1
|
DeWitt JT, Raghunathan M, Haricharan S. Nonrepair functions of DNA mismatch repair proteins: new avenues for precision oncology. Trends Cancer 2025; 11:49-61. [PMID: 39490324 PMCID: PMC12077842 DOI: 10.1016/j.trecan.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024]
Abstract
DNA damage repair (DDR) proteins are well recognized as guardians of the genome that are frequently lost during malignant transformation of normal cells across cancer types. To date, their tumor suppressor functions have been generally regarded as a consequence of their roles in maintaining genomic stability: more genomic instability increases the risk of oncogenic transformation events. However, recent discoveries centering around DNA mismatch repair (MMR) proteins suggest a broader impact of the loss of DDR proteins on cellular processes beyond genomic instability. Here, we explore the clinical implications of nonrepair roles for DDR proteins, using the growing evidence supporting roles for DNA MMR proteins in cell cycle and apoptosis regulation, metabolic function, the cellular secretome, and immunomodulation.
Collapse
Affiliation(s)
- Jerry Tyler DeWitt
- Department of Biology, San Diego State University, San Diego, CA, USA; Cancer Biology and Signaling Program, UCSD Moores Cancer Center, San Diego, CA, USA
| | - Megha Raghunathan
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Svasti Haricharan
- Department of Biology, San Diego State University, San Diego, CA, USA; Cancer Biology and Signaling Program, UCSD Moores Cancer Center, San Diego, CA, USA.
| |
Collapse
|
2
|
Beckman RA. Neutral evolution of rare cancer mutations in the computer and the clinic. NPJ Syst Biol Appl 2024; 10:110. [PMID: 39358357 PMCID: PMC11447017 DOI: 10.1038/s41540-024-00436-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
A distinct model of neutral evolution of rare cancer mutations is described and contrasted with models relying on the infinite sites approximation (that a specific mutation arises in only one cell at any instant). An explosion of genetic diversity is predicted at clinical cell numbers and may explain the progressive refractoriness of cancers during a clinical course. The widely used infinite sites assumption may not be applicable for clinical cancers.
Collapse
Affiliation(s)
- Robert A Beckman
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA.
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
3
|
Adler FR. A modelling framework for cancer ecology and evolution. J R Soc Interface 2024; 21:20240099. [PMID: 39013418 PMCID: PMC11251767 DOI: 10.1098/rsif.2024.0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/10/2024] [Indexed: 07/18/2024] Open
Abstract
Cancer incidence increases rapidly with age, typically as a polynomial. The somatic mutation theory explains this increase through the waiting time for enough mutations to build up to generate cells with the full set of traits needed to grow without control. However, lines of evidence ranging from tumour reversion and dormancy to the prevalence of presumed cancer mutations in non-cancerous tissues argue that this is not the whole story, and that cancer is also an ecological process, and that mutations only lead to cancer when the systems of control within and across cells have broken down. Aging thus has two effects: the build-up of mutations and the breakdown of control. This paper presents a mathematical modelling framework to unify these theories with novel approaches to model the mutation and diversification of cell lineages and of the breakdown of the layers of control both within and between cells. These models correctly predict the polynomial increase of cancer with age, show how germline defects in control accelerate cancer initiation, and compute how the positive feedback between cell replication, ecology and layers of control leads to a doubly exponential growth of cell populations.
Collapse
Affiliation(s)
- Frederick R. Adler
- Department of Mathematics, School of Biological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
4
|
Lei L, Cao Q, An G, Lv Y, Tang J, Yang J. DDI2 promotes tumor metastasis and resists antineoplastic drugs-induced apoptosis in colorectal cancer. Apoptosis 2022; 28:458-470. [PMID: 36520320 DOI: 10.1007/s10495-022-01796-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 12/23/2022]
Abstract
The normal colorectal mucosa undergoes precancerous lesions that can develop over time into colorectal cancer (CRC). In the stage of precancerous lesions, DNA replication stress may lead to genome instability. We have performed whole-exome sequencing on genomic DNA obtained from three cases of CRC tissues and identified a novel frameshift mutation of DNA damage inducible 1 homolog 2 gene (DDI2, c. 854 del T). To date, there is no direct evidence that DDI2 is involved in the carcinogenesis of CRC. In this study, we demonstrated that DDI2 is upregulated in the early stage of CRC based on clinical samples and public databases. We also found that 5FU, a standard chemotherapeutic agent for CRC treatment, increased DDI2 mRNA levels in a dose-dependent manner. Depression of DDI2 inhibited CRC cell proliferation, migration and invasion both in vitro and in vivo. Transcriptome sequencing revealed that DDI2 was involved in the mitogen-activated protein kinase (MAPK) pathway. Furthermore, DDI2 resists a MAPK kinase (MEK) inhibitor (trametinib) and a PolyADP-ribose polymerase 1 (PARP1) inhibitor (talazoparib) induced apoptosis in CRC cells. Thus, our results indicate that DDI2 may play a vital role in the carcinogenesis of CRC and could serve as a promising therapeutic target for CRC.
Collapse
|
5
|
Abstract
Choosing and optimizing treatment strategies for cancer requires
capturing its complex dynamics sufficiently well for understanding but
without being overwhelmed. Mathematical models are essential to
achieve this understanding, and we discuss the challenge of choosing
the right level of complexity to address the full range of tumor
complexity from growth, the generation of tumor heterogeneity, and
interactions within tumors and with treatments and the tumor
microenvironment. We discuss the differences between conceptual and
descriptive models, and compare the use of predator-prey models,
evolutionary game theory, and dynamic precision medicine approaches in
the face of uncertainty about mechanisms and parameter values.
Although there is of course no one-size-fits-all approach, we conclude
that broad and flexible thinking about cancer, based on combined
modeling approaches, will play a key role in finding creative and
improved treatments.
Collapse
Affiliation(s)
- Robert A Beckman
- Departments of Oncology and Biostatistics, Bioinformatics, & Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, 12231Georgetown University Medical Center, Washington, DC, USA
| | - Irina Kareva
- Mathematical and Computational Sciences Center, School of Human Evolution and Social Change, 7864Arizona State University, Tempe, AZ, USA
| | - Frederick R Adler
- School of Biological Sciences, 415772University of Utah, Salt Lake City, UT, USA.,Department of Mathematics, 415772University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
6
|
Beckman RA, Loeb LA. Rare Mutations in Cancer Drug Resistance and Implications for Therapy. Clin Pharmacol Ther 2020; 108:437-439. [PMID: 32648584 PMCID: PMC7484911 DOI: 10.1002/cpt.1938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/02/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Robert A Beckman
- Departments of Oncology and of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
| | - Lawrence A Loeb
- Departments of Pathology and Biochemistry, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
7
|
Abstract
Advances in our understanding of molecular mechanisms of tumorigenesis have translated into knowledge-based therapies directed against specific oncogenic signaling targets. These therapies often induce dramatic responses in susceptible tumors. Unfortunately, most advanced cancers, including those with robust initial responses, eventually acquire resistance to targeted therapies and relapse. Even though immune-based therapies are more likely to achieve complete cures, acquired resistance remains an obstacle to their success as well. Acquired resistance is the direct consequence of pre-existing intratumor heterogeneity and ongoing diversification during therapy, which enables some tumor cells to survive treatment and facilitates the development of new therapy-resistant phenotypes. In this review, we discuss the sources of intratumor heterogeneity and approaches to capture and account for it during clinical decision making. Finally, we outline potential strategies to improve therapeutic outcomes by directly targeting intratumor heterogeneity.
Collapse
Affiliation(s)
- Andriy Marusyk
- Department of Cancer Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Michalina Janiszewska
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
8
|
Loeb LA, Kohrn BF, Loubet-Senear KJ, Dunn YJ, Ahn EH, O’Sullivan JN, Salk JJ, Bronner MP, Beckman RA. Extensive subclonal mutational diversity in human colorectal cancer and its significance. Proc Natl Acad Sci U S A 2019; 116:26863-26872. [PMID: 31806761 PMCID: PMC6936702 DOI: 10.1073/pnas.1910301116] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Human colorectal cancers (CRCs) contain both clonal and subclonal mutations. Clonal driver mutations are positively selected, present in most cells, and drive malignant progression. Subclonal mutations are randomly dispersed throughout the genome, providing a vast reservoir of mutant cells that can expand, repopulate the tumor, and result in the rapid emergence of resistance, as well as being a major contributor to tumor heterogeneity. Here, we apply duplex sequencing (DS) methodology to quantify subclonal mutations in CRC tumor with unprecedented depth (104) and accuracy (<10-7). We measured mutation frequencies in genes encoding replicative DNA polymerases and in genes frequently mutated in CRC, and found an unexpectedly high effective mutation rate, 7.1 × 10-7. The curve of subclonal mutation accumulation as a function of sequencing depth, using DNA obtained from 5 different tumors, is in accord with a neutral model of tumor evolution. We present a theoretical approach to model neutral evolution independent of the infinite-sites assumption (which states that a particular mutation arises only in one tumor cell at any given time). Our analysis indicates that the infinite-sites assumption is not applicable once the number of tumor cells exceeds the reciprocal of the mutation rate, a circumstance relevant to even the smallest clinically diagnosable tumor. Our methods allow accurate estimation of the total mutation burden in clinical cancers. Our results indicate that no DNA locus is wild type in every malignant cell within a tumor at the time of diagnosis (probability of all cells being wild type, 10-308).
