | | | - Bin Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Lab, Peking University Cancer Hospital and Institute, Beijing, China
| | - Hong Cai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, China.
| | - Yang Ke
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
1592
|
Chaudhuri AA, Chabon JJ, Lovejoy AF, Newman AM, Stehr H, Azad TD, Khodadoust MS, Esfahani MS, Liu CL, Zhou L, Scherer F, Kurtz DM, Say C, Carter JN, Merriott DJ, Dudley JC, Binkley MS, Modlin L, Padda SK, Gensheimer MF, West RB, Shrager JB, Neal JW, Wakelee HA, Loo BW, Alizadeh AA, Diehn M. Early Detection of Molecular Residual Disease in Localized Lung Cancer by Circulating Tumor DNA Profiling. Cancer Discov 2017; 7:1394-1403. [PMID: 28899864 DOI: 10.1158/2159-8290.cd-17-0716] [Citation(s) in RCA: 696] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/07/2017] [Accepted: 08/31/2017] [Indexed: 12/15/2022]
Abstract
Identifying molecular residual disease (MRD) after treatment of localized lung cancer could facilitate early intervention and personalization of adjuvant therapies. Here, we apply cancer personalized profiling by deep sequencing (CAPP-seq) circulating tumor DNA (ctDNA) analysis to 255 samples from 40 patients treated with curative intent for stage I-III lung cancer and 54 healthy adults. In 94% of evaluable patients experiencing recurrence, ctDNA was detectable in the first posttreatment blood sample, indicating reliable identification of MRD. Posttreatment ctDNA detection preceded radiographic progression in 72% of patients by a median of 5.2 months, and 53% of patients harbored ctDNA mutation profiles associated with favorable responses to tyrosine kinase inhibitors or immune checkpoint blockade. Collectively, these results indicate that ctDNA MRD in patients with lung cancer can be accurately detected using CAPP-seq and may allow personalized adjuvant treatment while disease burden is lowest.Significance: This study shows that ctDNA analysis can robustly identify posttreatment MRD in patients with localized lung cancer, identifying residual/recurrent disease earlier than standard-of-care radiologic imaging, and thus could facilitate personalized adjuvant treatment at early time points when disease burden is lowest. Cancer Discov; 7(12); 1394-403. ©2017 AACR.See related commentary by Comino-Mendez and Turner, p. 1368This article is highlighted in the In This Issue feature, p. 1355.
Collapse
Affiliation(s)
- Aadel A Chaudhuri
- Department of Radiation Oncology, Stanford University, Stanford, California.,Stanford Cancer Institute, Stanford University, Stanford, California
| | - Jacob J Chabon
- Stanford Cancer Institute, Stanford University, Stanford, California.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Alexander F Lovejoy
- Department of Radiation Oncology, Stanford University, Stanford, California.,Stanford Cancer Institute, Stanford University, Stanford, California
| | - Aaron M Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California.,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Henning Stehr
- Stanford Cancer Institute, Stanford University, Stanford, California
| | - Tej D Azad
- Stanford Cancer Institute, Stanford University, Stanford, California
| | - Michael S Khodadoust
- Stanford Cancer Institute, Stanford University, Stanford, California.,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | | | - Chih Long Liu
- Stanford Cancer Institute, Stanford University, Stanford, California.,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Li Zhou
- Stanford Cancer Institute, Stanford University, Stanford, California.,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Florian Scherer
- Stanford Cancer Institute, Stanford University, Stanford, California.,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - David M Kurtz
- Stanford Cancer Institute, Stanford University, Stanford, California.,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California
| | - Carmen Say
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Justin N Carter
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - David J Merriott
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Jonathan C Dudley
- Stanford Cancer Institute, Stanford University, Stanford, California.,Department of Pathology, Stanford University, Stanford, California
| | - Michael S Binkley
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Leslie Modlin
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Sukhmani K Padda
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | | | - Robert B West
- Department of Pathology, Stanford University, Stanford, California
| | - Joseph B Shrager
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Stanford School of Medicine, Stanford University, Stanford, California
| | - Joel W Neal
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Heather A Wakelee
- Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, California. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California.,Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, California
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University, Stanford, California. .,Stanford Cancer Institute, Stanford University, Stanford, California.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| |
Collapse
|
1593
|
Ramón Y Cajal S, Capdevila C, Hernandez-Losa J, De Mattos-Arruda L, Ghosh A, Lorent J, Larsson O, Aasen T, Postovit LM, Topisirovic I. Cancer as an ecomolecular disease and a neoplastic consortium. Biochim Biophys Acta Rev Cancer 2017; 1868:484-499. [PMID: 28947238 DOI: 10.1016/j.bbcan.2017.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/26/2022]
Abstract
Current anticancer paradigms largely target driver mutations considered integral for cancer cell survival and tumor progression. Although initially successful, many of these strategies are unable to overcome the tremendous heterogeneity that characterizes advanced tumors, resulting in the emergence of resistant disease. Cancer is a rapidly evolving, multifactorial disease that accumulates numerous genetic and epigenetic alterations. This results in wide phenotypic and molecular heterogeneity within the tumor, the complexity of which is further amplified through specific interactions between cancer cells and the tumor microenvironment. In this context, cancer may be perceived as an "ecomolecular" disease that involves cooperation between several neoplastic clones and their interactions with immune cells, stromal fibroblasts, and other cell types present in the microenvironment. This collaboration is mediated by a variety of secreted factors. Cancer is therefore analogous to complex ecosystems such as microbial consortia. In the present article, we comment on the current paradigms and perspectives guiding the development of cancer diagnostics and therapeutics and the potential application of systems biology to untangle the complexity of neoplasia. In our opinion, conceptualization of neoplasia as an ecomolecular disease is warranted. Advances in knowledge pertinent to the complexity and dynamics of interactions within the cancer ecosystem are likely to improve understanding of tumor etiology, pathogenesis, and progression. This knowledge is anticipated to facilitate the design of new and more effective therapeutic approaches that target the tumor ecosystem in its entirety.
