1
|
Rodriguez A, Corchete LA, Alcazar JA, Montero JC, Rodriguez M, Chinchilla-Tábora LM, Vidal Tocino R, Moyano C, Muñoz-Bravo S, Sayagués JM, Abad M. Dysregulated Expression of Three Genes in Colorectal Cancer Stratifies Patients into Three Risk Groups. Cancers (Basel) 2022; 14:cancers14174076. [PMID: 36077612 PMCID: PMC9454483 DOI: 10.3390/cancers14174076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Despite advances in recent years in the study of the molecular profile of sporadic colorectal cancer (sCRC), the specific genetic events that lead to increased aggressiveness or the development of the metastatic process of tumours are not yet clear. In previous studies of the gene expression profile (GEP) using a high-density array (50,000 genes and 6000 miRNAs in a single assay) in sCRC tumours, we identified a 28-gene signature that was found to be associated with an adverse prognostic value for predicting patient survival. Here, we analyse the differential expression of these 28 genes for their possible association with tumour local aggressiveness and metastatic processes in 66 consecutive sCRC patients, followed for >5 years, using the NanoString nCounter platform. The global transcription profile (expression levels of the 28 genes studied simultaneously) allowed us to discriminate between sCRC tumours and nontumoral colonic tissues. Analysis of the biological and functional significance of the dysregulated GEPs observed in our sCRC tumours revealed 31 significantly altered canonical pathways. Among the most commonly altered pathways, we observed the increased expression of genes involved in signalling pathways and cellular processes, such as the PI3K-Akt pathway, the interaction with the extracellular matrix (ECM), and other functions related to cell signalling processes (SRPX2). From a prognostic viewpoint, the altered expression of BST2 and SRPX2 genes were the only independent variables predicting for disease-free survival (DFS). In addition to the pT stage at diagnosis, dysregulated transcripts of ADH1B, BST2, and FER1L4 genes showed a prognostic impact on OS in the multivariate analysis. Based on the altered expression of these three genes, a scoring system was built to stratify patients into low-, intermediate-, and high-risk groups with significantly different 5-year OS rates: 91%, 83%, and 52%, respectively. The prognostic impact was validated in two independent series of sCRC patients from the public GEO database (n = 562 patients). In summary, we show a strong association between the altered expression of three genes and the clinical outcome of sCRC patients, making them potential markers of suitability for adjuvant therapy after complete tumour resection. Additional prospective studies in larger series of patients are required to confirm the clinical utility of the newly identified biomarkers because the number of patients analysed remains small.
Collapse
Affiliation(s)
- Alba Rodriguez
- Department of Pathology and IBSAL, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Luís Antonio Corchete
- Cancer Research Center and Hematology Service, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - José Antonio Alcazar
- General and Gastrointestinal Surgery Service, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Juan Carlos Montero
- Department of Pathology and IBSAL, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Marta Rodriguez
- Department of Pathology and IBSAL, University Hospital of Salamanca, 37007 Salamanca, Spain
| | | | - Rosario Vidal Tocino
- Medical Oncology Service and IBSAL, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Carlos Moyano
- Clinical Biochemistry Service, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Saray Muñoz-Bravo
- Department of Pathology and IBSAL, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - José María Sayagués
- Department of Pathology and IBSAL, University Hospital of Salamanca, 37007 Salamanca, Spain
- Correspondence: (J.M.S.); (M.A.)
| | - Mar Abad
- Department of Pathology and IBSAL, University Hospital of Salamanca, 37007 Salamanca, Spain
- Correspondence: (J.M.S.); (M.A.)
| |
Collapse
|
2
|
Peng Y, Nie Y, Yu J, Wong CC. Microbial Metabolites in Colorectal Cancer: Basic and Clinical Implications. Metabolites 2021; 11:metabo11030159. [PMID: 33802045 PMCID: PMC8001357 DOI: 10.3390/metabo11030159] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading cancers that cause cancer-related deaths worldwide. The gut microbiota has been proved to show relevance with colorectal tumorigenesis through microbial metabolites. By decomposing various dietary residues in the intestinal tract, gut microbiota harvest energy and produce a variety of metabolites to affect the host physiology. However, some of these metabolites are oncogenic factors for CRC. With the advent of metabolomics technology, studies profiling microbiota-derived metabolites have greatly accelerated the progress in our understanding of the host-microbiota metabolism interactions in CRC. In this review, we briefly summarize the present metabolomics techniques in microbial metabolites researches and the mechanisms of microbial metabolites in CRC pathogenesis, furthermore, we discuss the potential clinical applications of microbial metabolites in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Yao Peng
- Department of Gastroenterology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, China; (Y.P.); (Y.N.)
| | - Yuqiang Nie
- Department of Gastroenterology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, China; (Y.P.); (Y.N.)
- Department of Gastroenterology, The Second Affiliated Hospital, Medical School, South China University of Technology, Guangzhou 510180, China
| | - Jun Yu
- Department of Gastroenterology, The Second Affiliated Hospital, Medical School, South China University of Technology, Guangzhou 510180, China
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Correspondence: (J.Y.); (C.C.W.)
| | - Chi Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Correspondence: (J.Y.); (C.C.W.)
| |
Collapse
|
3
|
Bhimani J, Ball K, Stebbing J. Patient-derived xenograft models-the future of personalised cancer treatment. Br J Cancer 2020; 122:601-602. [PMID: 31919403 PMCID: PMC7054515 DOI: 10.1038/s41416-019-0678-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 10/29/2019] [Accepted: 11/19/2019] [Indexed: 01/12/2023] Open
Abstract
For many tumours there is a lack of randomised data from which we can guide systemic treatments. Although gene expression profiling along with proteomics has led to advances in diagnosis, classification and prognosis, our ability to target many cancers has been further limited due to a lack of therapeutic options. The use of patient-derived xenograft (PDX) models in the setting of a rare malignancy is discussed here by Kamili et al, with the successful establishment of new model systems.
