151
|
Sousa DA, Carneiro M, Ferreira D, Moreira FTC, Sales MGFV, Rodrigues LR. Recent advances in the selection of cancer-specific aptamers for the development of biosensors. Curr Med Chem 2022; 29:5850-5880. [PMID: 35209816 DOI: 10.2174/0929867329666220224155037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/30/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022]
Abstract
An early diagnosis has the potential to greatly decrease cancer mortality. For that purpose, specific cancer biomarkers have been molecularly targeted by aptamer sequences to enable an accurate and rapid detection. Aptamer-based biosensors for cancer diagnostics are a promising alternative to those using antibodies, due to their high affinity and specificity to the target molecules and advantageous production. Synthetic nucleic acid aptamers are generated by in vitro Systematic Evolution of Ligands by Exponential enrichment (SELEX) methodologies that have been improved over the years to enhance the efficacy and to shorten the selection process. Aptamers have been successfully applied in electrochemical, optical, photoelectrochemical and piezoelectrical-based detection strategies. These aptasensors comprise a sensitive, accurate and inexpensive option for cancer detection being used as point-of-care devices. This review highlights the recent advances in cancer biomarkers, achievements and optimizations made in aptamer selection, as well as the different aptasensors developed for the detection of several cancer biomarkers.
Collapse
Affiliation(s)
- Diana A Sousa
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- MIT-Portugal Program, Lisbon, Portugal
| | - Mariana Carneiro
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- BioMark@ISEP, School of Engineering, Polytechnic of Porto, Porto, Portugal
| | - Débora Ferreira
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- MIT-Portugal Program, Lisbon, Portugal
| | - Felismina T C Moreira
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- BioMark@ISEP, School of Engineering, Polytechnic of Porto, Porto, Portugal
| | - Maria Goreti F V Sales
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- MIT-Portugal Program, Lisbon, Portugal
- BioMark@UC, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Lígia R Rodrigues
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
| |
Collapse
|
152
|
Han C, Zhong J, Zhang Q, Hu J, Liu R, Liu H, Mo Z, Chen P, Ling F. Development of a dynamic network biomarkers method and its application for detecting the tipping point of prior disease development. Comput Struct Biotechnol J 2022; 20:1189-1197. [PMID: 35317238 PMCID: PMC8907966 DOI: 10.1016/j.csbj.2022.02.019] [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: 10/14/2021] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 01/13/2023] Open
Abstract
The dynamic network biomarker (DNB) method has advanced since it was first proposed. This review discusses advances in the DNB method that can identify the dynamic change in the expression signature related to the critical time point of disease progression by utilizing different kinds of transcriptome data. The DNB method is good at identifying potential biomarkers for cancer and other disease development processes that are represented by a limited molecular profile change between the normal and critical stages. We highlight that the cancer tipping point or premalignant state has been widely discovered for different types of cancer by using the DNB method that utilizes bulk or single-cell RNA sequencing data. This method could also be applied to other dynamic research studies and help identify early warning signals, such as the prediction of a pre-outbreak of COVID-19. We also discuss how the identification of reliable biomarkers of cancer and the development of new methods can be utilized for early detection and intervention and provide insights into emerging paths of the widespread biomarker candidate pool for further validation and disease/health management.
Collapse
Affiliation(s)
- Chongyin Han
- Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Jiayuan Zhong
- School of Mathematics, South China University of Technology, Guangzhou, Guangdong, China
| | - Qinqin Zhang
- Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Jiaqi Hu
- Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Rui Liu
- Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Huisheng Liu
- Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Zongchao Mo
- Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Pei Chen
- School of Mathematics, South China University of Technology, Guangzhou, Guangdong, China
| | - Fei Ling
- Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
153
|
Vrba L, Futscher BW, Oshiro M, Watts GS, Menashi E, Hu C, Hammad H, Pennington DR, Golconda U, Gavini H, Roe DJ, Shroff RT, Nelson MA. Liquid biopsy, using a novel DNA methylation signature, distinguishes pancreatic adenocarcinoma from benign pancreatic disease. Clin Epigenetics 2022; 14:28. [PMID: 35193708 PMCID: PMC8864826 DOI: 10.1186/s13148-022-01246-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
Abstract
We tested the ability of a novel DNA methylation biomarker set to distinguish metastatic pancreatic cancer cases from benign pancreatic cyst patients and to monitor tumor dynamics using quantitative DNA methylation analysis of cell-free DNA (cfDNA) from blood samples. The biomarkers were able to distinguish malignant cases from benign disease with high sensitivity and specificity (AUC = 0.999). Furthermore, the biomarkers detected a consistent decline in tumor-derived cfDNA in samples from patients undergoing chemotherapy. The study indicates that our liquid biopsy assay could be useful for management of pancreatic cancer patients.