Collapse
Affiliation(s)
- Santiago Ramón Y Cajal
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Pathology Department, Vall d'Hebron Hospital, 08035 Barcelona, Spain; Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Spain.
| | - Claudia Capdevila
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Javier Hernandez-Losa
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Pathology Department, Vall d'Hebron Hospital, 08035 Barcelona, Spain; Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Spain
| | - Leticia De Mattos-Arruda
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Abhishek Ghosh
- Lady Davis Institute, JGH, SMBD, Gerald-Bronfman Department of Oncology, McGill University QC, Montreal H3T 1E2, Canada
| | - Julie Lorent
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, 171 65 Solna, Sweden
| | - Ola Larsson
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, 171 65 Solna, Sweden
| | - Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Spain
| | - Lynne-Marie Postovit
- Cancer Research Institute of Northern Alberta Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Ivan Topisirovic
- Lady Davis Institute, JGH, SMBD, Gerald-Bronfman Department of Oncology, McGill University QC, Montreal H3T 1E2, Canada
| |
Collapse
|
1594
|
Chen HZ, Bertino EM, He K. Tumor spread through air space (STAS) is an important predictor of clinical outcome in stage IA lung adenocarcinoma. J Thorac Dis 2017; 9:2283-2285. [PMID: 28932525 DOI: 10.21037/jtd.2017.07.69] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Hui-Zi Chen
- The James Cancer Hospital and Solove Research Institute, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.,Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.,Hematology and Medical Oncology Fellowship Program, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Erin M Bertino
- The James Cancer Hospital and Solove Research Institute, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.,Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kai He
- The James Cancer Hospital and Solove Research Institute, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.,Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
1595
|
Abstract
Acquired molecular changes can promote the spreading of primary tumor cells to distant tissues. In this issue of Cell, Roe et al. show that metastatic progression of pancreatic cancer involves large-scale enhancer reprogramming by Foxa1, which activates transcriptional program specifying early endodermal stem cells.
Collapse
Affiliation(s)
- Raul Mostoslavsky
- The Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Massachusetts General Hospital Center for Regenerative Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Nabeel Bardeesy
- The Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Massachusetts General Hospital Center for Regenerative Medicine, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
1596
|
Attili I, Passaro A, Pavan A, Conte P, De Marinis F, Bonanno L. Combination immunotherapy strategies in advanced non-small cell lung cancer (NSCLC): Does biological rationale meet clinical needs? Crit Rev Oncol Hematol 2017; 119:30-39. [PMID: 29065983 DOI: 10.1016/j.critrevonc.2017.09.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/26/2017] [Accepted: 09/13/2017] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have emerged as one of the main new therapeutic options for advanced non-small cell lung cancer (NSCLC) patients. Even though they demonstrated superiority towards standard chemotherapy in different disease settings, the response rates do not exceed 45% in highly molecularly selected patients. This is related to known limitations of the available biomarkers, as well to the complex and dynamic nature of tumor microenvironment. The study of the different strategies adopted by tumor cells to escape the immune system lays the basis of the new combination strategies. This review focuses on analyzing the biological rationale and early clinical data available concerning therapeutic strategies combining ICIs together, ICIs with different regimens and schedules of standard chemotherapy, ICIs with tyrosine kinase inhibitors, ICIs with antiangiogenic agents and ICs with radiotherapy.
Collapse
Affiliation(s)
- Ilaria Attili
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institue of Oncology, Milan, Italy
| | - Alberto Pavan
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - PierFranco Conte
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Filippo De Marinis
- Division of Thoracic Oncology, European Institue of Oncology, Milan, Italy
| | - Laura Bonanno
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy.