Collapse
Affiliation(s)
- Jenna Bhimani
- Department of Oncology, Charing Cross Hospital, Imperial College and Imperial College Healthcare NHS Trust, Fulham Palace Road, London, W6 8RF, UK.
| | - Katie Ball
- Department of Oncology, Charing Cross Hospital, Imperial College and Imperial College Healthcare NHS Trust, Fulham Palace Road, London, W6 8RF, UK
| | - Justin Stebbing
- Department of Oncology, Charing Cross Hospital, Imperial College and Imperial College Healthcare NHS Trust, Fulham Palace Road, London, W6 8RF, UK.,Imperial College Centre for Translational and Experimental Medicine, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
4
|
Chen Z, Jiang L. The clinical application of fruquintinib on colorectal cancer. Expert Rev Clin Pharmacol 2019; 12:713-721. [PMID: 31177854 DOI: 10.1080/17512433.2019.1630272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Zhongguang Chen
- Department of Pharmaceutical, Central Hospital of Linyi City, Yishui, Shandong, China
| | - Lili Jiang
- Ultrasound Medical Department, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
5
|
Susman S, Berindan-Neagoe I, Petrushev B, Pirlog R, Florian IS, Mihu CM, Berce C, Craciun L, Grewal R, Tomuleasa C. The role of the pathology department in the preanalytical phase of molecular analyses. Cancer Manag Res 2018; 10:745-753. [PMID: 29695931 PMCID: PMC5903845 DOI: 10.2147/cmar.s150851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
After introducing the new molecules for the treatment of patients with tumoral pathology, the therapeutical decision will be taken depending on the molecular profile performed upon the harvested tissues. This major modification makes the molecular and morphological analysis an essential part in the clinical management of patients and the pathologist plays an important role in this process. The quality and reproducibility of the results are imperative today and they depend on both the reliability of the molecular techniques and the quality of the tissue we use in the process. Also, the genomics and proteomics techniques, used increasingly often, require high-quality tissues, and pathology laboratories play a very significant role in the management of all phases of this process. In this paper the parameters which must be followed in order to obtain optimal results within the techniques which analyze nucleic acids and proteins were reviewed.
Collapse
Affiliation(s)
- Sergiu Susman
- Department of Pathology, Imogen Research Center.,Department of Morphological Sciences
| | | | - Bobe Petrushev
- Research Center for Functional Genomics and Translational Medicine
| | | | - Ioan-Stefan Florian
- Department of Neurosurgery, Iuliu Hatieganu University of Medicine and Pharmacy
| | | | - Cristian Berce
- Research Center for Functional Genomics and Translational Medicine
| | | | - Ravnit Grewal
- Department of Hematology, Ion Chiricuta Oncology Institute
| | - Ciprian Tomuleasa
- Research Center for Functional Genomics and Translational Medicine.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Haematopathology, Tygerberg Academic Hospital, Tygerberg, South Africa
| |
Collapse
|
6
|
Sayagués JM, Corchete LA, Gutiérrez ML, Sarasquete ME, Del Mar Abad M, Bengoechea O, Fermiñán E, Anduaga MF, Del Carmen S, Iglesias M, Esteban C, Angoso M, Alcazar JA, García J, Orfao A, Muñoz-Bellvis L. Genomic characterization of liver metastases from colorectal cancer patients. Oncotarget 2018; 7:72908-72922. [PMID: 27662660 PMCID: PMC5341953 DOI: 10.18632/oncotarget.12140] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/13/2016] [Indexed: 01/09/2023] Open
Abstract
Metastatic dissemination is the most frequent cause of death of sporadic colorectal cancer (sCRC) patients. Genomic abnormalities which are potentially characteristic of such advanced stages of the disease are complex and so far, they have been poorly described and only partially understood. We evaluated the molecular heterogeneity of sCRC tumors based on simultaneous assessment of the overall GEP of both coding mRNA and non-coding RNA genes in primary sCRC tumor samples from 23 consecutive patients and their paired liver metastases. Liver metastases from the sCRC patients analyzed, systematically showed deregulated transcripts of those genes identified as also deregulated in their paired primary colorectal carcinomas. However, some transcripts were found to be specifically deregulated in liver metastases (vs. non-tumoral colorectal tissues) while expressed at normal levels in their primary tumors, reflecting either an increased genomic instability of metastatic cells or theiradaption to the liver microenvironment. Newly deregulated metastatic transcripts included overexpression of APOA1, HRG, UGT2B4, RBP4 and ADH4 mRNAS and the miR-3180-3p, miR-3197, miR-3178, miR-4793 and miR-4440 miRNAs, together with decreased expression of the IGKV1-39, IGKC, IGKV1-27, FABP4 and MYLK mRNAS and the miR-363, miR-1, miR-143, miR-27b and miR-28-5p miRNAs. Canonical pathways found to be specifically deregulated in liver metastatic samples included multiple genes related with intercellular adhesion and the metastatic processes (e.g., IGF1R, PIK3CA, PTEN and EGFR), endocytosis (e.g., the PDGFRA, SMAD2, ERBB3, PML and FGFR2), and the cell cycle (e.g., SMAD2, CCND2, E2F5 and MYC). Our results also highlighted the activation of genes associated with the TGFβ signaling pathway, -e.g. RHOA, SMAD2, SMAD4, SMAD5, SMAD6, BMPR1A, SMAD7 and MYC-, which thereby emerge as candidate genes to play an important role in CRC tumor metastasis.