Collapse
Affiliation(s)
- Lawrence A. Loeb
- Department of Pathology, University of Washington, Seattle, WA 98195
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Brendan F. Kohrn
- Department of Pathology, University of Washington, Seattle, WA 98195
| | | | - Yasmin J. Dunn
- Department of Pathology, University of Washington, Seattle, WA 98195
| | - Eun Hyun Ahn
- Department of Pathology, University of Washington, Seattle, WA 98195
| | - Jacintha N. O’Sullivan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, Dublin 8, Ireland
| | - Jesse J. Salk
- Division of Medical Oncology, University of Washington, Seattle, WA 98195
- TwinStrand Biosciences, Inc., Seattle, WA 98121
| | - Mary P. Bronner
- Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Robert A. Beckman
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20007
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University Medical Center, Washington, DC 20007
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC 20007
| |
Collapse
|
9
|
Raynes Y, Weinreich DM. Genomic clustering of fitness-affecting mutations favors the evolution of chromosomal instability. Evol Appl 2019; 12:301-313. [PMID: 30697341 PMCID: PMC6346662 DOI: 10.1111/eva.12717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/16/2018] [Accepted: 09/16/2018] [Indexed: 12/19/2022] Open
Abstract
Most solid cancers are characterized by chromosomal instability (CIN)-an elevated rate of large-scale chromosomal aberrations and ploidy changes. Chromosomal instability may arise through mutations in a range of genomic integrity loci and is commonly associated with fast disease progression, poor prognosis, and multidrug resistance. However, the evolutionary forces promoting CIN-inducing alleles (hereafter, CIN mutators) during carcinogenesis remain poorly understood. Here, we develop a stochastic, individual-based model of indirect selection experienced by CIN mutators via genomic associations with fitness-affecting mutations. Because mutations associated with CIN affect large swaths of the genome and have the potential to simultaneously comprise many individual loci, we show that indirect selection on CIN mutators is critically influenced by genome organization. In particular, we find strong support for a key role played by the spatial clustering of loci with either beneficial or deleterious mutational effects. Genomic clustering of selected loci allows CIN mutators to generate favorable chromosomal changes that facilitate their rapid expansion within a neoplasm and, in turn, accelerate carcinogenesis. We then examine the distribution of oncogenic and tumor-suppressing loci in the human genome and find both to be potentially more clustered along the chromosome than expected, leading us to speculate that human genome may be susceptible to CIN hitchhiking. More quantitative data on fitness effects of individual mutations will be necessary, though, to assess the true levels of clustering in the human genome and the effectiveness of indirect selection for CIN. Finally, we use our model to examine how therapeutic strategies that increase the deleterious burden of genetically unstable cells by raising either the rate of CIN or the cost of deleterious mutations affect CIN evolution. We find that both can inhibit CIN hitchhiking and delay carcinogenesis in some circumstances, yet, in line with earlier work, we find the latter to be considerably more effective.
Collapse
Affiliation(s)
- Yevgeniy Raynes
- Department of Ecology and Evolutionary Biology, Center for Computational Molecular BiologyBrown UniversityProvidenceRhode Island
| | - Daniel M. Weinreich
- Department of Ecology and Evolutionary Biology, Center for Computational Molecular BiologyBrown UniversityProvidenceRhode Island
| |
Collapse
|
10
|
Temko D, Van Gool IC, Rayner E, Glaire M, Makino S, Brown M, Chegwidden L, Palles C, Depreeuw J, Beggs A, Stathopoulou C, Mason J, Baker A, Williams M, Cerundolo V, Rei M, Taylor JC, Schuh A, Ahmed A, Amant F, Lambrechts D, Smit VTHBM, Bosse T, Graham TA, Church DN, Tomlinson I. Somatic POLE exonuclease domain mutations are early events in sporadic endometrial and colorectal carcinogenesis, determining driver mutational landscape, clonal neoantigen burden and immune response. J Pathol 2018; 245:283-296. [PMID: 29604063 PMCID: PMC6032922 DOI: 10.1002/path.5081] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/05/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023]
Abstract
Genomic instability, which is a hallmark of cancer, is generally thought to occur in the middle to late stages of tumourigenesis, following the acquisition of permissive molecular aberrations such as TP53 mutation or whole genome doubling. Tumours with somatic POLE exonuclease domain mutations are notable for their extreme genomic instability (their mutation burden is among the highest in human cancer), distinct mutational signature, lymphocytic infiltrate, and excellent prognosis. To what extent these characteristics are determined by the timing of POLE mutations in oncogenesis is unknown. Here, we have shown that pathogenic POLE mutations are detectable in non-malignant precursors of endometrial and colorectal cancer. Using genome and exome sequencing, we found that multiple driver mutations in POLE-mutant cancers show the characteristic POLE mutational signature, including those in genes conventionally regarded as initiators of tumourigenesis. In POLE-mutant cancers, the proportion of monoclonal predicted neoantigens was similar to that in other cancers, but the absolute number was much greater. We also found that the prominent CD8+ T-cell infiltrate present in POLE-mutant cancers was evident in their precursor lesions. Collectively, these data indicate that somatic POLE mutations are early, quite possibly initiating, events in the endometrial and colorectal cancers in which they occur. The resulting early onset of genomic instability may account for the striking immune response and excellent prognosis of these tumours, as well as their early presentation. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Female
- Humans
- Middle Aged
- Adenocarcinoma/enzymology
- Adenocarcinoma/genetics
- Adenocarcinoma/immunology
- Adenocarcinoma/pathology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Colorectal Neoplasms/enzymology
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/pathology
- Databases, Genetic
- DNA Polymerase II/genetics
- DNA Polymerase II/metabolism
- Endometrial Neoplasms/enzymology
- Endometrial Neoplasms/genetics
- Endometrial Neoplasms/immunology
- Endometrial Neoplasms/pathology
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Genomic Instability
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mutation
- Neoplasm Grading
- Neoplasm Staging
- Phenotype
- Poly-ADP-Ribose Binding Proteins/genetics
- Poly-ADP-Ribose Binding Proteins/metabolism
- Tumor Microenvironment
- Whole Genome Sequencing
Collapse
Affiliation(s)
- Daniel Temko
- Evolution and Cancer Laboratory, Barts Cancer Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Centre for Maths and Physics in the Life Sciences and Experimental Biology (CoMPLEX)University College LondonLondonUK
- Department of Computer ScienceUniversity College LondonLondonUK
| | - Inge C Van Gool
- Department of PathologyLeiden University Medical CentreLeidenThe Netherlands
| | - Emily Rayner
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Mark Glaire
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Seiko Makino
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Matthew Brown
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Laura Chegwidden
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Claire Palles
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Jeroen Depreeuw
- KU Leuven (University of Leuven), University Hospitals Leuven, Department of Obstetrics and GynaecologyDivision of Gynaecological OncologyLeuvenBelgium
- KU Leuven, Department of Human GeneticsLaboratory for Translational GeneticsLeuvenBelgium
- VIB Centre for Cancer BiologyLaboratory for Translational GeneticsLeuvenBelgium
| | - Andrew Beggs
- Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
| | | | - John Mason
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Ann‐Marie Baker
- Evolution and Cancer Laboratory, Barts Cancer Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Marc Williams
- Evolution and Cancer Laboratory, Barts Cancer Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Margarida Rei
- MRC Human Immunology Unit, Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Jenny C Taylor
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Anna Schuh
- Department of OncologyUniversity of OxfordOxfordUK
| | - Ahmed Ahmed
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
- Nuffield Department of Obstetrics & GynaecologyUniversity of OxfordOxfordUK
| | - Frédéric Amant
- Centre for Gynaecological Oncology AmsterdamNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Diether Lambrechts
- KU Leuven, Department of Human GeneticsLaboratory for Translational GeneticsLeuvenBelgium
- VIB Centre for Cancer BiologyLaboratory for Translational GeneticsLeuvenBelgium
| | - Vincent THBM Smit
- Department of PathologyLeiden University Medical CentreLeidenThe Netherlands
| | - Tjalling Bosse
- Department of PathologyLeiden University Medical CentreLeidenThe Netherlands
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Barts Cancer Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - David N Church
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Ian Tomlinson
- Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
11
|
Abstract
A large number of chemicals and several physical agents, such as UV light and γ-radiation, have been associated with the etiology of human cancer. Generation of DNA damage (also known as DNA adducts or lesions) induced by these agents is an important first step in the process of carcinogenesis. Evolutionary processes gave rise to DNA repair tools that are efficient in repairing damaged DNA; yet replication of damaged DNA may take place prior to repair, particularly when they are induced at a high frequency. Damaged DNA replication may lead to gene mutations, which in turn may give rise to altered proteins. Mutations in an oncogene, a tumor-suppressor gene, or a gene that controls the cell cycle can generate a clonal cell population with a distinct advantage in proliferation. Many such events, broadly divided into the stages of initiation, promotion, and progression, which may occur over a long period of time and transpire in the context of chronic exposure to carcinogens, can lead to the induction of human cancer. This is exemplified in the long-term use of tobacco being responsible for an increased risk of lung cancer. This mini-review attempts to summarize this wide area that centers on DNA damage as it relates to the development of human cancer.
Collapse
Affiliation(s)
- Ashis K Basu
- Department of Chemistry, University of Connecticut, Storrs, CT 06269-3060, USA.
| |
Collapse
|
12
|
Alekseenko IV, Vinogradova TV, Sverdlov ED. Genetic Regulatory Mechanisms of Evolution and Embryogenesis in a Distorting Mirror of Carcinogenesis. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418020023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
13
|
Beckman RA, Loeb LA. Evolutionary dynamics and significance of multiple subclonal mutations in cancer. DNA Repair (Amst) 2017; 56:7-15. [PMID: 28652129 DOI: 10.1016/j.dnarep.2017.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
For the last 40 years the authors have collaborated on trying to understand the complexities of human cancer by formulating testable mathematical models that are based on mutation accumulation in human malignancies. We summarize the concepts encompassed by multiple mutations in human cancers in the context of source, accumulation during carcinogenesis and tumor progression, and therapeutic consequences. We conclude that the efficacious treatment of human cancer by targeted therapy will involve individualized, uniquely directed specific agents singly and in simultaneous combinations, and take into account the importance of targeting resistant subclonal mutations, particularly those subclones with alterations in DNA repair genes, DNA polymerase, and other genes required to maintain genetic stability.