Collapse
Affiliation(s)
- Lukas Vrba
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Bernard W Futscher
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA.,Precision Epigenomics Inc, Tucson, AZ, USA
| | - Marc Oshiro
- The University of Arizona Cancer Center, Tucson, AZ, USA
| | - George S Watts
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | | | - Charles Hu
- Dignity Health Chandler Regional Medical Center, Chandler, AZ, USA
| | - Hytham Hammad
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Division of Hematology/Oncology, Department of Medicine, University of Arizona Caner Center, Tucson, AZ, USA
| | - Daniel R Pennington
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Division of Hematology/Oncology, Department of Medicine, University of Arizona Caner Center, Tucson, AZ, USA
| | | | - Hemanth Gavini
- Division of Hematology/Oncology, Department of Medicine, University of Arizona Caner Center, Tucson, AZ, USA
| | - Denise J Roe
- The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Rachna T Shroff
- The University of Arizona Cancer Center, Tucson, AZ, USA.,Division of Hematology/Oncology, Department of Medicine, University of Arizona Caner Center, Tucson, AZ, USA
| | - Mark A Nelson
- The University of Arizona Cancer Center, Tucson, AZ, USA. .,Department of Pathology, College of Medicine, University of Arizona, Tucson, AZ, USA. .,Precision Epigenomics Inc, Tucson, AZ, USA.
| |
Collapse
|
154
|
Juat DJ, Hachey SJ, Billimek J, Del Rosario MP, Nelson EL, Hughes CCW, Zell JA. Adoptive T-Cell Therapy in Advanced Colorectal Cancer: A Systematic Review. Oncologist 2022; 27:210-219. [PMID: 35274719 PMCID: PMC8914488 DOI: 10.1093/oncolo/oyab038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/15/2021] [Indexed: 11/12/2022] Open
Abstract
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths in the US. For the vast majority of patients with advanced CRC (ie, for those in whom metastatic tumors are unresectable), treatment is palliative and typically involves chemotherapy, biologic therapy, and/or immune checkpoint inhibition. In recent years, the use of adoptive T-cell therapy (ACT), leveraging the body’s own immune system to recognize and target cancer, has become increasingly popular. Unfortunately, while ACT has been successful in the treatment of hematological malignancies, it is less efficacious in advanced CRC due in part to a lack of productive immune infiltrate. This systematic review was conducted to summarize the current data for the efficacy and safety of ACT in advanced CRC. We report that ACT is well tolerated in patients with advanced CRC. Favorable survival estimates among patients with advanced CRC receiving ACT demonstrate promise for this novel treatment paradigm. However, additional stage I/II clinical trials are needed to establish the efficacy and safety of ACT in patients with CRC.