Collapse
Affiliation(s)
- José María Sayagués
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center, IBMCC-CSIC/USAL and IBSAL, University of Salamanca, Salamanca, Spain
| | - Luís Antonio Corchete
- Cáncer Research Center and Service of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - María Laura Gutiérrez
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center, IBMCC-CSIC/USAL and IBSAL, University of Salamanca, Salamanca, Spain
| | - Maria Eugenia Sarasquete
- Cáncer Research Center and Service of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - María Del Mar Abad
- Department of Pathology, University Hospital of Salamanca, Salamanca, Spain
| | - Oscar Bengoechea
- Department of Pathology, University Hospital of Salamanca, Salamanca, Spain
| | - Encarna Fermiñán
- Genomics Unit, Cancer Research Center, IBMCC-CSIC/USAL, Salamanca, Spain
| | - María Fernanda Anduaga
- Service of General and Gastrointestinal Surgery and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Sofia Del Carmen
- Department of Pathology, University Hospital of Salamanca, Salamanca, Spain
| | - Manuel Iglesias
- Service of General and Gastrointestinal Surgery and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Carmen Esteban
- Service of General and Gastrointestinal Surgery and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - María Angoso
- Service of General and Gastrointestinal Surgery and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Jose Antonio Alcazar
- Service of General and Gastrointestinal Surgery and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Jacinto García
- Service of General and Gastrointestinal Surgery and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Alberto Orfao
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center, IBMCC-CSIC/USAL and IBSAL, University of Salamanca, Salamanca, Spain
| | - Luís Muñoz-Bellvis
- Service of General and Gastrointestinal Surgery and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| |
Collapse
|
7
|
Zhang L. Personalized medicine and blood disorders. Per Med 2016; 13:587-596. [PMID: 29754548 DOI: 10.2217/pme-2016-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Personalized medicine has been using genomics approaches to elucidate the etiology of a disease as well as to personalize the management for patients of a particular disease based on that individual's genetic features. It benefits patients across a multitude of therapeutic areas and advancements are particularly evident in hematology/oncology. The importance of genomics discoveries and development in nonmalignant blood disorders generally goes unrecognized, but it becomes critical now due to the global disease burden and a high mortality. This paper focuses on the exploration of personalized medicine applications in hemoglobin diseases, and thrombotic and bleeding disorders. It discusses the challenges which slow down the implementation as well. The available data suggest that the ability to understand the clinical features of a patient's genetic profile and the knowledge of disease mechanisms are the keys to facilitate new diagnosis, new therapies, new prescriptions and better healthcare.
Collapse
Affiliation(s)
- Li Zhang
- Clinical Research Hematology, Baxalta, Inc., Cambridge, MA 02142, USA
| |
Collapse
|
8
|
Hendlisz A, Bleiberg H. Too little benefit for too much toxicity: rethinking the adjuvant development model in colon cancer. Curr Opin Oncol 2016; 28:345-7. [PMID: 27138572 DOI: 10.1097/cco.0000000000000298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Alain Hendlisz
- Department of Medicine, Digestive Oncology Unit, Medical Oncology, Institut Jules Bordet, ULB, Brussels, Belgium
| | | |
Collapse
|
9
|
Hayashi H, Beppu T, Sakamoto Y, Miyamoto Y, Yokoyama N, Higashi T, Nitta H, Hashimoto D, Chikamoto A, Baba H. Prognostic value of Ki-67 expression in conversion therapy for colorectal liver-limited metastases. Am J Cancer Res 2015; 5:1225-1233. [PMID: 26046001 PMCID: PMC4449450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 12/25/2014] [Indexed: 06/04/2023] Open
Abstract
INTRODUCTION The objective of this study was to examine the prognostic value of Ki-67 expression in conversion therapy for colorectal liver-confined metastases. METHODS We enrolled a total of 96 patients including 54 patients who received oxaliplatin- or irinotecan-based chemotherapy and curative hepatectomy for initially unresectable metastases (conversion group) and 42 patients with initially resectable liver metastases (straight hepatectomy group). Ki-67 expression was examined in 96 resected specimens but excluded the 2 specimens that revealed no residual cancer cells in conversion group. RESULTS Conversion therapy leads to greater survival that is equivalent to that straight hepatectomy group. In conversion group, high Ki-67 expression (> 30%) levels were detectable in 33 patients (64%) after chemotherapy prior to conversion therapy. High Ki-67 expression was significantly associated with shorter disease-free survival and worse overall survival (P < 0.01 in both), and was an independent worse prognostic factor of disease-free survival and overall survival (hazard ratio [HR] and P-values were 5.608, 0.001 and 5.366, 0.04, respectively) in patients with conversion therapy. Interestingly, even in the patients with RECIST PR (n = 32), high Ki-67 expression was significantly shorter disease-free survival compared to low Ki-67 expression (P < 0.001). In contrast to conversion group, there was no significant difference in disease free survival and overall survival between low (n = 14, 33%) and high (n = 28, 67%) Ki-67 expressions in patients with straight hepatectomy (P = 0.14 and 0.74, respectively). CONCLUSIONS Residual Ki-67 expression is a useful biomarker for worse prognostic outcomes after conversion therapy. High Ki-67 expression may be a biomarker of micrometastases containing aggressive cancer cells.
Collapse
Affiliation(s)
- Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Toru Beppu
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Yasuo Sakamoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Naomi Yokoyama
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Takaaki Higashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hidetoshi Nitta
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Daisuke Hashimoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Akira Chikamoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University 1-1-1 Honjo, Kumamoto 860-8556, Japan
| |
Collapse
|
10
|
Baker JJ, Meyers MO, Deal AM, Frank JF, Stitzenberg KB, Ollila DW. Prognostic significance of tumor mitotic rate in T2 melanoma staged with sentinel lymphadenectomy. J Surg Oncol 2015; 111:711-5. [DOI: 10.1002/jso.23880] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 12/06/2014] [Indexed: 12/29/2022]
Affiliation(s)
| | - Michael O. Meyers
- Division of Surgical Oncology; Department of Surgery; University of North Carolina-Chapel Hill; North Carolina
- Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; North Carolina
| | - Allison M. Deal
- Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; North Carolina
- Biostatistics Core Facility; University of North Carolina-Chapel Hill; North Carolina
| | - Jill F. Frank
- Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; North Carolina
| | - Karyn B. Stitzenberg
- Division of Surgical Oncology; Department of Surgery; University of North Carolina-Chapel Hill; North Carolina
- Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; North Carolina
| | - David W. Ollila
- Division of Surgical Oncology; Department of Surgery; University of North Carolina-Chapel Hill; North Carolina
- Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; North Carolina
| |
Collapse
|
11
|
Abstract
Significant progress has been made in understanding the molecular genetic basis of colorectal cancer (CRC). That information is paving the way to understanding the genetic basis of other tumors, as well. Oncology nurses should anticipate the routine integration of this information and testing of CRC tumors to understand the molecular basis of the disease in clinical practice. Molecular testing can lead to the identification of families at risk for hereditary cancer syndromes, particularly Lynch syndrome, which sometimes is referred to as hereditary nonpolyposis colorectal cancer. Knowledge of the genetic basis of CRC also contributes valuable information aimed at selecting appropriate and effective targeted therapy.