Collapse
Affiliation(s)
- Robert A Beckman
- Departments of Oncology and Biostatistics, Bioinformatics, & Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC 20007 USA
| | - Lawrence A Loeb
- Joseph Gottstein Memorial Cancer Research Laboratory, Departments of Pathology and Biochemistry, University of Washington School of Medicine, Seattle, WA, 98195 USA.
| |
Collapse
|
14
|
Alekseenko IV, Kuzmich AI, Pleshkan VV, Tyulkina DV, Zinovyeva MV, Kostina MB, Sverdlov ED. The cause of cancer mutations: Improvable bad life or inevitable stochastic replication errors? Mol Biol 2016. [DOI: 10.1134/s0026893316060030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
15
|
Passerini V, Storchová Z. Too much to handle - how gaining chromosomes destabilizes the genome. Cell Cycle 2016; 15:2867-2874. [PMID: 27636196 PMCID: PMC5105935 DOI: 10.1080/15384101.2016.1231285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/08/2016] [Accepted: 08/26/2016] [Indexed: 10/21/2022] Open
Abstract
Most eukaryotic organisms are diploid, with 2 chromosome sets in their nuclei. Whole chromosomal aneuploidy, a deviation from multiples of the haploid chromosome number, arises from chromosome segregation errors and often has detrimental consequences for cells. In humans, numerical aneuploidy severely impairs embryonic development and the rare survivors develop disorders characterized by multiple pathologies. Moreover, as many as 75 % of malignant tumors display aneuploidy. Although the exact contribution of aneuploidy to tumorigenesis remains unclear, previous studies have suggested that aneuploidy may affect the maintenance of genome integrity. We found that human cells with extra chromosomes showed phenotypes suggestive of replication defects, a phenomenon which we went on to characterize as being due to the aneuploidy-driven downregulation of replication factors, in particular of the replicative helicase MCM2-7. Thus, missegregation of even a single chromosome can further promote genomic instability and thereby contribute to tumor development. In this review we will examine the possible causes of downregulation of replicative factors and discuss the consequences of genomic instability in aneuploid cells.
Collapse
Affiliation(s)
- Verena Passerini
- Group Maintenance of Genome Stability, Max Planck Institute of Biochemistry, Martinsried, Germany
- Center for Integrated Protein Science, Ludwig-Maximilian-University, Munich, Germany
| | - Zuzana Storchová
- Group Maintenance of Genome Stability, Max Planck Institute of Biochemistry, Martinsried, Germany
- Center for Integrated Protein Science, Ludwig-Maximilian-University, Munich, Germany
- Technical University Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
16
|
Yeang CH, Beckman RA. Long range personalized cancer treatment strategies incorporating evolutionary dynamics. Biol Direct 2016; 11:56. [PMID: 27770811 PMCID: PMC5075220 DOI: 10.1186/s13062-016-0153-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 09/21/2016] [Indexed: 02/07/2023] Open
Abstract
Background Current cancer precision medicine strategies match therapies to static consensus molecular properties of an individual’s cancer, thus determining the next therapeutic maneuver. These strategies typically maintain a constant treatment while the cancer is not worsening. However, cancers feature complicated sub-clonal structure and dynamic evolution. We have recently shown, in a comprehensive simulation of two non-cross resistant therapies across a broad parameter space representing realistic tumors, that substantial improvement in cure rates and median survival can be obtained utilizing dynamic precision medicine strategies. These dynamic strategies explicitly consider intratumoral heterogeneity and evolutionary dynamics, including predicted future drug resistance states, and reevaluate optimal therapy every 45 days. However, the optimization is performed in single 45 day steps (“single-step optimization”). Results Herein we evaluate analogous strategies that think multiple therapeutic maneuvers ahead, considering potential outcomes at 5 steps ahead (“multi-step optimization”) or 40 steps ahead (“adaptive long term optimization (ALTO)”) when recommending the optimal therapy in each 45 day block, in simulations involving both 2 and 3 non-cross resistant therapies. We also evaluate an ALTO approach for situations where simultaneous combination therapy is not feasible (“Adaptive long term optimization: serial monotherapy only (ALTO-SMO)”). Simulations utilize populations of 764,000 and 1,700,000 virtual patients for 2 and 3 drug cases, respectively. Each virtual patient represents a unique clinical presentation including sizes of major and minor tumor subclones, growth rates, evolution rates, and drug sensitivities. While multi-step optimization and ALTO provide no significant average survival benefit, cure rates are significantly increased by ALTO. Furthermore, in the subset of individual virtual patients demonstrating clinically significant difference in outcome between approaches, by far the majority show an advantage of multi-step or ALTO over single-step optimization. ALTO-SMO delivers cure rates superior or equal to those of single- or multi-step optimization, in 2 and 3 drug cases respectively. Conclusion In selected virtual patients incurable by dynamic precision medicine using single-step optimization, analogous strategies that “think ahead” can deliver long-term survival and cure without any disadvantage for non-responders. When therapies require dose reduction in combination (due to toxicity), optimal strategies feature complex patterns involving rapidly interleaved pulses of combinations and high dose monotherapy. Reviewers This article was reviewed by Wendy Cornell, Marek Kimmel, and Andrzej Swierniak. Wendy Cornell and Andrzej Swierniak are external reviewers (not members of the Biology Direct editorial board). Andrzej Swierniak was nominated by Marek Kimmel. Electronic supplementary material The online version of this article (doi:10.1186/s13062-016-0153-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Robert A Beckman
- Departments of Oncology and of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
17
|
Abstract
We propose a modification of the Crow-Kimura and Eigen models of biological molecular evolution to include a mutator gene that causes both an increase in the mutation rate and a change in the fitness landscape. This mutator effect relates to a wide range of biomedical problems. There are three possible phases: mutator phase, mixed phase and non-selective phase. We calculate the phase structure, the mean fitness and the fraction of the mutator allele in the population, which can be applied to describe cancer development and RNA viruses. We find that depending on the genome length, either the normal or the mutator allele dominates in the mixed phase. We analytically solve the model for a general fitness function. We conclude that the random fitness landscape is an appropriate choice for describing the observed mutator phenomenon in the case of a small fraction of mutators. It is shown that the increase in the mutation rates in the regular and the mutator parts of the genome should be set independently; only some combinations of these increases can push the complex biomedical system to the non-selective phase, potentially related to the eradication of tumors.
Collapse
|
18
|
Alekseenko IV, Pleshkan VV, Monastyrskaya GS, Kuzmich AI, Snezhkov EV, Didych DA, Sverdlov ED. Fundamentally low reproducibility in molecular genetic cancer research. RUSS J GENET+ 2016; 52:650-663. [DOI: 10.1134/s1022795416070036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
19
|
Schmitt MW, Loeb LA, Salk JJ. The influence of subclonal resistance mutations on targeted cancer therapy. Nat Rev Clin Oncol 2016; 13:335-47. [PMID: 26483300 PMCID: PMC4838548 DOI: 10.1038/nrclinonc.2015.175] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Clinical oncology is being revolutionized by the increasing use of molecularly targeted therapies. This paradigm holds great promise for improving cancer treatment; however, allocating specific therapies to the patients who are most likely to derive a durable benefit continues to represent a considerable challenge. Evidence continues to emerge that cancers are characterized by extensive intratumour genetic heterogeneity, and that patients being considered for treatment with a targeted agent might, therefore, already possess resistance to the drug in a minority of cells. Indeed, multiple examples of pre-existing subclonal resistance mutations to various molecularly targeted agents have been described, which we review herein. Early detection of pre-existing or emerging drug resistance could enable more personalized use of targeted cancer therapy, as patients could be stratified to receive the therapies that are most likely to be effective. We consider how monitoring of drug resistance could be incorporated into clinical practice to optimize the use of targeted therapies in individual patients.
Collapse
Affiliation(s)
- Michael W Schmitt
- Departments of Biochemistry and Pathology, University of Washington, 1959 Northeast Pacific Street, Box 357705, Seattle, WA 98195, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Box 357705, Seattle, WA 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Box 19024, Seattle, WA 98109, USA
| | - Lawrence A Loeb
- Departments of Biochemistry and Pathology, University of Washington, 1959 Northeast Pacific Street, Box 357705, Seattle, WA 98195, USA
| | - Jesse J Salk
- Departments of Biochemistry and Pathology, University of Washington, 1959 Northeast Pacific Street, Box 357705, Seattle, WA 98195, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Box 357705, Seattle, WA 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Box 19024, Seattle, WA 98109, USA
| |
Collapse
|
20
|
Ta HQ, Ivey ML, Frierson HF, Conaway MR, Dziegielewski J, Larner JM, Gioeli D. Checkpoint Kinase 2 Negatively Regulates Androgen Sensitivity and Prostate Cancer Cell Growth. Cancer Res 2015; 75:5093-105. [PMID: 26573794 DOI: 10.1158/0008-5472.can-15-0224] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 09/12/2015] [Indexed: 12/18/2022]
Abstract
Prostate cancer is the second leading cause of cancer death in American men, and curing metastatic disease remains a significant challenge. Nearly all patients with disseminated prostate cancer initially respond to androgen deprivation therapy (ADT), but virtually all patients will relapse and develop incurable castration-resistant prostate cancer (CRPC). A high-throughput RNAi screen to identify signaling pathways regulating prostate cancer cell growth led to our discovery that checkpoint kinase 2 (CHK2) knockdown dramatically increased prostate cancer growth and hypersensitized cells to low androgen levels. Mechanistic investigations revealed that the effects of CHK2 were dependent on the downstream signaling proteins CDC25C and CDK1. Moreover, CHK2 depletion increased androgen receptor (AR) transcriptional activity on androgen-regulated genes, substantiating the finding that CHK2 affects prostate cancer proliferation, partly, through the AR. Remarkably, we further show that CHK2 is a novel AR-repressed gene, suggestive of a negative feedback loop between CHK2 and AR. In addition, we provide evidence that CHK2 physically associates with the AR and that cell-cycle inhibition increased this association. Finally, IHC analysis of CHK2 in prostate cancer patient samples demonstrated a decrease in CHK2 expression in high-grade tumors. In conclusion, we propose that CHK2 is a negative regulator of androgen sensitivity and prostate cancer growth, and that CHK2 signaling is lost during prostate cancer progression to castration resistance. Thus, perturbing CHK2 signaling may offer a new therapeutic approach for sensitizing CRPC to ADT and radiation.