Collapse
Affiliation(s)
- Damie J Juat
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Stephanie J Hachey
- Department of Family Medicine, University of California Irvine, Irvine, CA, USA
| | - John Billimek
- Department of Family Medicine, University of California Irvine, Irvine, CA, USA
| | - Michael P Del Rosario
- Division of Hematology/Oncology, Department of Medicine, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Edward L Nelson
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
- Division of Hematology/Oncology, Department of Medicine, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Christopher C W Hughes
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Jason A Zell
- Division of Hematology/Oncology, Department of Medicine, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
155
|
Brand RE, Persson J, Bratlie SO, Chung DC, Katona BW, Carrato A, Castillo M, Earl J, Kokkola A, Lucas AL, Moser AJ, DeCicco C, Mellby LD, King TC. Detection of Early-Stage Pancreatic Ductal Adenocarcinoma From Blood Samples: Results of a Multiplex Biomarker Signature Validation Study. Clin Transl Gastroenterol 2022; 13:e00468. [PMID: 35166713 PMCID: PMC8963856 DOI: 10.14309/ctg.0000000000000468] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The IMMray PanCan-d test combines an 8-plex biomarker signature with CA19-9 in a proprietary algorithm to detect pancreatic ductal adenocarcinoma (PDAC) in serum samples. This study aimed to validate the clinical performance of the IMMray PanCan-d test and to better understand test performance in Lewis-null (le/le) individuals who cannot express CA19-9. METHODS Serum samples from 586 individuals were analyzed with the IMMray PanCan-d biomarker signature and CA19-9 assay, including 167 PDAC samples, 203 individuals at high risk of familial/hereditary PDAC, and 216 healthy controls. Samples were collected at 11 sites in the United States and Europe. The study was performed by Immunovia, Inc (Marlborough, MA), and sample identity was blinded throughout the study. Test results were automatically generated using validated custom software with a locked algorithm and predefined decision value cutoffs for sample classification. RESULTS The IMMray PanCan-d test distinguished PDAC stages I and II (n = 56) vs high-risk individuals with 98% specificity and 85% sensitivity and distinguished PDAC stages I-IV vs high-risk individuals with 98% specificity and 87% sensitivity. We identified samples with a CA19-9 value of 2.5 U/mL or less as probable Lewis-null (le/le) individuals. Excluding these 55 samples from the analysis increased the IMMray PanCan-d test sensitivity to 92% for PDAC stages I-IV (n = 157) vs controls (n = 379) while maintaining specificity at 99%; test sensitivity for PDAC stages I and II increased from 85% to 89%. DISCUSSION These results demonstrate the IMMray PanCan-d blood test can detect PDAC with high specificity (99%) and sensitivity (92%).
Collapse
Affiliation(s)
- Randall E. Brand
- Division of Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA;
| | - Jan Persson
- Sahlgrenska University Hospital, Department of Surgery, Gothenburg, Sweden;
| | - Svein Olav Bratlie
- Sahlgrenska University Hospital, Department of Surgery, Gothenburg, Sweden;
| | - Daniel C. Chung
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA;
| | - Bryson W. Katona
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA;
| | - Alfredo Carrato
- Molecular Epidemiology and Predictive Markers in Cancer Group, Ramon y Cajal University Hospital, Alcala University, IRYCIS, CIBERONC, Madrid, Spain, Pancreatic Cancer Europe Chairperson, Brussels, Belgium
| | - Marién Castillo
- Molecular Epidemiology and Predictive Markers in Cancer Group, Ramón y Cajal University Hospital, IRYCIS, CIBERONC, Madrid, Spain;
| | - Julie Earl
- Molecular Epidemiology and Predictive Markers in Cancer Group, Ramón y Cajal University Hospital, IRYCIS, CIBERONC, Madrid, Spain;
| | - Arto Kokkola
- Helsinki University Hospital, Helsinki, Finland;
| | - Aimee L. Lucas
- Division of Gastroenterology, Mt. Sinai Medical Center, New York, New York, USA;
| | - A. James Moser
- Division of Surgical Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA;
| | - Corinne DeCicco
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA;
| | | | | |
Collapse
|
156
|
Marques-Garcia F, Boned B, González-Lao E, Braga F, Carobene A, Coskun A, Díaz-Garzón J, Fernández-Calle P, Perich MC, Simon M, Jonker N, Aslan B, Bartlett WA, Sandberg S, Aarsand AK. Critical review and meta-analysis of biological variation estimates for tumor markers. Clin Chem Lab Med 2022; 60:494-504. [PMID: 35143717 DOI: 10.1515/cclm-2021-0725] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 02/01/2022] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Biological variation data (BV) can be used for different applications, but this depends on the availability of robust and relevant BV data. In this study, we aimed to summarize and appraise BV studies for tumor markers, to examine the influence of study population characteristics and concentrations on BV estimates and to discuss the applicability of BV data for tumor markers in clinical practice. METHODS Studies reporting BV data for tumor markers related to gastrointestinal, prostate, breast, ovarian, haematological, lung, and dermatological cancers were identified by a systematic literature search. Relevant studies were evaluated by the Biological Variation Data Critical Appraisal Checklist (BIVAC) and meta-analyses were performed for BIVAC compliant studies to deliver global estimates of within-subject (CVI) and between-subject (CVG) BV with 95% CI. RESULTS The systematic review identified 49 studies delivering results for 22 tumor markers; four papers received BIVAC grade A, 3 B, 27 C and 15 D. Out of these, 29 CVI and 29 CVG estimates met the criteria to be included in the meta-analysis. Robust data are lacking to conclude on the relationship between BV and different disease states and tumor marker concentrations. CONCLUSIONS This review identifies a lack of high-quality BV studies for many tumor markers and a need for delivery of BIVAC compliant studies, including in different, disease states and tumor marker concentrations. As of yet, the state-of-the-art may still be the most appropriate model to establish analytical performance specifications for the majority of tumor markers.