Collapse
Affiliation(s)
- Suzanne M Mahon
- Department of Internal Medicine and in the School of Nursing, Saint Louis University in Missouri
| |
Collapse
|
12
|
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
|
13
|
Yasui H, Tsurita G, Imai K. DNA synthesis inhibitors for the treatment of gastrointestinal cancer. Expert Opin Pharmacother 2014; 15:2361-72. [PMID: 25256052 DOI: 10.1517/14656566.2014.958074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Intensive laboratory, preclinical and clinical studies have identified and validated molecular targets in cancers, leading to a shift toward the development of novel, rationally designed and specific therapeutic agents. However, gastrointestinal cancers continue to have a poor prognosis, largely due to drug resistance. AREAS COVERED Here, we discuss the current understanding of DNA synthesis inhibitors and their mechanisms of action for the treatment of gastrointestinal malignancies. EXPERT OPINION Conventional agents, including DNA synthesis inhibitors such as fluoropyrimidines and platinum analogs, remain the most effective therapeutics and are the standards against which new drugs are compared. Novel DNA synthesis inhibitors for the treatment of gastrointestinal malignancies include a combination of the antimetabolite TAS-102, which consists of trifluorothymidine with a thymidine phosphorylase inhibitor, and a novel micellar formulation of cisplatin NC-6004 that uses a nanotechnology-based drug delivery system. The challenges of translational cancer research using DNA synthesis inhibitors include the identification of drugs that are specific to tumor cells to reduce toxicity and increase antitumor efficacy, biomarkers to predict pharmacological responses to chemotherapeutic drugs, identification of ways to overcome drug resistance and development of novel combination therapies with DNA synthesis inhibitors and other cancer therapies, such as targeted molecular therapeutics. Here, we discuss the current understanding of DNA synthesis inhibitors and their mechanisms of action for the treatment of gastrointestinal malignancies.
Collapse
Affiliation(s)
- Hiroshi Yasui
- The University of Tokyo, The Institute of Medical Science, Center for Antibody and Vaccine , 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639 , Japan
| | | | | |
Collapse
|
14
|
Bank PC, Swen JJ, Guchelaar HJ. Pharmacogenetic biomarkers for predicting drug response. Expert Rev Mol Diagn 2014; 14:723-35. [PMID: 24857685 DOI: 10.1586/14737159.2014.923759] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Drug response shows significant interpatient variability and evidence that genetics influences outcome of drug therapy has been known for more than five decades. However, the translation of this knowledge to clinical practice remains slow. Using examples from clinical practice six considerations about the implementation of pharmacogenetics (PGx) into routine care are discussed: the need for PGx biomarkers; the sources of genetic variability in drug response; the amount of variability explained by PGx; whether PGx test results are actionable; the level of evidence needed for implementation of PGx and the sources of information regarding interpretation of PGx data.
Collapse
Affiliation(s)
- Paul Christiaan Bank
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | | | | |
Collapse
|
15
|
The impact of Aldehyde dehydrogenase 1 expression on prognosis for metastatic colon cancer. J Surg Res 2014; 192:82-9. [PMID: 24953984 DOI: 10.1016/j.jss.2014.05.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/20/2014] [Accepted: 05/16/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cancer stem cells may be associated with tumor progression and prognosis for colon cancer. We hypothesized that expression of Aldehyde dehydrogenase 1 (ALDH1) would increase with tumor progression and be associated with survival. METHODS Tissue was obtained from resection specimens for isolation of cancer stem cells. In addition, paraffin blocks from resected colon cancers with normal colon, primary tumor, and lymph node and liver metastasis from 2000 to 2010 were identified and stained with ALDH1. RESULTS In in vitro models (adherent and tumor spheres) ALHD1+ cells grew more efficiently than ALDH1- cells. ALDH1 expression was highest in peritumoral crypt cells (0.137 μm(2), 95% confidence interval [CI] 0.125-0.356) and normal crypts (median 0.091 μm(2), 95% CI 0.064-0.299) followed by lymph node metastasis (median 0.025 μm(2), 95% CI 0-0.131) and the primary cancers (median 0.014 μm(2), 95% CI 0.0123-0.154). Samples were divided into high and low ALDH1 expression. Survival was associated with expression in the primary tumor (9 versus 23 mo, P = 0.0016) expression but not peritumoral tissue (21 versus 20.5 mo, P = 0.32), normal colon (19 versus 27 mo, P = 0.289), or lymph node metastasis (23 versus 21 mo, P = 0.69). On univariate analysis, ALDH1 expression and grade were associated with survival but ages, number of lymph node metastasis, race, or grade were not associated. On multivariate analysis, only ALDH1 status continued to be associated with survival, odds ratio 4.4, and P = 0.011. CONCLUSIONS ALDH1 is indicative of stemness and is a biomarker marker in colon cancer. Expression did not increase with progression from normal colon to primary tumors and metastasis.