Collapse
Affiliation(s)
- Huy Q Ta
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Melissa L Ivey
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Henry F Frierson
- Department of Pathology, University of Virginia Health System, Charlottesville, Virginia. Cancer Center Member, University of Virginia, Charlottesville, Virginia
| | - Mark R Conaway
- Cancer Center Member, University of Virginia, Charlottesville, Virginia. Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia
| | - Jaroslaw Dziegielewski
- Cancer Center Member, University of Virginia, Charlottesville, Virginia. Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia
| | - James M Larner
- Cancer Center Member, University of Virginia, Charlottesville, Virginia. Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia
| | - Daniel Gioeli
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia. Cancer Center Member, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
21
|
Beckman RA, Chen C. Translating predictive biomarkers within oncology clinical development programs. Biomark Med 2015; 9:851-62. [PMID: 26330133 DOI: 10.2217/bmm.15.56] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Predictive biomarkers provide essential information to enable personalized medicine, and hold the promise for enhancing the effectiveness and value of cancer therapies. However, they do not always work. This review provides a framework for managing the risk of predictive biomarkers and maximally harvesting their benefit. Methods are provided which permit data-driven, adaptive decision making about the use of predictive biomarkers during clinical development, applying them to the extent they are validated by the clinical data. Techniques for optimizing overall development efficiency, measured as the number of successful drug indications approved per patient utilized, are also presented.
Collapse
Affiliation(s)
- Robert A Beckman
- Departments of Oncology & Biostatistics, Bioinformatics & Biomathematics, Lombardi Comprehensive Cancer Center & Innovation Center for Biomedical Informatics, Georgetown University Medical Center, 4000 Reservoir Road NW, Suite 120 Washington, DC 20007, USA
| | - Cong Chen
- Biostatistics & Research Decision Sciences, Merck Research Laboratories, Rahway, NJ, USA
| |
Collapse
|
22
|
Kumar Y, Yang J, Hu T, Chen L, Xu Z, Xu L, Hu XX, Tang G, Wang JM, Li Y, Poon WS, Wan W, Zhang L, Mat WK, Pun FW, Lee P, Cheong THY, Ding X, Ng SK, Tsang SY, Chen JF, Zhang P, Li S, Wang HY, Xue H. Massive interstitial copy-neutral loss-of-heterozygosity as evidence for cancer being a disease of the DNA-damage response. BMC Med Genomics 2015. [PMID: 26208496 PMCID: PMC4515014 DOI: 10.1186/s12920-015-0104-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background The presence of loss-of-heterozygosity (LOH) mutations in cancer cell genomes is commonly encountered. Moreover, the occurrences of LOHs in tumor suppressor genes play important roles in oncogenesis. However, because the causative mechanisms underlying LOH mutations in cancer cells yet remain to be elucidated, enquiry into the nature of these mechanisms based on a comprehensive examination of the characteristics of LOHs in multiple types of cancers has become a necessity. Methods We performed next-generation sequencing on inter-Alu sequences of five different types of solid tumors and acute myeloid leukemias, employing the AluScan platform which entailed amplification of such sequences using multiple PCR primers based on the consensus sequences of Alu elements; as well as the whole genome sequences of a lung-to-liver metastatic cancer and a primary liver cancer. Paired-end sequencing reads were aligned to the reference human genome to identify major and minor alleles so that the partition of LOH products between homozygous-major vs. homozygous-minor alleles could be determined at single-base resolution. Strict filtering conditions were employed to avoid false positives. Measurements of LOH occurrences in copy number variation (CNV)-neutral regions were obtained through removal of CNV-associated LOHs. Results We found: (a) average occurrence of copy-neutral LOHs amounting to 6.9 % of heterologous loci in the various cancers; (b) the mainly interstitial nature of the LOHs; and (c) preference for formation of homozygous-major over homozygous-minor, and transitional over transversional, LOHs. Conclusions The characteristics of the cancer LOHs, observed in both AluScan and whole genome sequencings, point to the formation of LOHs through repair of double-strand breaks by interhomolog recombination, or gene conversion, as the consequence of a defective DNA-damage response, leading to a unified mechanism for generating the mutations required for oncogenesis as well as the progression of cancer cells. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0104-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yogesh Kumar
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Jianfeng Yang
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Taobo Hu
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Lei Chen
- Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China.
| | - Zhi Xu
- Department of Oncology, Nanjing First Hospital, and Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Lin Xu
- Jiangsu Key Laboratory of Cancer Molecular Biology and Translational Medicine, Jiangsu Cancer Hospital, Nanjing, China.
| | - Xiao-Xia Hu
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Gusheng Tang
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Jian-Min Wang
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Yi Li
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China.
| | - Wai-Sang Poon
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China.
| | - Weiqing Wan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China.
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China.
| | - Wai-Kin Mat
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Frank W Pun
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Peggy Lee
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Timothy H Y Cheong
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Xiaofan Ding
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Siu-Kin Ng
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Shui-Ying Tsang
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| | - Jin-Fei Chen
- Department of Oncology, Nanjing First Hospital, and Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Peng Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST, and Department of Automation, Tsinghua University, Beijing, 100084, China.
| | - Shao Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, TNLIST, and Department of Automation, Tsinghua University, Beijing, 100084, China.
| | - Hong-Yang Wang
- Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, China.
| | - Hong Xue
- Division of Life Science, Applied Genomics Centre and Centre for Statistical Science, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| |
Collapse
|
23
|
Ciprotti M, Tebbutt NC, Lee FT, Lee ST, Gan HK, McKee DC, O'Keefe GJ, Gong SJ, Chong G, Hopkins W, Chappell B, Scott FE, Brechbiel MW, Tse AN, Jansen M, Matsumura M, Kotsuma M, Watanabe R, Venhaus R, Beckman RA, Greenberg J, Scott AM. Phase I Imaging and Pharmacodynamic Trial of CS-1008 in Patients With Metastatic Colorectal Cancer. J Clin Oncol 2015; 33:2609-16. [PMID: 26124477 DOI: 10.1200/jco.2014.60.4256] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE CS-1008 (tigatuzumab) is a humanized, monoclonal immunoglobulin G1 (IgG1) agonistic antibody to human death receptor 5. The purpose of this study was to investigate the impact of CS-1008 dose on the biodistribution, quantitative tumor uptake, and antitumor response in patients with metastatic colorectal cancer (mCRC). PATIENTS AND METHODS Patients with mCRC who had received at least one course of chemotherapy were assigned to one of five dosage cohorts and infused with a weekly dose of CS-1008. Day 1 and day 36 doses were trace-labeled with indium-111 ((111)In), followed by whole-body planar and regional single-photon emission computed tomography (SPECT) imaging at several time points over the course of 10 days. RESULTS Nineteen patients were enrolled. (111)In-CS-1008 uptake in tumor was observed in only 12 patients (63%). (111)In-CS-1008 uptake and pharmacokinetics were not affected by dose or repeated drug administration. (111)In-CS-1008 biodistribution showed gradual blood-pool clearance and no abnormal uptake in normal tissue. No anti-CS-1008 antibody development was detected. One patient achieved partial response (3.7 months duration), eight patients had stable disease, and 10 patients had progressive disease. Clinical benefit rate (stable disease + partial response) in patients with (111)In-CS-1008 uptake in tumor was 58% versus 28% in patients with no uptake. An analysis of individual lesions showed that lesions with antibody uptake were one third as likely to progress as those without antibody uptake (P = .07). Death-receptor-5 expression in archived tumor samples did not correlate with (111)In-CS-1008 uptake (P = .5) or tumor response (P = .6). CONCLUSION Death-receptor-5 imaging with (111)In-CS-1008 reveals interpatient and intrapatient heterogeneity of uptake in tumor, is not dose dependent, and is predictive of clinical benefit in the treatment of patients who have mCRC.
Collapse
Affiliation(s)
- Marika Ciprotti
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Niall C Tebbutt
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Fook-Thean Lee
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Sze-Ting Lee
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Hui K Gan
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - David C McKee
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Graeme J O'Keefe
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Sylvia J Gong
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Geoffrey Chong
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Wendie Hopkins
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Bridget Chappell
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Fiona E Scott
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Martin W Brechbiel
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Archie N Tse
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Mendel Jansen
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Manabu Matsumura
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Masakatsu Kotsuma
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Rira Watanabe
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Ralph Venhaus
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Robert A Beckman
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Jonathan Greenberg
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Andrew M Scott
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY.