Collapse
Affiliation(s)
- Fernando Marques-Garcia
- Biochemistry Department, Metropolitan North Clinical Laboratory (LCMN), Germans Trias i Pujol Universitary Hospital, Badalona, Barcelona, Spain.,Spanish Society of Laboratory Medicine (SEQCML), Analytical Quality Commission, Barcelona, Spain
| | - Beatriz Boned
- Spanish Society of Laboratory Medicine (SEQCML), Analytical Quality Commission, Barcelona, Spain.,Royo Villanova Hospital, Zaragoza, Spain
| | - Elisabet González-Lao
- Spanish Society of Laboratory Medicine (SEQCML), Analytical Quality Commission, Barcelona, Spain.,Quality Healthcare Consulting, Grupo ACMS, Barcelona, Spain
| | - Federica Braga
- Research Centre for Metrological Traceability in Laboratory Medicine (CIRME), University of Milan, Milan, Italy
| | - Anna Carobene
- Servizio Medicina di Laboratorio, Ospedale San Raffaele, Milan, Italy
| | - Abdurrahman Coskun
- School of Medicine, Acibadem Mehmet Ali Aydınlar University, Atasehir, Istanbul, Turkey
| | - Jorge Díaz-Garzón
- Spanish Society of Laboratory Medicine (SEQCML), Analytical Quality Commission, Barcelona, Spain.,Department of Laboratory Medicine, La Paz University Hospital, Madrid, Spain
| | - Pilar Fernández-Calle
- Spanish Society of Laboratory Medicine (SEQCML), Analytical Quality Commission, Barcelona, Spain.,Department of Laboratory Medicine, La Paz University Hospital, Madrid, Spain
| | - Maria Carmen Perich
- Spanish Society of Laboratory Medicine (SEQCML), Analytical Quality Commission, Barcelona, Spain
| | - Margarida Simon
- Spanish Society of Laboratory Medicine (SEQCML), Analytical Quality Commission, Barcelona, Spain.,Consortium of Laboratory Intercomarcal Alt Penedès and Garraf l'Anoia, Vilafranca del Penedès, Spain
| | - Niels Jonker
- Certe-Wilhelmina Ziekenhuis Assen, Assen, The Netherlands
| | - Berna Aslan
- Institute for Quality Management in Healthcare (IQMH), Centre for Proficiency Testing, Toronto, Ontario, Canada
| | | | - Sverre Sandberg
- Department of Medical Biochemistry and Pharmacology, Norwegian Porphyria Centre, Haukeland University Hospital, Bergen, Norway.,Norwegian Organization for Quality Improvement of Laboratory Examinations (NOKLUS), Haraldsplass Deaconess Hospital, Bergen, Norway.,Department of Global Health and Primary Care, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Aasne K Aarsand
- Department of Medical Biochemistry and Pharmacology, Norwegian Porphyria Centre, Haukeland University Hospital, Bergen, Norway.,Norwegian Organization for Quality Improvement of Laboratory Examinations (NOKLUS), Haraldsplass Deaconess Hospital, Bergen, Norway
| | | |
Collapse
|
157
|
Ionica E, Gaina G, Tica M, Chifiriuc MC, Gradisteanu-Pircalabioru G. Contribution of Epithelial and Gut Microbiome Inflammatory Biomarkers to the Improvement of Colorectal Cancer Patients' Stratification. Front Oncol 2022; 11:811486. [PMID: 35198435 PMCID: PMC8859258 DOI: 10.3389/fonc.2021.811486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
In order to ensure that primary endpoints of clinical studies are attained, the patients' stratification is an important aspect. Selection criteria include age, gender, and also specific biomarkers, such as inflammation scores. These criteria are not sufficient to achieve a straightforward selection, however, in case of multifactorial diseases, with unknown or partially identified mechanisms, occasionally including host factors, and the microbiome. In these cases, the efficacy of interventions is difficult to predict, and as a result, the selection of subjects is often random. Colorectal cancer (CRC) is a highly heterogeneous disease, with variable clinical features, outcomes, and response to therapy; the CRC onset and progress involves multiple sequential