Collapse
|
16
|
Clinical prognostic value of combined analysis of Aldh1, Survivin, and EpCAM expression in colorectal cancer. Br J Cancer 2014; 110:2935-44. [PMID: 24786601 PMCID: PMC4056050 DOI: 10.1038/bjc.2014.226] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/28/2014] [Accepted: 04/08/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Tumour aggressiveness might be related to the degree of main cancer hallmark acquirement of tumour cells, reflected by expression levels of specific biomarkers. We investigated the expression of Aldh1, Survivin, and EpCAM, together reflecting main cancer hallmarks, in relation to clinical outcome of colorectal cancer (CRC) patients. METHODS Immunohistochemistry was performed using a tumour tissue microarray of TNM (Tumour, Node, Metastasis)-stage I-IV CRC tissues. Single-marker expression or their combination was assessed for associations with the clinical outcome of CRC patients (N=309). RESULTS Increased expression of Aldh1 or Survivin, or decreased expression of EpCAM was each associated with poor clinical outcome, and was therefore identified as clinically unfavourable expression. Analyses of the combination of all three markers showed worse clinical outcome, specifically in colon cancer patients, with an increasing number of markers showing unfavourable expression. Hazard ratios ranged up to 8.3 for overall survival (P<0.001), 36.6 for disease-specific survival (P<0.001), and 27.1 for distant recurrence-free survival (P<0.001). CONCLUSIONS Our data identified combined expression levels of Aldh1, Survivin, and EpCAM as strong independent prognostic factors, with high hazard ratios, for survival and tumour recurrence in colon cancer patients, and therefore reflect tumour aggressiveness.
Collapse
|
17
|
Wong A, Ma BBY. Personalizing therapy for colorectal cancer. Clin Gastroenterol Hepatol 2014; 12:139-44. [PMID: 24025538 DOI: 10.1016/j.cgh.2013.08.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/26/2013] [Accepted: 08/26/2013] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide. Several important scientific discoveries in the molecular biology of CRC have changed clinical practice in oncology. These included the comprehensive genome-wide profiling of CRC by the Cancer Genome Atlas Network, the discovery of mutations along the RAS-RAF signaling pathway as major determinants of response to antibodies against the epidermal growth factor receptor, the elucidation of new molecular subsets of CRC or gene signatures that may predict clinical outcome after adjuvant chemotherapy, and the innovative targeting of the family of vascular endothelial growth factor and receptors. These new data have allowed oncologists to individualize drug therapy on the basis of a patient's tumor's unique molecular profile, especially in the management of metastatic CRC. This review article will discuss the progress of personalized medicine in the contemporary management of CRC.
Collapse
Affiliation(s)
- Ashley Wong
- Sir Y. K. Pao Centre for Cancer, Department of Clinical Oncology, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Brigette B Y Ma
- Sir Y. K. Pao Centre for Cancer, Department of Clinical Oncology, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
18
|
Yothers G, O'Connell MJ, Lee M, Lopatin M, Clark-Langone KM, Millward C, Paik S, Sharif S, Shak S, Wolmark N. Validation of the 12-gene colon cancer recurrence score in NSABP C-07 as a predictor of recurrence in patients with stage II and III colon cancer treated with fluorouracil and leucovorin (FU/LV) and FU/LV plus oxaliplatin. J Clin Oncol 2013; 31:4512-9. [PMID: 24220557 PMCID: PMC3871512 DOI: 10.1200/jco.2012.47.3116] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Accurate assessments of recurrence risk and absolute treatment benefit are needed to inform colon cancer adjuvant therapy. The 12-gene Recurrence Score assay has been validated in patients with stage II colon cancer from the Cancer and Leukemia Group B 9581 and Quick and Simple and Reliable (QUASAR) trials. We conducted an independent, prospectively designed clinical validation study of Recurrence Score, with prespecified end points and analysis plan, in archival specimens from patients with stage II and III colon cancer randomly assigned to fluorouracil (FU) or FU plus oxaliplatin in National Surgical Adjuvant Breast and Bowel Project C-07. METHODS Recurrence Score was assessed in 892 fixed, paraffin-embedded tumor specimens (randomly selected 50% of patients with tissue). Data were analyzed by Cox regression adjusting for stage and treatment. RESULTS Continuous Recurrence Score predicted recurrence (hazard ratio for a 25-unit increase in score, 1.96; 95% CI, 1.50 to 2.55; P < .001), as well as disease-free and overall survival (both P < .001). Recurrence Score predicted recurrence risk (P = .001) after adjustment for stage, mismatch repair, nodes examined, grade, and treatment. Recurrence Score did not have significant interaction with stage (P = .90) or age (P = .76). Relative benefit of oxaliplatin was similar across the range of Recurrence Score (interaction P = .48); accordingly, absolute benefit of oxaliplatin increased with higher scores, most notably in patients with stage II and IIIA/B disease. CONCLUSION The 12-gene Recurrence Score predicts recurrence risk in stage II and stage III colon cancer and provides additional information beyond conventional clinical and pathologic factors. Incorporating Recurrence Score into the clinical context may better inform adjuvant therapy decisions in stage III as well as stage II colon cancer.