| |
Collapse
|
24
|
Beckman RA, Chen C. Efficient, Adaptive Clinical Validation of Predictive Biomarkers in Cancer Therapeutic Development. ADVANCES IN CANCER BIOMARKERS 2015; 867:81-90. [DOI: 10.1007/978-94-017-7215-0_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
25
|
Li SC, Tachiki LML, Kabeer MH, Dethlefs BA, Anthony MJ, Loudon WG. Cancer genomic research at the crossroads: realizing the changing genetic landscape as intratumoral spatial and temporal heterogeneity becomes a confounding factor. Cancer Cell Int 2014; 14:115. [PMID: 25411563 PMCID: PMC4236490 DOI: 10.1186/s12935-014-0115-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 10/24/2014] [Indexed: 02/06/2023] Open
Abstract
The US National Cancer Institute (NCI) and the National Human Genome Research Institute (NHGRI) created the Cancer Genome Atlas (TCGA) Project in 2006. The TCGA's goal was to sequence the genomes of 10,000 tumors to identify common genetic changes among different types of tumors for developing genetic-based treatments. TCGA offered great potential for cancer patients, but in reality has little impact on clinical applications. Recent reports place the past TCGA approach of testing a small tumor mass at a single time-point at a crossroads. This crossroads presents us with the conundrum of whether we should sequence more tumors or obtain multiple biopsies from each individual tumor at different time points. Sequencing more tumors with the past TCGA approach of single time-point sampling can neither capture the heterogeneity between different parts of the same tumor nor catch the heterogeneity that occurs as a function of time, error rates, and random drift. Obtaining multiple biopsies from each individual tumor presents multiple logistical and financial challenges. Here, we review current literature and rethink the utility and application of the TCGA approach. We discuss that the TCGA-led catalogue may provide insights into studying the functional significance of oncogenic genes in reference to non-cancer genetic background. Different methods to enhance identifying cancer targets, such as single cell technology, real time imaging of cancer cells with a biological global positioning system, and cross-referencing big data sets, are offered as ways to address sampling discrepancies in the face of tumor heterogeneity. We predict that TCGA landmarks may prove far more useful for cancer prevention than for cancer diagnosis and treatment when considering the effect of non-cancer genes and the normal genetic background on tumor microenvironment. Cancer prevention can be better realized once we understand how therapy affects the genetic makeup of cancer over time in a clinical setting. This may help create novel therapies for gene mutations that arise during a tumor's evolution from the selection pressure of treatment.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
- />Department of Neurology, University of California Irvine School of Medicine, Irvine, CA 92697-4292 USA
- />Department of Biological Science, California State University, Fullerton, CA 92834 USA
| | - Lisa May Ling Tachiki
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
- />University of California Irvine School of Medicine, Irvine, CA 92697 USA
| | - Mustafa H Kabeer
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
- />Department of Pediatric Surgery, CHOC Children’s Hospital, 1201 West La Veta Ave, Orange, CA 92868 USA
- />Department of Surgery, University of California Irvine School of Medicine, 333 City Blvd. West, Suite 700, Orange, CA 92868 USA
| | - Brent A Dethlefs
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
| | | | - William G Loudon
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
- />Department of Neurological Surgery, Saint Joseph Hospital, Orange, CA 92868 USA
- />Department of Neurological Surgery, University of California Irvine School of Medicine, Orange, CA 92862 USA
- />Department of Biological Science, California State University, Fullerton, CA 92834 USA
| |
Collapse
|
26
|
Beckman RA, Yeang CH. Nonstandard personalized medicine strategies for cancer may lead to improved patient outcomes. Per Med 2014; 11:705-719. [PMID: 29764056 DOI: 10.2217/pme.14.57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer is an evolutionary process that is driven by mutation and selection. Tumors are genetically unstable, and research has shown that this is the most efficient way for cancers to evolve. Genetic instability leads to genetic heterogeneity and dynamic change within a single individual's tumor, in turn leading to therapeutic resistance. Cancer treatment has also evolved from an empirical science of killing dividing cells to the current era of 'personalized medicine', exquisitely targeting the molecular features of individual cancers. However, current personalized medicine regards a single individual's cancer as largely uniform and static. Moreover, from a strategic perspective, current personalized medicine thinks primarily of the immediate therapy selection. Ongoing research suggests that new, nonstandard personalized treatment strategies that plan further ahead and consider intratumoral heterogeneity and the evolving nature of cancer (due to genetic instability) may lead to the next level of therapeutic benefit beyond current personalized medicine.
Collapse
Affiliation(s)
- Robert A Beckman
- Center for Evolution & Cancer, Helen Diller Family Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - Chen-Hsiang Yeang
- Institute of Statistical Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, Taiwan
| |
Collapse
|
27
|
Trusina A. Stress induced telomere shortening: longer life with less mutations? BMC SYSTEMS BIOLOGY 2014; 8:27. [PMID: 24580844 PMCID: PMC4015310 DOI: 10.1186/1752-0509-8-27] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Accepted: 02/17/2014] [Indexed: 01/15/2023]
Abstract
Background Mutations accumulate as a result of DNA damage and imperfect DNA repair machinery. In higher eukaryotes the accumulation and spread of mutations is limited in two primary ways: through p53-mediated programmed cell death and cellular senescence mediated by telomeres. Telomeres shorten at every cell division and cell stops dividing once the shortest telomere reaches a critical length. It has been shown that the rate of telomere attrition is accelerated when cells are exposed to DNA damaging agents. However the implications of this mechanism are not fully understood. Results With the help of in silico model we investigate the effect of genotoxic stress on telomere attrition and apoptosis in a population of non-identical replicating cells. When comparing the populations of cells with constant vs. stress-induced rate of telomere shortening we find that stress induced telomere shortening (SITS) increases longevity while reducing mutation rate. Interestingly, however, the effect takes place only when genotoxic stresses (e.g. reactive oxygen species due to metabolic activity) are distributed non-equally among cells. Conclusions Our results for the first time show how non-equal distribution of metabolic load (and associated genotoxic stresses) combined with stress induced telomere shortening can delay aging and minimize mutations.
Collapse
Affiliation(s)
- Ala Trusina
- Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, DK 2100, Copenhagen, Denmark.
| |
Collapse
|
28
|
Abdel-Fatah TMA, Albarakati N, Bowell L, Agarwal D, Moseley P, Hawkes C, Ball G, Chan S, Ellis IO, Madhusudan S. Single-strand selective monofunctional uracil-DNA glycosylase (SMUG1) deficiency is linked to aggressive breast cancer and predicts response to adjuvant therapy. Breast Cancer Res Treat 2013; 142:515-27. [PMID: 24253812 DOI: 10.1007/s10549-013-2769-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 11/08/2013] [Indexed: 01/26/2023]
Abstract
Uracil in DNA is an important cause of mutagenesis. SMUG1 is a uracil-DNA glycosylase that removes uracil through base excision repair. SMUG1 also processes radiation-induced oxidative base damage as well as 5-fluorouracil incorporated into DNA during chemotherapy. We investigated SMUG1 mRNA expression in 249 primary breast cancers. SMUG1 protein expression was investigated in 1,165 breast tumours randomised into two cohorts [training set (n = 583) and test set (n = 582)]. SMUG1 and chemotherapy response was also investigated in a series of 315 ER-negative tumours (n = 315). For mechanistic insights, SMUG1 was correlated to biomarkers of aggressive phenotype, DNA repair, cell cycle and apoptosis. Low SMUG1 mRNA expression was associated with adverse disease specific survival (p = 0.008) and disease-free survival (p = 0.008). Low SMUG1 protein expression (25 %) was associated with high histological grade (p < 0.0001), high mitotic index (p < 0.0001), pleomorphism (p < 0.0001), glandular de-differentiation (p = 0.0001), absence of hormonal receptors (ER-/PgR-/AR) (p < 0.0001), presence of basal-like (p < 0.0001) and triple-negative phenotypes (p < 0.0001). Low SMUG1 protein expression was associated with loss of BRCA1 (p < 0.0001), ATM (p < 0.0001) and XRCC1 (p < 0.0001). Low p27 (p < 0.0001), low p21 (p = 0.023), mutant p53 (p = 0.037), low MDM2 (p < 0.0001), low MDM4 (p = 0.004), low Bcl-2 (p = 0.001), low Bax (p = 0.003) and high MIB1 (p < 0.0001) were likely in low SMUG1 tumours. Low SMUG1 protein expression was associated with poor prognosis in univariate (p < 0.001) and multivariate analysis (p < 0.01). In ER+ cohort that received adjuvant endocrine therapy, low SMUG1 protein expression remains associated with poor survival (p < 0.01). In ER- cohort that received adjuvant chemotherapy, low SMUG1 protein expression is associated with improved survival (p = 0.043). Our study suggests that low SMUG1 expression may correlate to adverse clinicopathological features and predict response to adjuvant therapy in breast cancer.
Collapse
|
29
|
Holbek S, Bendtsen KM, Juul J. Moderate stem-cell telomere shortening rate postpones cancer onset in a stochastic model. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2013; 88:042706. [PMID: 24229212 DOI: 10.1103/physreve.88.042706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 09/24/2013] [Indexed: 06/02/2023]
Abstract
Mammalian cells are restricted from proliferating indefinitely. Telomeres at the end of each chromosome are shortened at cell division and when they reach a critical length, the cell will enter permanent cell cycle arrest-a state known as senescence. This mechanism is thought to be tumor suppressing, as it helps prevent precancerous cells from dividing uncontrollably. Stem cells express the enzyme telomerase, which elongates the telomeres, thereby postponing senescence. However, unlike germ cells and most types of cancer cells, stem cells only express telomerase at levels insufficient to fully maintain the length of their telomeres, leading to a slow decline in proliferation potential. It is not yet fully understood how this decline influences the risk of cancer and the longevity of the organism. We here develop a stochastic model to explore the role of telomere dynamics in relation to both senescence and cancer. The model describes the accumulation of cancerous mutations in a multicellular organism and creates a coherent theoretical framework for interpreting the results of several recent experiments on telomerase regulation. We demonstrate that the longest average cancer-free lifespan before cancer onset is obtained when stem cells start with relatively long telomeres that are shortened at a steady rate at cell division. Furthermore, the risk of cancer early in life can be reduced by having a short initial telomere length. Finally, our model suggests that evolution will favor a shorter than optimal average cancer-free lifespan in order to postpone cancer onset until late in life.
Collapse
Affiliation(s)
- Simon Holbek
- University of Copenhagen, Niels Bohr Institute, Blegdamsvej 17, DK-2100 Copenhagen, Denmark
| | | | | |
Collapse
|
30
|
Gentry SN, Jackson TL. A mathematical model of cancer stem cell driven tumor initiation: implications of niche size and loss of homeostatic regulatory mechanisms. PLoS One 2013; 8:e71128. [PMID: 23990931 PMCID: PMC3747196 DOI: 10.1371/journal.pone.0071128] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/28/2013] [Indexed: 12/23/2022] Open
Abstract
Hierarchical organized tissue structures, with stem cell driven cell differentiation, are critical to the homeostatic maintenance of most tissues, and this underlying cellular architecture is potentially a critical player in the development of a many cancers. Here, we develop a mathematical model of mutation acquisition to investigate how deregulation of the mechanisms preserving stem cell homeostasis contributes to tumor initiation. A novel feature of the model is the inclusion of both extrinsic and intrinsic chemical signaling and interaction with the niche to control stem cell self-renewal. We use the model to simulate the effects of a variety of types and sequences of mutations and then compare and contrast all mutation pathways in order to determine which ones generate cancer cells fastest. The model predicts that the sequence in which mutations occur significantly affects the pace of tumorigenesis. In addition, tumor composition varies for different mutation pathways, so that some sequences generate tumors that are dominated by cancerous cells with all possible mutations, while others are primarily comprised of cells that more closely resemble normal cells with only one or two mutations. We are also able to show that, under certain circumstances, healthy stem cells diminish due to the displacement by mutated cells that have a competitive advantage in the niche. Finally, in the event that all homeostatic regulation is lost, exponential growth of the cancer population occurs in addition to the depletion of normal cells. This model helps to advance our understanding of how mutation acquisition affects mechanisms that influence cell-fate decisions and leads to the initiation of cancers.