steps with accumulation of genetic alterations, namely, mutations, gene amplification, and epigenetic changes. The gut microbes, either eubiotic or dysbiotic, could influence the CRC evolution through a complex and versatile crosstalk with the intestinal and immune cells, permanently changing the tumor microenvironment. There have been significant advances in the development of personalized approaches for CRC screening, treatment, and potential prevention. Advances in molecular techniques bring new criteria for patients' stratification-mutational analysis at the time of diagnosis to guide treatment, for example. Gut microbiome has emerged as the main trigger of gut mucosal homeostasis. This may impact cancer susceptibility through maintenance of the epithelial/mucus barrier and production of protective metabolites, such as short-chain fatty acids (SCFAs) via interactions with the hosts' diet and metabolism. Microbiome dysbiosis leads to the enrichment of cancer-promoting bacterial populations, loss of protective populations or maintaining an inflammatory chronic state, all of which contribute to the development and progression of CRC. Meanwhile, variations in patient responses to anti-cancer immuno- and chemotherapies were also linked to inter-individual differences in intestine microbiomes. The authors aim to highlight the contribution of epithelial and gut microbiome inflammatory biomarkers in the improvement of CRC patients' stratification towards a personalized approach of early diagnosis and treatment.
Collapse
Affiliation(s)
- Elena Ionica
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Gisela Gaina
- Laboratory of Cell Biology, Neuroscience and Experimental Miology, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Mihaela Tica
- Bucharest Emergency University Hospital, Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Biological Science Division, Romanian Academy of Sciences, Bucharest, Romania
| | | |
Collapse
|
158
|
Abstract
Oxidative stress is caused by homeostasis disrupted by excessively increased reactive oxygen species (ROS) due to intrinsic or extrinsic causes. Among diseases caused by the abnormal induction of ROS, cancer is a representative disease that shows gender specificity in the development and malignancy. Females have the advantage of longer life expectancy than males because of the genetic advantages derived from X chromosomes, the antioxidant protective function by estrogen, and the decrease in exposure to extrinsic risk factors such as alcohol and smoking. This study first examines the ordinary biological responses to oxidative stress and the effects of ROS on the cancer progression and describes the differences in cancer incidence and mortality by gender and the differences in oxidative stress affected by sex hormones. This paper summarized how several important transcription factors regulate ROS-induced stress and in vivo responses, and how their expression is changed by sex hormones. Estrogen is associated with disease resistance and greater mitochondrial function, and reduces mitochondrial damage and ROS production in females than in males. In addition, estrogen affects the activation of nuclear factor-erythroid 2 p45-related factor (NRF) 2 and the regulation of other antioxidant-related transcription factors through NRF2, leading to benefits in females. Because ROS have a variety of molecular targets in cells, the effective cancer treatment requires understanding the potential of ROS and focusing on the characteristics of the research target such as patient's gender. Therefore, this review intends to emphasize the necessity of discussing gender specificity as a new therapeutic approach for efficient regulation of ROS considering individual specificity.