| |
Collapse
|
1597
|
Cutts A, Venn O, Dilthey A, Gupta A, Vavoulis D, Dreau H, Middleton M, McVean G, Taylor JC, Schuh A. Characterisation of the changing genomic landscape of metastatic melanoma using cell free DNA. NPJ Genom Med 2017; 2:25. [PMID: 29075515 PMCID: PMC5654504 DOI: 10.1038/s41525-017-0030-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer is characterised by complex somatically acquired genetic aberrations that manifest as intra-tumour and inter-tumour genetic heterogeneity and can lead to treatment resistance. In this case study, we characterise the genome-wide somatic mutation dynamics in a metastatic melanoma patient during therapy using low-input (50 ng) PCR-free whole genome sequencing of cell-free DNA from pre-treatment and post-relapse blood samples. We identify de novo tumour-specific somatic mutations from cell-free DNA, while the sequence context of single nucleotide variants showed the characteristic UV-damage mutation signature of melanoma. To investigate the behaviour of individual somatic mutations during proto-oncogene B-Raf -targeted and immune checkpoint inhibition, amplicon-based deep sequencing was used to verify and track frequencies of 212 single nucleotide variants at 10 distinct time points over 13 months of treatment. Under checkpoint inhibition therapy, we observed an increase in mutant allele frequencies indicating progression on therapy 88 days before clinical determination of non-response positron emission tomogrophy-computed tomography. We also revealed mutations from whole genome sequencing of cell-free DNA that were not present in the tissue biopsy, but that later contributed to relapse. Our findings have potential clinical applications where high quality tumour-tissue derived DNA is not available.
Collapse
Affiliation(s)
- Anthony Cutts
- Nuffield Division of Clinical Laboratory Sciences (NDCLS), Oxford Molecular Diagnostics Centre, University of Oxford, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Oliver Venn
- Lighthouse Cancer Diagnostics Ltd, Oxford, UK.,The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alexander Dilthey
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Avinash Gupta
- University of Oxford Department of Oncology, Churchill Hospital, Oxford, UK
| | - Dimitris Vavoulis
- Nuffield Division of Clinical Laboratory Sciences (NDCLS), Oxford Molecular Diagnostics Centre, University of Oxford, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Helene Dreau
- Nuffield Division of Clinical Laboratory Sciences (NDCLS), Oxford Molecular Diagnostics Centre, University of Oxford, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Mark Middleton
- NIHR Oxford Biomedical Research Centre, Oxford, UK.,University of Oxford Department of Oncology, Churchill Hospital, Oxford, UK
| | - Gil McVean
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Jenny C Taylor
- NIHR Oxford Biomedical Research Centre, Oxford, UK.,The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Anna Schuh
- NIHR Oxford Biomedical Research Centre, Oxford, UK.,University of Oxford Department of Oncology, Churchill Hospital, Oxford, UK.,Department of Haematology, Oxford University Hospital Trust, Oxford, UK
| |
Collapse
|
1598
|
Mechanisms and clinical implications of tumor heterogeneity and convergence on recurrent phenotypes. J Mol Med (Berl) 2017; 95:1167-1178. [PMID: 28871446 DOI: 10.1007/s00109-017-1587-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/11/2017] [Accepted: 08/20/2017] [Indexed: 10/18/2022]
Abstract
Tumor heterogeneity has been identified at various -omic levels. The tumor genome, transcriptome, proteome, and phenome can vary widely across cells in patient tumors and are influenced by tumor cell interactions with heterogeneous physical conditions and cellular components of the tumor microenvironment. Here, we explore the concept that while variation exists at multiple -omic levels, changes at each of these levels converge on the same pathways and lead to convergent phenotypes in tumors that can provide common drug targets. These phenotypes include cellular growth and proliferation, sustained oncogenic signaling, and immune avoidance, among others. Tumor heterogeneity complicates treatment of patient cancers as it leads to varied response to therapies. Identification of convergent cellular phenotypes arising in patient cancers and targeted therapies that reverse them has the potential to transform the way clinicians treat these cancers and to improve patient outcome.
Collapse
|
1599
|
Rostami A, Bratman SV. Utilizing circulating tumour DNA in radiation oncology. Radiother Oncol 2017; 124:357-364. [DOI: 10.1016/j.radonc.2017.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/01/2017] [Accepted: 07/05/2017] [Indexed: 12/25/2022]
|
1600
|
McClelland SE. Role of chromosomal instability in cancer progression. Endocr Relat Cancer 2017; 24:T23-T31. [PMID: 28696210 DOI: 10.1530/erc-17-0187] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 12/24/2022]
Abstract
Cancer cells often display chromosomal instability (CIN), a defect that involves loss or rearrangement of the cell's genetic material - chromosomes - during cell division. This process results in the generation of aneuploidy, a deviation from the haploid number of chromosomes, and structural alterations of chromosomes in over 90% of solid tumours and many haematological cancers. This trait is unique to cancer cells as normal cells in the body generally strictly maintain the correct number and structure of chromosomes. This key difference between cancer and normal cells has led to two important hypotheses: (i) cancer cells have had to overcome inherent barriers to changes in chromosomes that are not tolerated in non-cancer cells and (ii) CIN represents a cancer-specific target to allow the specific elimination of cancer cells from the body. To exploit these hypotheses and design novel approaches to treat cancer, a full understanding of the mechanisms driving CIN and how CIN contributes to cancer progression is required. Here, we will discuss the possible mechanisms driving chromosomal instability, how CIN may contribute to the progression at multiple stages of tumour evolution and possible future therapeutic directions based on targeting cancer chromosomal instability.
Collapse
|