Collapse
Affiliation(s)
- Sara N. Gentry
- Department of Mathematics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Trachette L. Jackson
- Department of Mathematics, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
31
|
Ruchirawat M, Cebulska-Wasilewska A, Au WW. Evidence for exposure-induced DNA repair abnormality is indicative of health and genetic risk. Int J Hyg Environ Health 2013; 216:566-73. [PMID: 23545294 DOI: 10.1016/j.ijheh.2013.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 03/03/2013] [Accepted: 03/07/2013] [Indexed: 01/06/2023]
Abstract
A recent focus has been targeted toward the development of functional biomarkers that can be used to predict disease more reliably. One such biomarker is the challenge assay for DNA repair deficiency. Briefly, the assay involves challenging lymphocytes in culture to a DNA damaging agent in vitro and determining the repair outcome in chromosome aberrations and/or DNA strand breaks. The aim is to show that individuals who have chronic exposure to toxic substances will develop exposure-induced DNA repair deficiencies. Many studies around the world have shown that the assay detects DNA repair deficiency in environmentally/occupationally exposed populations and with significant exposure dose-response relationship. The prediction of health risk was also validated. In addition, exposure-induced repair deficiency which was apparently passed through the germ cells had caused genetic consequences in a 3-generation population. The assay is simple to conduct and is more sensitive than some traditional biomarker assays. Together with the functional significance of the assay, the challenge assay can be used with confidence in population studies for health risk assessment.
Collapse
|
32
|
Shull AY, Clendenning ML, Ghoshal-Gupta S, Farrell CL, Vangapandu HV, Dudas L, Wilkerson BJ, Buckhaults PJ. Somatic mutations, allele loss, and DNA methylation of the Cub and Sushi Multiple Domains 1 (CSMD1) gene reveals association with early age of diagnosis in colorectal cancer patients. PLoS One 2013; 8:e58731. [PMID: 23505554 PMCID: PMC3591376 DOI: 10.1371/journal.pone.0058731] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/05/2013] [Indexed: 01/01/2023] Open
Abstract
Background The Cub and Sushi Multiple Domains 1 (CSMD1) gene, located on the short arm of chromosome 8, codes for a type I transmembrane protein whose function is currently unknown. CSMD1 expression is frequently lost in many epithelial cancers. Our goal was to characterize the relationships between CSMD1 somatic mutations, allele imbalance, DNA methylation, and the clinical characteristics in colorectal cancer patients. Methods We sequenced the CSMD1 coding regions in 54 colorectal tumors using the 454FLX pyrosequencing platform to interrogate 72 amplicons covering the entire coding sequence. We used heterozygous SNP allele ratios at multiple CSMD1 loci to determine allelic balance and infer loss of heterozygosity. Finally, we performed methylation-specific PCR on 76 colorectal tumors to determine DNA methylation status for CSMD1 and known methylation targets ALX4, RUNX3, NEUROG1, and CDKN2A. Results Using 454FLX sequencing and confirming with Sanger sequencing, 16 CSMD1 somatic mutations were identified in 6 of the 54 colorectal tumors (11%). The nonsynonymous to synonymous mutation ratio of the 16 somatic mutations was 15∶1, a ratio significantly higher than the expected 2∶1 ratio (p = 0.014). This ratio indicates a presence of positive selection for mutations in the CSMD1 protein sequence. CSMD1 allelic imbalance was present in 19 of 37 informative cases (56%). Patients with allelic imbalance and CSMD1 mutations were significantly younger (average age, 41 years) than those without somatic mutations (average age, 68 years). The majority of tumors were methylated at one or more CpG loci within the CSMD1 coding sequence, and CSMD1 methylation significantly correlated with two known methylation targets ALX4 and RUNX3. C:G>T:A substitutions were significantly overrepresented (47%), suggesting extensive cytosine methylation predisposing to somatic mutations. Conclusions Deep amplicon sequencing and methylation-specific PCR reveal that CSMD1 alterations can correlate with earlier clinical presentation in colorectal tumors, thus further implicating CSMD1 as a tumor suppressor gene.
Collapse
Affiliation(s)
- Austin Y. Shull
- Georgia Health Sciences University Cancer Center, Georgia Health Sciences University, Augusta, Georgia, United States of America
- Department of Biochemistry and Cancer Biology, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Megan L. Clendenning
- Georgia Health Sciences University Cancer Center, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Sampa Ghoshal-Gupta
- Georgia Health Sciences University Cancer Center, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Christopher L. Farrell
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, South Carolina, United States of America
| | - Hima V. Vangapandu
- Graduate School of Biomedical Sciences, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Larry Dudas
- Northeast Georgia Medical Center, Department of Internal Medicine, Gainesville, Georgia, United States of America
| | - Brent J. Wilkerson
- Department of Otolaryngology-Head and Neck Surgery, University of California Davis Medical Center, Sacramento, California, United States of America
| | - Phillip J. Buckhaults
- Georgia Health Sciences University Cancer Center, Georgia Health Sciences University, Augusta, Georgia, United States of America
- Department of Biochemistry and Cancer Biology, Georgia Health Sciences University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
33
|
S Datta R, Gutteridge A, Swanton C, Maley CC, Graham TA. Modelling the evolution of genetic instability during tumour progression. Evol Appl 2013; 6:20-33. [PMID: 23396531 PMCID: PMC3567468 DOI: 10.1111/eva.12024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 09/28/2012] [Indexed: 12/19/2022] Open
Abstract
The role of genetic instability in driving carcinogenesis remains controversial. Genetic instability should accelerate carcinogenesis by increasing the rate of advantageous driver mutations; however, genetic instability can also potentially retard tumour growth by increasing the rate of deleterious mutation. As such, it is unclear whether genetically unstable clones would tend to be more selectively advantageous than their genetically stable counterparts within a growing tumour. Here, we show the circumstances where genetic instability evolves during tumour progression towards cancer. We employ a Wright-Fisher type model that describes the evolution of tumour subclones. Clones can acquire both advantageous and deleterious mutations, and mutator mutations that increase a cell's intrinsic mutation rate. Within the model, cancers evolve with a mutator phenotype when driver mutations bestow only moderate increases in fitness: very strong or weak selection for driver mutations suppresses the evolution of a mutator phenotype. Genetic instability occurs secondarily to selectively advantageous driver mutations. Deleterious mutations have relatively little effect on the evolution of genetic instability unless selection for additional driver mutations is very weak or if deleterious mutations are very common. Our model provides a framework for studying the evolution of genetic instability in tumour progression. Our analysis highlights the central role of selection in shaping patterns of mutation in carcinogenesis.
Collapse
Affiliation(s)
- Ruchira S Datta
- Center for Evolution and Cancer, University of California San Francisco San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
34
|
Sultana R, Abdel-Fatah T, Abbotts R, Hawkes C, Albarakati N, Seedhouse C, Ball G, Chan S, Rakha EA, Ellis IO, Madhusudan S. Targeting XRCC1 deficiency in breast cancer for personalized therapy. Cancer Res 2012; 73:1621-34. [PMID: 23253910 DOI: 10.1158/0008-5472.can-12-2929] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
XRCC1 is a key component of DNA base excision repair, single strand break repair, and backup nonhomologous end-joining pathway. XRCC1 (X-ray repair cross-complementing gene 1) deficiency promotes genomic instability, increases cancer risk, and may have clinical application in breast cancer. We investigated XRCC1 expression in early breast cancers (n = 1,297) and validated in an independent cohort of estrogen receptor (ER)-α-negative breast cancers (n = 281). Preclinically, we evaluated XRCC1-deficient and -proficient Chinese hamster and human cancer cells for synthetic lethality application using double-strand break (DSB) repair inhibitors [KU55933 (ataxia telangectasia-mutated; ATM inhibitor) and NU7441 (DNA-PKcs inhibitor)]. In breast cancer, loss of XRCC1 (16%) was associated with high grade (P < 0.0001), loss of hormone receptors (P < 0.0001), triple-negative (P < 0.0001), and basal-like phenotypes (P = 0.001). Loss of XRCC1 was associated with a two-fold increase in risk of death (P < 0.0001) and independently with poor outcome (P < 0.0001). Preclinically, KU55933 [2-(4-Morpholinyl)-6-(1-thianthrenyl)-4H-pyran-4-one] and NU7441 [8-(4-Dibenzothienyl)-2-(4-morpholinyl)-4H-1-benzopyran-4-one] were synthetically lethal in XRCC1-deficient compared with proficient cells as evidenced by hypersensitivity to DSB repair inhibitors, accumulation of DNA DSBs, G2-M cell-cycle arrest, and induction of apoptosis. This is the first study to show that XRCC1 deficiency in breast cancer results in an aggressive phenotype and that XRCC1 deficiency could also be exploited for a novel synthetic lethality application using DSB repair inhibitors. Cancer Res; 73(5); 1621-34. ©2012 AACR.
Collapse
Affiliation(s)
- Rebeka Sultana
- Laboratory of Molecular Oncology, Academic Unit of Oncology, School of Molecular Medical Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Prindle MJ, Loeb LA. DNA polymerase delta in DNA replication and genome maintenance. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2012; 53:666-82. [PMID: 23065663 PMCID: PMC3694620 DOI: 10.1002/em.21745] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 09/09/2012] [Accepted: 09/12/2012] [Indexed: 05/12/2023]
Abstract
The eukaryotic genome is in a constant state of modification and repair. Faithful transmission of the genomic information from parent to daughter cells depends upon an extensive system of surveillance, signaling, and DNA repair, as well as accurate synthesis of DNA during replication. Often, replicative synthesis occurs over regions of DNA that have not yet been repaired, presenting further challenges to genomic stability. DNA polymerase δ (pol δ) occupies a central role in all of these processes: catalyzing the accurate replication of a majority of the genome, participating in several DNA repair synthetic pathways, and contributing structurally to the accurate bypass of problematic lesions during translesion synthesis. The concerted actions of pol δ on the lagging strand, pol ϵ on the leading strand, associated replicative factors, and the mismatch repair (MMR) proteins results in a mutation rate of less than one misincorporation per genome per replication cycle. This low mutation rate provides a high level of protection against genetic defects during development and may prevent the initiation of malignancies in somatic cells. This review explores the role of pol δ in replication fidelity and genome maintenance.