Collapse
Affiliation(s)
- Greg Yothers
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| | - Michael J. O'Connell
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| | - Mark Lee
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| | - Margarita Lopatin
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| | - Kim M. Clark-Langone
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| | - Carl Millward
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| | - Soonmyung Paik
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| | - Saima Sharif
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| | - Steven Shak
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| | - Norman Wolmark
- Greg Yothers, Michael J. O'Connell, Soonmyung Paik, Saima Sharif, and Norman Wolmark, National Surgical Adjuvant Breast and Bowel Project (NSABP) Operations and Biostatistical Centers; Greg Yothers, University of Pittsburgh Graduate School of Public Health; Saima Sharif and Norman Wolmark, Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA; Mark Lee, Margarita Lopatin, Kim M. Clark-Langone, and Steven Shak, Genomic Health, Redwood City, CA
| |
Collapse
|
19
|
Affiliation(s)
- Mark R Wick
- Departments of Pathology, University of Virginia Health System, Charlottesville, VA.
| | | | | |
Collapse
|
20
|
F-18 FDG PET/CT in the assessment of patients with unexplained CEA rise after surgical curative resection for colorectal cancer. Int J Colorectal Dis 2013; 28:1699-705. [PMID: 23846517 DOI: 10.1007/s00384-013-1747-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2013] [Indexed: 02/04/2023]
Abstract
PURPOSE We evaluated the role of quantitative assessment by maximum standardized uptake value (SUVmax) on F-18 fluorodeoxyglucose [F-18]FDG positron emission tomography/computed tomography (PET/CT) in stratifying colorectal cancer (CRC) patients with unexplained carcinoembryonic antigen (CEA) rise after surgical curative resection. MATERIAL AND METHODS Forty asymptomatic patients (mean age, 64 ± 12 years) with previous CRC and current serum CEA levels >5 ng/ml underwent [F-18] FDG PET/CT 13 ± 3 months after complete surgical resection. The SUVmax was registered on anastomosis and peri-anastomotic tissue lesions, if present. The patients were followed for 24 ± 9 months thereafter. Re-intervention, evidence of newly discovered distant metastases, and death were recognized as main events and constituted surrogate end points. The receiver-operator-curve (ROC) analysis was performed to estimate the optimal SUVmax cut-off to predict patients at high risk of main events. PET/CT results were then related to disease outcome (overall survival; OS). RESULTS The mean SUVmax at the anastomotic site was 6.2 ± 3 (range 2.6-15). At multivariate logistic regression analysis, the anastomotic SUVmax remained as the only significant contributor to the prediction of the events (p = 0.004; OR 1.97). The ROC analysis recognized that the optimal threshold of SUVmax to differentiate patients was 5.7. A worse OS was observed in patients presenting with a SUVmax greater than 5.7 as compared to those having lesser (median survival: 16 vs. 31 months; p = 0.002). CONCLUSIONS The quantitative assessment by SUVmax on [F-18]FDG PET/CT may be helpful in patients presenting with unexplained CEA rise after curative resection of CRC, by identifying those at risk of main events.
Collapse
|
21
|
Qin R, Kohli M. Pharmacogenetics- and pharmacogenomics-based rational clinical trial designs in oncology. Per Med 2013; 10:859-869. [PMID: 29776282 DOI: 10.2217/pme.13.78] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The rapid evolution of molecular technologies that can identify genetic markers and lead to dissecting the inherent variance of individual cancer biology has had a tangible impact on trial designs in oncology. Rational trial designs based on molecular marker expression coupled with drug–marker interactions have started to be adopted, challenging the previous paradigms of morphology-based, single-arm efficacy studies. This review summarizes novel trials being developed based on molecular predictive factor therapeutics and the potential impact these novel trial designs will have on the practice of oncology in future. A variety of clinical trial designs based on tumor and drug–host genetic interactions are discussed and the example of advanced prostate cancer is used to illustrate the changing landscape of clinical trial designs in cancer.
Collapse
Affiliation(s)
- Rui Qin
- Department of Health Sciences Research, Mayo Clinic, 200 First Street South West, Rochester, MN 55905, USA
| | - Manish Kohli
- Department of Oncology, Mayo Clinic, 200 First Street South West, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Maspin is a marker for early recurrence in primary stage III and IV colorectal cancer. Br J Cancer 2013; 109:1636-47. [PMID: 24002600 PMCID: PMC3776998 DOI: 10.1038/bjc.2013.489] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 07/26/2013] [Accepted: 07/30/2013] [Indexed: 12/12/2022] Open
Abstract
Background: Little is known about the factors that drive metastasis formation in colorectal cancer (CRC). Here, we set out to identify genes and proteins in patients with colorectal liver metastases that correlate with early disease recurrence. Such factors may predict a propensity for metastasis in earlier stages of CRC. Methods: Gene expression profiling and proteomics were used to identify differentially expressed genes/proteins in resected liver metastases that recurred within 6 months following liver surgery vs those that did not recur for >24 months. Expression of the identified genes/proteins in stage II (n=243) and III (n=176) tumours was analysed by immunohistochemistry on tissue microarrays. Correlation of protein levels with stage-specific outcome was assessed by uni- and multivariable analyses. Results: Both gene expression profiling and proteomics identified Maspin to be differentially expressed in colorectal liver metastases with early (<6 months) and prolonged (>24 months) time to recurrence. Immunohistochemical analysis of Maspin expression on tumour sections revealed that it was an independent predictor of time to recurrence (log-rank P=0.004) and CRC-specific survival (P=0.000) in stage III CRC. High Maspin expression was also correlated with mucinous differentiation. In stage II CRC patients, high Maspin expression did not correlate with survival but was correlated with a right-sided tumour location. Conclusion: High Maspin expression correlates with poor outcome in CRC after spread to the local lymph nodes. Therefore, Maspin may have a stage-specific function possibly related to tumour cell dissemination and/or metastatic outgrowth.