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Chemistry, College of Science and Technology, Duksung Women's University, Seoul 01369, Republic of Korea
| |
Collapse
|
159
|
Choe EK, Shivakumar M, Verma A, Verma SS, Choi SH, Kim JS, Kim D. Leveraging deep phenotyping from health check-up cohort with 10,000 Korean individuals for phenome-wide association study of 136 traits. Sci Rep 2022; 12:1930. [PMID: 35121771 PMCID: PMC8817039 DOI: 10.1038/s41598-021-04580-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022] Open
Abstract
The expanding use of the phenome-wide association study (PheWAS) faces challenges in the context of using International Classification of Diseases billing codes for phenotype definition, imbalanced study population ethnicity, and constrained application of the results in research. We performed a PheWAS utilizing 136 deep phenotypes corroborated by comprehensive health check-ups in a Korean population, along with trans-ethnic comparisons through using the UK Biobank and Biobank Japan Project. Meta-analysis with Korean and Japanese population was done. The PheWAS associated 65 phenotypes with 14,101 significant variants (P < 4.92 × 10-10). Network analysis, visualization of cross-phenotype mapping, and causal inference mapping with Mendelian randomization were conducted. Among phenotype pairs from the genotype-driven cross-phenotype associations, we evaluated penetrance in correlation analysis using a clinical database. We focused on the application of PheWAS in order to make it robust and to aid the derivation of biological meaning post-PheWAS. This comprehensive analysis of PheWAS results based on a health check-up database will provide researchers and clinicians with a panoramic overview of the networks among multiple phenotypes and genetic variants, laying groundwork for the practical application of precision medicine.
Collapse
Affiliation(s)
- Eun Kyung Choe
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, B304 Richards Building, 3700 Hamilton Walk, Philadelphia, PA, 19104-6116, USA.,Department of Surgery, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, 06236, South Korea
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, B304 Richards Building, 3700 Hamilton Walk, Philadelphia, PA, 19104-6116, USA
| | - Anurag Verma
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shefali Setia Verma
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Seung Ho Choi
- Department of Internal Medicine, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, 06236, South Korea
| | - Joo Sung Kim
- Department of Internal Medicine, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, 06236, South Korea. .,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, B304 Richards Building, 3700 Hamilton Walk, Philadelphia, PA, 19104-6116, USA. .,Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
160
|
Yalcin S, Philip PA, Athanasiadis I, Bazarbashi S, Shamseddine A. Classification of early-stage colon cancer with Immunoscore ®: clinical evidence and case studies. Future Oncol 2022; 18:613-623. [PMID: 34904905 DOI: 10.2217/fon-2021-0712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/05/2021] [Indexed: 02/08/2023] Open
Abstract
Immunoscore® is a digital pathology diagnostic immunoassay used to complement tumor node metastasis staging for the prediction of recurrence risk in patients with early-stage colon cancer. In combination with standard clinicopathological features, Immunoscore informs adjuvant chemotherapy decision-making for patients with early-stage colon cancer. Immunoscore has been validated in patients with stage II/III colon cancer and demonstrated to be a stronger prognostic factor for survival than tumor node metastasis staging alone. Immunoscore improves the prognostic definition of patients with colon cancer, the identification of those patients at high risk of tumor recurrence, and the ability to predict which patients will derive most benefit from the use of adjuvant chemotherapy. Immunoscore has robust analytical performance characteristics which include good interlaboratory reproducibility and overall assay precision.
Collapse
Affiliation(s)
- Suayib Yalcin
- Department of Medical Oncology, Hacettepe University Institute of Cancer, Hacettepe University, Ankara, Turkey
| | - Philip A Philip
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ilias Athanasiadis
- Department of Medical Oncology, Mitera Hospital, part of Hygeia-Polis, Athens, Greece
| | - Shouki Bazarbashi
- Oncology Centre, KFSHRC King Faisal Specialist Hospital & Research Centre, Saudi Arabia
| | - Ali Shamseddine
- American University of Beirut Medical Center (AUBMC) & Lebanese Society of Medical Oncology, Beirut, Lebanon
| |
Collapse
|
161
|
Demirli Atici S, Kamer E. Is CA19-9 effective in predicting chemotherapeutic response in patients with synchronous liver metastases with colorectal cancer? World J Gastroenterol 2022; 28:500-501. [PMID: 35125833 PMCID: PMC8790559 DOI: 10.3748/wjg.v28.i4.500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/11/2021] [Accepted: 01/13/2022] [Indexed: 02/06/2023] Open
Abstract
Evaluation of response to chemotherapy in colorectal cancer patients with synchronous liver metastases is important in terms of treatment management. In this Letter to the Editor, several issues in the article are discussed. For the comparison of carbohydrate antigen 19-9 (CA19-9) values referenced in the study, the patient group was not matched for cancer stage. Therefore, it may be more appropriate to select and compare CA19-9 values in patients with same-stage cancer.