Collapse
Affiliation(s)
- Marc J Prindle
- Department of Pathology, The Joseph Gottstien Memorial Cancer Research Laboratory, University of Washington, Seattle, WA 98195-7705, USA
| | | |
Collapse
|
36
|
Abstract
DNA sequencing studies have established that many cancers contain tens of thousands of clonal mutations throughout their genomes, which is difficult to reconcile with the very low rate of mutation in normal human cells. This observation provides strong evidence for the mutator phenotype hypothesis, which proposes that a genome-wide elevation in the spontaneous mutation rate is an early step in carcinogenesis. An elevated mutation rate implies that cancers undergo continuous evolution, generating multiple subpopulations of cells that differ from one another in DNA sequence. The extensive heterogeneity in DNA sequence and continual tumor evolution that would occur in the context of a mutator phenotype have important implications for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Michael W Schmitt
- Joseph Gottstein Memorial Cancer Research Laboratory, Department of Pathology, University of Washington, Seattle, Washington, USA
| | | | | |
Collapse
|
37
|
Impact of genetic dynamics and single-cell heterogeneity on development of nonstandard personalized medicine strategies for cancer. Proc Natl Acad Sci U S A 2012; 109:14586-91. [PMID: 22891318 DOI: 10.1073/pnas.1203559109] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancers are heterogeneous and genetically unstable. Current practice of personalized medicine tailors therapy to heterogeneity between cancers of the same organ type. However, it does not yet systematically address heterogeneity at the single-cell level within a single individual's cancer or the dynamic nature of cancer due to genetic and epigenetic change as well as transient functional changes. We have developed a mathematical model of personalized cancer therapy incorporating genetic evolutionary dynamics and single-cell heterogeneity, and have examined simulated clinical outcomes. Analyses of an illustrative case and a virtual clinical trial of over 3 million evaluable "patients" demonstrate that augmented (and sometimes counterintuitive) nonstandard personalized medicine strategies may lead to superior patient outcomes compared with the current personalized medicine approach. Current personalized medicine matches therapy to a tumor molecular profile at diagnosis and at tumor relapse or progression, generally focusing on the average, static, and current properties of the sample. Nonstandard strategies also consider minor subclones, dynamics, and predicted future tumor states. Our methods allow systematic study and evaluation of nonstandard personalized medicine strategies. These findings may, in turn, suggest global adjustments and enhancements to translational oncology research paradigms.
Collapse
|
38
|
Kelley MR, Georgiadis MM, Fishel ML. APE1/Ref-1 role in redox signaling: translational applications of targeting the redox function of the DNA repair/redox protein APE1/Ref-1. Curr Mol Pharmacol 2012; 5:36-53. [PMID: 22122463 DOI: 10.2174/1874467211205010036] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 08/18/2010] [Accepted: 08/25/2010] [Indexed: 12/22/2022]
Abstract
The heterogeneity of most cancers diminishes the treatment effectiveness of many cancer-killing regimens. Thus, treatments that hold the most promise are ones that block multiple signaling pathways essential to cancer survival. One of the most promising proteins in that regard is APE1, whose reduction-oxidation activity influences multiple cancer survival mechanisms, including growth, proliferation, metastasis, angiogenesis, and stress responses. With the continued research using APE1 redox specific inhibitors alone or coupled with developing APE1 DNA repair inhibitors it will now be possible to further delineate the role of APE1 redox, repair and protein-protein interactions. Previously, use of siRNA or over expression approaches, while valuable, do not give a clear picture of the two major functions of APE1 since both techniques severely alter the cellular milieu. Additionally, use of the redox-specific APE1 inhibitor, APX3330, now makes it possible to study how inhibition of APE1's redox signaling can affect multiple tumor pathways and can potentiate the effectiveness of existing cancer regimens. Because APE1 is an upstream effector of VEGF, as well as other molecules that relate to angiogenesis and the tumor microenvironment, it is also being studied as a possible treatment for agerelated macular degeneration and diabetic retinopathy. This paper reviews all of APE1's functions, while heavily focusing on its redox activities. It also discusses APE1's altered expression in many cancers and the therapeutic potential of selective inhibition of redox regulation, which is the subject of intense preclinical studies.
Collapse
Affiliation(s)
- Mark R Kelley
- Department of Pediatrics (Section of Hematology/Oncology), Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
39
|
Abstract
Recent data on DNA sequencing of human tumours have established that cancer cells contain thousands of mutations. These data support the concept that cancer cells express a mutator phenotype. This Perspective considers the evidence supporting the mutator phenotype hypothesis, the origin and consequences of a mutator phenotype, the implications for personalized medicine and the feasibility of ablating tumours by error catastrophe.
Collapse
Affiliation(s)
- Lawrence A Loeb
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA.
| |
Collapse
|
40
|
Satoh N, Maniwa Y, Bermudez VP, Nishimura K, Nishio W, Yoshimura M, Okita Y, Ohbayashi C, Hurwitz J, Hayashi Y. Oncogenic phosphatase Wip1 is a novel prognostic marker for lung adenocarcinoma patient survival. Cancer Sci 2011; 102:1101-6. [PMID: 21281403 PMCID: PMC11158119 DOI: 10.1111/j.1349-7006.2011.01898.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
DNA damage response pathways are important for maintaining genomic stability. The oncogenic phosphatase Wip1 plays a crucial role in DNA damage response by inhibiting several cell cycle proteins, including p53. Although Wip1 gene amplification has been reported in various primary tumors, including lung cancer, its biological significance for survival of primary lung tumor patients remains unclear. We investigated the expression of Wip1 in cancer epithelial cells immunohistochemically in 84 consecutive resected cases of lung adenocarcinoma. Increased Wip1 expression was observed in 54 (64.3%) of the 84 cases. Wip1 expression was found to be correlated significantly with two clinicopathological factors: γ-H2AX expression, and invasion to the pulmonary vein. A univariate analysis and log-rank test indicated a significant association between Wip1 expression and lower overall survival rate (P = 0.019 and P = 0.0099, respectively). A multivariate analysis also indicated a statistically significant association between increased Wip1 expression and lower overall survival rate (hazard ratio, 4.3; P = 0.026). The Ki67 index level was higher in the Wip1-positive group than in the negative group (P < 0.04, Mann-Whitney U-test). Moreover, in a subgroup analysis of only stage I patients, increased Wip1 expression was also significantly associated with a lower overall survival rate (P = 0.023, log-rank test). These results indicate that the increased expression of Wip1 in cancer epithelial cells has significant value for tumor progression and the clinical prognosis of patients with primary lung adenocarcinoma.
Collapse
Affiliation(s)
- Naoyuki Satoh
- Division of Molecular Medicine and Medical Genetics, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Recent advances in the research of hepatitis B virus-related hepatocellular carcinoma: epidemiologic and molecular biological aspects. Adv Cancer Res 2011; 108:21-72. [PMID: 21034965 DOI: 10.1016/b978-0-12-380888-2.00002-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the commonest cancers worldwide, and more than half of HCC patients are attributable to persistent hepatitis B virus (HBV) infections. The best and cheapest way to prevent HBV-related HCC is the implementation of universal hepatitis B vaccination program, by which the incidence rates of childhood HCC have been reduced in several countries, including Taiwan. However, there are still hundreds of millions of HBV carriers in the world that remain a global health challenge. In the past decade, several hepatitis B viral factors such as serum HBV DNA level, genotype, and naturally occurring mutants have already been identified to influence liver disease progression and HCC development in HBV carriers. Several easy-to-use scoring systems based on clinical and viral characteristics are developed to predict HCC risk in HBV carriers and may facilitate the communication between practicing physicians and patients in clinical practice. In addition, the role of nonviral factors in HBV-related HCC has also been increasingly recognized. On the basis of these emerging data, it is recommended that HBV carriers should be screened and monitored to identify those who have a higher risk of liver disease progression and require antiviral treatments. Regarding the molecular carcinogenesis of HCC development, despite some progress in the research of cell biology of HCC in the past decade, aberrant pathways involved in maintaining HCC phenotypes have not been completely elucidated yet. In the future, through comprehensive and integrated approaches to analyze the genomes of human HCC, novel target genes or pathways critically involved in hepatocarcinogenesis may hopefully be identified.
Collapse
|
42
|
Prindle MJ, Fox EJ, Loeb LA. The mutator phenotype in cancer: molecular mechanisms and targeting strategies. Curr Drug Targets 2011; 11:1296-303. [PMID: 20840072 DOI: 10.2174/1389450111007011296] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 03/01/2010] [Indexed: 02/04/2023]
Abstract
Normal human cells replicate their DNA with exceptional accuracy. It has been estimated that approximately one error occurs during DNA replication for each 10(9) to 10(10) nucleotides polymerized. In contrast, malignant cells exhibit multiple chromosomal abnormalities and contain tens of thousands of alterations in the nucleotide sequence of nuclear DNA. To account for the disparity between the rarity of mutations in normal cells and the large numbers of mutations present in cancer, we have hypothesized that during tumor development, cancer cells exhibit a mutator phenotype. As a defining feature of cancer, the mutator phenotype remains an as-yet unexplored therapeutic target: by reducing the rate at which mutations accumulate it may be possible to significantly delay tumor development; conversely, the large number of mutations in cancer may make cancer cells more sensitive to cell killing by increasing the mutation rate. Here we summarize the evidence for the mutator phenotype hypothesis in cancer and explore how the increased frequency of random mutations during the evolution of human tumors provides new approaches for the design of cancer chemotherapy.