Collapse
|
23
|
Hlubocky FJ, Webster K, Beaumont J, Cashy J, Paul D, Abernethy A, Syrjala KL, Von Roenn J, Cella D. A preliminary study of a health related quality of life assessment of priority symptoms in advanced lymphoma: the National Comprehensive Cancer Network-Functional Assessment of Cancer Therapy - Lymphoma Symptom Index. Leuk Lymphoma 2013; 54:1942-6. [PMID: 23320888 PMCID: PMC5889924 DOI: 10.3109/10428194.2012.762977] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Despite the recent advances in cancer therapeutics for lymphoma (Lym), a continuum of disease, treatment and psychological challenges, adversely impacting health-related quality of life, remain for the clinical management of the patient with Lym. In response, this study presents the development and validation of the National Comprehensive Cancer Network-Functional Assessment of Cancer Therapy (NCCN-FACT) Lymphoma Symptom Index-18 (FLymSI-18). Patients with advanced Lym (n = 50) rated the significance of 40 symptoms, and hematologist-oncologists (n = 10) rated these symptoms according to importance and disease-related or treatment-related origin. Patient symptom priorities were unified with clinician priorities for symptom measurement in Lym for instrument development. Reliability estimates indicate that FLymSI-18 has acceptable internal consistency (α = 0.87), content validity and concurrent validity as indicated by moderate to strong correlations with the FACIT (Functional Assessment of Chronic Illness Therapy). Overall, the FLymSI-18 provides evidence for its reliability and validity as a brief assessment of the most important symptoms associated with advanced Lym in the clinical trial research environment.
Collapse
Affiliation(s)
- Fay J Hlubocky
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Northwestern Medical Faculty Foundation, Evanston, IL, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Venook AP, Niedzwiecki D, Lopatin M, Ye X, Lee M, Friedman PN, Frankel W, Clark-Langone K, Millward C, Shak S, Goldberg RM, Mahmoud NN, Warren RS, Schilsky RL, Bertagnolli MM. Biologic determinants of tumor recurrence in stage II colon cancer: validation study of the 12-gene recurrence score in cancer and leukemia group B (CALGB) 9581. J Clin Oncol 2013; 31:1775-81. [PMID: 23530100 PMCID: PMC3641698 DOI: 10.1200/jco.2012.45.1096] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE A greater understanding of the biology of tumor recurrence should improve adjuvant treatment decision making. We conducted a validation study of the 12-gene recurrence score (RS), a quantitative assay integrating stromal response and cell cycle gene expression, in tumor specimens from patients enrolled onto Cancer and Leukemia Group B (CALGB) 9581. PATIENTS AND METHODS CALGB 9581 randomly assigned 1,713 patients with stage II colon cancer to treatment with edrecolomab or observation and found no survival difference. The analysis reported here included all patients with available tissue and recurrence (n = 162) and a random (approximately 1:3) selection of nonrecurring patients. RS was assessed in 690 formalin-fixed paraffin-embedded tumor samples with quantitative reverse transcriptase polymerase chain reaction by using prespecified genes and a previously validated algorithm. Association of RS and recurrence was analyzed by weighted Cox proportional hazards regression. RESULTS Continuous RS was significantly associated with risk of recurrence (P = .013) as was mismatch repair (MMR) gene deficiency (P = .044). In multivariate analyses, RS was the strongest predictor of recurrence (P = .004), independent of T stage, MMR, number of nodes examined, grade, and lymphovascular invasion. In T3 MMR-intact (MMR-I) patients, prespecified low and high RS groups had average 5-year recurrence risks of 13% (95% CI, 10% to 16%) and 21% (95% CI, 16% to 26%), respectively. CONCLUSION The 12-gene RS predicts recurrence in stage II colon cancer in CALGB 9581. This is consistent with the importance of stromal response and cell cycle gene expression in colon tumor recurrence. RS appears to be most discerning for patients with T3 MMR-I tumors, although markers such as grade and lymphovascular invasion did not add value in this subset of patients.
Collapse
Affiliation(s)
- Alan P Venook
- University of California San Francisco Cancer Center, 1600 Divisadero Street, San Francisco, CA 94115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ross W, Lynch P, Raju G, Rodriguez A, Burke T, Hafemeister L, Hawk E, Wu X, Dubois RN, Mishra L. Biomarkers, bundled payments, and colorectal cancer care. Genes Cancer 2012; 3:16-22. [PMID: 22893787 DOI: 10.1177/1947601912448958] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 04/29/2012] [Indexed: 01/17/2023] Open
Abstract
Changes in the management of cancers such as colorectal cancer (CRC) are urgently needed, as such cancers continue to be one of the most commonly diagnosed cancers; CRC accounts for 21% of all cancers and is responsible for mortalities second only to lung cancer in the United States. A comprehensive science-driven approach towards markedly improved early detection/screening to efficacious targeted therapeutics with clear diagnostic and prognostic markers is essential. In addition, further changes addressing rising costs, stemming from recent health care reform measures, will be brought about in part by changes in how care is reimbursed. For oncology, the advances in genomics and biomarkers have the potential to define subsets of patients who have a prognosis or response to a particular type of therapy that differs from the mean. Better definition of a cancer's behavior will facilitate developing care plans tailored to the patient. One method under study is episode-based payment or bundling, where one payment is made to a provider organization to cover all expenses associated with a discrete illness episode. Payments will be based on the average cost of care, with providers taking on a risk for overutilization and outliers. For providers to thrive in this environment, they will need to know what care a patient will require and the costs of that care. A science-driven "personalized approach" to cancer care has the potential to produce better outcomes with reductions in the use of ineffectual therapies and costs. This promising scenario is still in the future, but progress is being made, and the shape of things to come for cancer care in the age of genomics is becoming clearer.
Collapse
Affiliation(s)
- William Ross
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Verma M. Personalized medicine and cancer. J Pers Med 2012; 2:1-14. [PMID: 25562699 PMCID: PMC4251363 DOI: 10.3390/jpm2010001] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Revised: 01/18/2012] [Accepted: 01/21/2012] [Indexed: 12/31/2022] Open
Abstract
Cancer is one of the leading causes of death in the United States, and more than 1.5 million new cases and more than 0.5 million deaths were reported during 2010 in the United States alone. Following completion of the sequencing of the human genome, substantial progress has been made in characterizing the human epigenome, proteome, and metabolome; a better understanding of pharmacogenomics has been developed, and the potential for customizing health care for the individual has grown tremendously. Recently, personalized medicine has mainly involved the systematic use of genetic or other information about an individual patient to select or optimize that patient’s preventative and therapeutic care. Molecular profiling in healthy and cancer patient samples may allow for a greater degree of personalized medicine than is currently available. Information about a patient’s proteinaceous, genetic, and metabolic profile could be used to tailor medical care to that individual’s needs. A key attribute of this medical model is the development of companion diagnostics, whereby molecular assays that measure levels of proteins, genes, or specific mutations are used to provide a specific therapy for an individual’s condition by stratifying disease status, selecting the proper medication, and tailoring dosages to that patient’s specific needs. Additionally, such methods can be used to assess a patient’s risk factors for a number of conditions and to tailor individual preventative treatments. Recent advances, challenges, and future perspectives of personalized medicine in cancer are discussed.