Collapse
Affiliation(s)
- Semra Demirli Atici
- Department of General Surgery, University of Health Sciences Tepecik Training and Research Hospital, İzmir 35180, Turkey
| | - Erdinc Kamer
- Department of General Surgery, University of Health Sciences Tepecik Training and Research Hospital, İzmir 35180, Turkey
| |
Collapse
|
162
|
Manojlovic N, Savic G, Nikolic B, Rancic N. Dynamic monitoring of carcinoembryonic antigen, CA19-9 and inflammation-based indices in patients with advanced colorectal cancer undergoing chemotherapy. World J Clin Cases 2022; 10:899-918. [PMID: 35127905 PMCID: PMC8790463 DOI: 10.12998/wjcc.v10.i3.899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/21/2021] [Accepted: 12/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The roles of carcinoembryonic antigen (CEA) and carbohydrate antigen (CA19-9) in monitoring the patient response to chemotherapy for metastatic colorectal cancer (mCRC) are not clearly defined, and inflammatory indices, including the neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), platelet-to-lymphocyte ratio (PLR) and systemic immune-inflammation index (SII), have been sparsely investigated for this purpose.
AIM To aim of this study was to evaluate the relationship between the kinetics of CEA, CA19-9, NLR, LMR, PLR and SII in serum and patient response to chemotherapy estimated by computed tomography (CT) in patients with unresectable mCRC.
METHODS Patients with mCRC treated with a 1st-line and 2nd-line chemotherapy underwent at least 3 whole-body spiral CT scans during response monitoring according to the Response Evaluation Criteria in Solid Tumour 1.1 (RECIST 1.1), and simultaneous determination of CEA, CA19-9, neutrophil, lymphocyte, platelet and monocyte levels was performed. The kinetics of changes in the tumour markers and inflammatory indices were calculated as the percentage change from baseline or nadir, while receiver operating characteristic curves were drawn to select the thresholds to define patients with progressive or responsive disease with the highest sensitivity (Se) and specificity (Sp). The correlation of tumour marker kinetics with inflammatory index changes and RECIST response was determined by univariate and multivariate logistic regression analysis and the clinical utility index (CUI).
RESULTS A total of 102 patients with mCRC treated with chemotherapy were included. Progressive disease (PD), defined as a CEA increase of 25.52%, resulted in an Se of 80.3%, an Sp of 84%, a good CUI negative [CUI (Ve-)] value of 0.75 and a good fraction correct (FC) value of 81.2; at a CEA cut-off of -60.85% with an Se of 100% and an Sp of 35.7% for PD, CT could be avoided in 25.49% of patients. The 21.49% CA19-9 cut-off for PD had an Se of 66.5%, an Sp of 87.4%, an acceptable CUI (Ve-) value of 0.65 and an acceptable FC value of 75. An NLR increase of 11.5% for PD had an Se of 67% and an Sp of 66%; a PLR increase of 5.9% had an Se of 53% and an Sp of 69%; an SII increase above -6.04% had an Se of 72% and an Sp of 63%; and all had acceptable CUI (Ve-) values at 0.55. In the univariate logistic regression analysis, CEA (P < 0.001), CA19-9 (P < 0.05), NLR (P < 0.05), PLR (P < 0.05) and SII (P < 0.05) were important predictors of tumour progression, but in the multivariate logistic regression analysis, CEA was the only independent predictor of PD (P < 0.05).