Collapse
Affiliation(s)
- Marc J Prindle
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | | | | |
Collapse
|
43
|
Jinka R, Kapoor R, Pavuluri S, Raj AT, Kumar MJ, Rao L, Pande G. Differential gene expression and clonal selection during cellular transformation induced by adhesion deprivation. BMC Cell Biol 2010; 11:93. [PMID: 21122158 PMCID: PMC3012028 DOI: 10.1186/1471-2121-11-93] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 12/02/2010] [Indexed: 12/27/2022] Open
Abstract
Background Anchorage independent growth is an important hallmark of oncogenic transformation. Previous studies have shown that when adhesion dependent fibroblasts were prevented from adhering to a substrate they underwent anoikis. In the present study we have demonstrated how anoikis resistant cells gain the transformation related properties with sequential selection of genes. We have proposed this process as a model system for selection of transformed cells from normal cells. Results This report demonstrates that some fibroblasts can survive during late stages of anoikis, at which time they exhibit transformation-associated properties such as in vitro colony formation in soft agar and in vivo subcutaneous tumour formation in nude mice. Cytogenetic characterisation of these cells revealed that they contained a t (2; 2) derivative chromosome and they have a selective survival advantage in non adherent conditions. Gene expression profile indicated that these cells over expressed genes related to hypoxia, glycolysis and tumor suppression/metastasis which could be helpful in their retaining a transformed phenotype. Conclusion Our results reveal some new links between anoikis and cell transformation and they provide a reproducible model system which can potentially be useful to study multistage cancer and to identify new targets for drug development.
Collapse
Affiliation(s)
- Rajeswari Jinka
- Department of Biochemistry, Acharya Nagarjuna University, Guntur 522510,India
| | | | | | | | | | | | | |
Collapse
|
44
|
Tian T, Olson S, Whitacre JM, Harding A. The origins of cancer robustness and evolvability. Integr Biol (Camb) 2010; 3:17-30. [PMID: 20944865 DOI: 10.1039/c0ib00046a] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Unless diagnosed early, many adult cancers remain incurable diseases. This is despite an intense global research effort to develop effective anticancer therapies, calling into question the use of rational drug design strategies in targeting complex disease states such as cancer. A fundamental challenge facing researchers and clinicians is that cancers are inherently robust biological systems, able to survive, adapt and proliferate despite the perturbations resulting from anticancer drugs. It is essential that the mechanisms underlying tumor robustness be formally studied and characterized, as without a thorough understanding of the principles of tumor robustness, strategies to overcome therapy resistance are unlikely to be found. Degeneracy describes the ability of structurally distinct system components (e.g. proteins, pathways, cells, organisms) to be conditionally interchangeable in their contribution to system traits and it has been broadly implicated in the robustness and evolvability of complex biological systems. Here we focus on one of the most important mechanisms underpinning tumor robustness and degeneracy, the cellular heterogeneity that is the hallmark of most solid tumors. Based on a combination of computational, experimental and clinical studies we argue that stochastic noise is an underlying cause of tumor heterogeneity and particularly degeneracy. Drawing from a number of recent data sets, we propose an integrative model for the evolution of therapy resistance, and discuss recent computational studies that propose new therapeutic strategies aimed at defeating the adaptable cancer phenotype.
Collapse
|
45
|
Beckman RA. Efficiency of carcinogenesis: is the mutator phenotype inevitable? Semin Cancer Biol 2010; 20:340-52. [PMID: 20934514 DOI: 10.1016/j.semcancer.2010.10.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 10/01/2010] [Indexed: 11/15/2022]
Abstract
Cancer development requires multiple oncogenic mutations. Pathogenic mechanisms which accelerate this process may be favored carcinogenic pathways. Mutator mutations are mutations in genetic stability genes, and increase the mutation rate, speeding up the accumulation of oncogenic mutations. The mutator hypothesis states that mutator mutations play a critical role in carcinogenesis. Alternatively, tumors might arise by mutations occurring at the normal rate followed by selection and expansion of various premalignant lineages on the path to cancer. This alternative pathway is a significant argument against the mutator hypothesis. Mutator mutations may also lead to accumulation of deleterious mutations, which could lead to extinction of premalignant lineages before they become cancerous, another argument against the mutator hypothesis. Finally, the need for acquisition of a mutator mutation imposes an additional step on the carcinogenic process. Accordingly, the mutator hypothesis has been a seminal but controversial idea for several decades despite considerable experimental and theoretical work. To resolve this debate, the concept of efficiency has been introduced as a metric for comparing carcinogenic mechanisms, and a new theoretical approach of focused quantitative modeling has been applied. The results demonstrate that, given what is already known, the predominance of mutator mechanisms is likely inevitable, as they overwhelm less efficient non-mutator pathways to cancer.
Collapse
Affiliation(s)
- Robert A Beckman
- Department of Oncology Clinical Research, Daiichi Sankyo Pharmaceutical Development, Edison, NJ 08837, USA.
| |
Collapse
|
46
|
Schmitt MW, Venkatesan RN, Pillaire MJ, Hoffmann JS, Sidorova JM, Loeb LA. Active site mutations in mammalian DNA polymerase delta alter accuracy and replication fork progression. J Biol Chem 2010; 285:32264-72. [PMID: 20628184 DOI: 10.1074/jbc.m110.147017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
DNA polymerase δ (pol δ) is one of the two main replicative polymerases in eukaryotes; it synthesizes the lagging DNA strand and also functions in DNA repair. In previous work, we demonstrated that heterozygous expression of the pol δ L604G variant in mice results in normal life span and no apparent phenotype, whereas a different substitution at the same position, L604K, is associated with shortened life span and accelerated carcinogenesis. Here, we report in vitro analysis of the homologous mutations at position Leu-606 in human pol δ. Four-subunit human pol δ variants that harbor or lack 3' → 5'-exonucleolytic proofreading activity were purified from Escherichia coli. The pol δ L606G and L606K holoenzymes retain catalytic activity and processivity similar to that of wild type pol δ. pol δ L606G is highly error prone, incorporating single noncomplementary nucleotides at a high frequency during DNA synthesis, whereas pol δ L606K is extremely accurate, with a higher fidelity of single nucleotide incorporation by the active site than that of wild type pol δ. However, pol δ L606K is impaired in the bypass of DNA adducts, and the homologous variant in mouse embryonic fibroblasts results in a decreased rate of replication fork progression in vivo. These results indicate that different substitutions at a single active site residue in a eukaryotic polymerase can either increase or decrease the accuracy of synthesis relative to wild type and suggest that enhanced fidelity of base selection by a polymerase active site can result in impaired lesion bypass and delayed replication fork progression.
Collapse
Affiliation(s)
- Michael W Schmitt
- Department of Biochemistry, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | | | | | | | | | | |
Collapse
|
47
|
Fox EJ, Beckman RA, Loeb LA. Reply: Is There Any Genetic Instability in Human Cancer? DNA Repair (Amst) 2010; 9:859-860. [PMID: 20703319 DOI: 10.1016/j.dnarep.2010.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Edward J Fox
- Department of Pathology Box 357705, School of Medicine, K-072 HSB, University of Washington
| | | | | |
Collapse
|
48
|
Luo M, He H, Kelley MR, Georgiadis MM. Redox regulation of DNA repair: implications for human health and cancer therapeutic development. Antioxid Redox Signal 2010; 12:1247-69. [PMID: 19764832 PMCID: PMC2864659 DOI: 10.1089/ars.2009.2698] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Redox reactions are known to regulate many important cellular processes. In this review, we focus on the role of redox regulation in DNA repair both in direct regulation of specific DNA repair proteins as well as indirect transcriptional regulation. A key player in the redox regulation of DNA repair is the base excision repair enzyme apurinic/apyrimidinic endonuclease 1 (APE1) in its role as a redox factor. APE1 is reduced by the general redox factor thioredoxin, and in turn reduces several important transcription factors that regulate expression of DNA repair proteins. Finally, we consider the potential for chemotherapeutic development through the modulation of APE1's redox activity and its impact on DNA repair.
Collapse
Affiliation(s)
- Meihua Luo
- Department of Pediatrics (Section of Hematology/Oncology), Herman B. Wells Center for Pediatric Research, Indiana University, Indiana
| | - Hongzhen He
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indiana
| | - Mark R. Kelley
- Department of Pediatrics (Section of Hematology/Oncology), Herman B. Wells Center for Pediatric Research, Indiana University, Indiana
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indiana
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indiana
| | - Millie M. Georgiadis
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indiana
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana
| |
Collapse
|
49
|
Abstract
Although now dogma, the idea that nonvertebrate organisms such as yeast, worms, and flies could inform, and in some cases even revolutionize, our understanding of oncogenesis in humans was not immediately obvious. Aided by the conservative nature of evolution and the persistence of a cohort of devoted researchers, the role of model organisms as a key tool in solving the cancer problem has, however, become widely accepted. In this review, we focus on the nematode Caenorhabditis elegans and its diverse and sometimes surprising contributions to our understanding of the tumorigenic process. Specifically, we discuss findings in the worm that address a well-defined set of processes known to be deregulated in cancer cells including cell cycle progression, growth factor signaling, terminal differentiation, apoptosis, the maintenance of genome stability, and developmental mechanisms relevant to invasion and metastasis.
Collapse
Affiliation(s)
- Natalia V. Kirienko
- University of Wyoming, College of Agriculture, Department of Molecular Biology, Dept 3944, 1000 E. University Avenue, Laramie, WY 82071
| | - Kumaran Mani
- University of Wyoming, College of Agriculture, Department of Molecular Biology, Dept 3944, 1000 E. University Avenue, Laramie, WY 82071
| | - David S. Fay
- University of Wyoming, College of Agriculture, Department of Molecular Biology, Dept 3944, 1000 E. University Avenue, Laramie, WY 82071
| |
Collapse
|
50
|
Abstract
Cancer recapitulates Darwinian evolution. Mutations acquired during life that provide cells with a growth or survival advantage will preferentially multiply to form a tumor. As a result of The Cancer Genome Atlas Project, we have gathered detailed information on the nucleotide sequence changes in a number of human cancers. The sources of mutations in cancer are diverse, and the complexity of those found to be clonally present in tumors has increasingly made it difficult to identify key rate-limiting genes for tumor growth that could serve as potential targets for directed therapies. The impact of DNA sequencing on future cancer research and personalized therapy is likely to be profound and merits critical evaluation.
Collapse
Affiliation(s)
- Jesse J Salk
- Joseph Gottstein Memorial Cancer Research Laboratory, Department of Pathology, University of Washington, Seattle, Washington 98195, USA
| | | | | |
Collapse
|