Collapse
Affiliation(s)
- Mukesh Verma
- Epidemiology and Genetics Research Program (EGRP), Division of Cancer Control and Population Sciences, National Cancer Institute (NCI), National Institutes of Health (NIH), 6130 Executive Boulevard, Rockville, MD 20852, USA.
| |
Collapse
|
27
|
Shuster JR, Lance RS, Troyer DA. Molecular preservation by extraction and fixation, mPREF: a method for small molecule biomarker analysis and histology on exactly the same tissue. BMC Clin Pathol 2011; 11:14. [PMID: 22188997 PMCID: PMC3280163 DOI: 10.1186/1472-6890-11-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Accepted: 12/21/2011] [Indexed: 02/06/2023] Open
Abstract
Background Histopathology is the standard method for cancer diagnosis and grading to assess aggressiveness in clinical biopsies. Molecular biomarkers have also been described that are associated with cancer aggressiveness, however, the portion of tissue analyzed is often processed in a manner that is destructive to the tissue. We present here a new method for performing analysis of small molecule biomarkers and histology in exactly the same biopsy tissue. Methods Prostate needle biopsies were taken from surgical prostatectomy specimens and first fixed, each in a separate vial, in 2.5 ml of 80% methanol:water. The biopsies were fixed for 24 hrs at room temperature and then removed and post-processed using a non-formalin-based fixative (UMFIX), embedded, and analyzed by hematoxylin and eosin (H&E) and by immunohistochemical (IHC) staining. The retained alcohol pre-fixative was analyzed for small molecule biomarkers by mass spectrometry. Results H&E analysis was successful following the pre-fixation in 80% methanol. The presence or absence of tumor could be readily determined for all 96 biopsies analyzed. A subset of biopsy sections was analyzed by IHC, and cancerous and non-cancerous regions could be readily visualized by PIN4 staining. To demonstrate the suitability for analysis of small molecule biomarkers, 28 of the alcohol extracts were analyzed using a mass spectrometry-based metabolomics platform. All extracts tested yielded successful metabolite profiles. 260 named biochemical compounds were detected in the alcohol extracts. A comparison of the relative levels of compounds in cancer containing vs. non-cancer containing biopsies showed differences for 83 of the compounds. A comparison of the results with prior published reports showed good agreement between the current method and prior reported biomarker discovery methods that involve tissue destructive methods. Conclusions The Molecular Preservation by Extraction and Fixation (mPREF) method allows for the analysis of small molecule biomarkers from exactly the same tissue that is processed for histopathology.
Collapse
Affiliation(s)
- Jeffrey R Shuster
- Depts, Of Pathology and Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, USA.
| | | | | |
Collapse
|
28
|
Pilati P, Mocellin S, Bertazza L, Galdi F, Briarava M, Mammano E, Tessari E, Zavagno G, Nitti D. Prognostic Value of Putative Circulating Cancer Stem Cells in Patients Undergoing Hepatic Resection for Colorectal Liver Metastasis. Ann Surg Oncol 2011; 19:402-8. [DOI: 10.1245/s10434-011-2132-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Indexed: 12/11/2022]
|
29
|
Cordero OJ, Imbernon M, Chiara LD, Martinez-Zorzano VS, Ayude D, de la Cadena MP, Rodriguez-Berrocal FJ. Potential of soluble CD26 as a serum marker for colorectal cancer detection. World J Clin Oncol 2011; 2:245-61. [PMID: 21773075 PMCID: PMC3139035 DOI: 10.5306/wjco.v2.i6.245] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 03/28/2011] [Accepted: 04/05/2011] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is characterized by a low survival rate even though the basis for colon cancer development, which involves the evolution of adenomas to carcinoma, is known. Moreover, the mortality rates continue to rise in economically transitioning countries although there is the opportunity to intervene in the natural history of the adenoma–cancer sequence through risk factors, screening, and treatment. Screening in particular accounted for most of the decline in colorectal cancer mortality achieved in the USA during the period 1975-2000. Patients show a better prognosis when the neoplasm is diagnosed early. Among the variety of screening strategies, the methods range from invasive and costly procedures such as colonoscopy to more low-cost and non-invasive tests such as the fecal occult blood test (guaiac and immunochemical). As a non-invasive biological serum marker would be of great benefit because of the performance of the test, several biomarkers, including cytologic assays, DNA and mRNA, and soluble proteins, have been studied. We found that the soluble CD26 (sCD26) concentration is diminished in serum of colorectal cancer patients compared to healthy donors, suggesting the potential utility of a sCD26 immunochemical detection test for early diagnosis. sCD26 originates from plasma membrane CD26 lacking its transmembrane and cytoplasmic domains. Some 90%–95% of sCD26 has been associated with serum dipeptidyl peptidase IV (DPP-IV) activity. DPP-IV, assigned to the CD26 cluster, is a pleiotropic enzyme expressed mainly on epithelial cells and lymphocytes. Our studies intended to validate this test for population screening to detect colorectal cancer and advanced adenomas are reviewed here.
Collapse
Affiliation(s)
- Oscar J Cordero
- Oscar J Cordero, Monica Imbernon, Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, School of Biology, CIBUS Building, Campus Vida, 15782 Santiago de Compostela, Spain
| | | | | | | | | | | | | |
Collapse
|