CONCLUSION CEA is a useful marker for monitoring the chemotherapy response of patients with unresectable mCRC and could replace a quarter of CT examinations. CA19-9 has poorer diagnostic characteristics than CEA but could be useful in some clinical circumstances, particularly when CEA is not increased. Dynamic changes in the inflammatory indices NLR, PLR and SII could be promising for further investigation as markers of the chemotherapy response.
Collapse
Affiliation(s)
- Nebojsa Manojlovic
- Clinic for Gastroenterology and Hepatology, Military Medical Academy, Faculty of Medicine of the Military Medical Academy, University of Defence, Belgrade 11000, Serbia
| | - Goran Savic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Belgrade, Serbia, Military Medical Academy, Belgrade 11000, Serbia
| | - Bojan Nikolic
- Institute for Radiology, Military Medical Academy, Belgrade 11000, Serbia
| | - Nemanja Rancic
- Center for Clinical Pharmacology, Institute for Radiology, Military Medical Academy, Faculty of Medicine of the Military Medical Academy, University of Defence, Belgrade 11000, Serbia
| |
Collapse
|
163
|
Aires F, Rodrigues D, Lamas MP, Herdeiro MT, Figueiras A, Oliveira MJ, Marques M, Pinto AT. C-Reactive Protein as Predictive Biomarker for Response to Chemoradiotherapy in Patients with Locally Advanced Rectal Cancer: A Retrospective Study. Cancers (Basel) 2022; 14:491. [PMID: 35158759 PMCID: PMC8833484 DOI: 10.3390/cancers14030491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
The standard of care for the treatment of locally advanced rectal cancer is neoadjuvant chemoradiotherapy (nCRT) followed by surgery, but complete response rates are reduced. To find predictive biomarkers of response to therapy, we conducted a retrospective study evaluating blood biomarkers before nCRT. Hemoglobin (Hg), C-reactive protein (CRP), platelets, carcinoembryonic antigen, carbohydrate antigen 19.9 levels, and neutrophil/lymphocyte ratio were obtained from 171 rectal cancer patients before nCRT. Patients were classified as responders (Ryan 0-1; ycT0N0), 59.6% (n = 102), or nonresponders (Ryan 2-3), 40.3% (n = 69), in accordance with the Ryan classification. A logistic regression using prognostic pretreatment factors identified CRP ≤ 3.5 (OR = 0.05; 95%CI: 0.01-0.21) as a strong independent predictor of response to treatment. Multivariate analysis showed that CRP was an independent predictor of disease-free survival (DFS) (HR = 5.48; 95%CI: 1.54-19.48) and overall survival (HR = 6.10; 95%CI 1.27-29.33) in patients treated with nCRT. Platelets were an independent predictor of DFS (HR = 3.068; 95%CI: 1.29-7.30) and OS (HR= 4.65; 95%CI: 1.66-13.05) and Hg was revealed to be an independent predictor of DFS (HR = 0.37; 95%CI: 0.15-0.90) in rectal cancer patients treated with nCRT. The lower expression of CRP is independently associated with an improved response to nCRT, DFS, and OS.
Collapse
Affiliation(s)
- Fátima Aires
- Radiotherapy Department of Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal; (D.R.); (M.M.)
| | - Darlene Rodrigues
- Radiotherapy Department of Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal; (D.R.); (M.M.)
- ICBAS–Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal;
- CINTESIS–Center for Health Technology and Services Research, University of Porto, 4200-450 Porto, Portugal
| | - María Piñeiro Lamas
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiology and Public Health–CIBERESP), 15706 Santiago de Compostela, Spain; (M.P.L.); (A.F.)
- Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Maria Teresa Herdeiro
- Department of Medical Sciences, Institute of Biomedicine–iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal; (M.T.H.); (A.T.P.)
| | - Adolfo Figueiras
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiology and Public Health–CIBERESP), 15706 Santiago de Compostela, Spain; (M.P.L.); (A.F.)
- Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Faculty of Medicine, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Maria José Oliveira
- ICBAS–Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal;
- i3S–Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- INEB–Instituto de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Margarida Marques
- Radiotherapy Department of Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal; (D.R.); (M.M.)
| | - Ana Teresa Pinto
- Department of Medical Sciences, Institute of Biomedicine–iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal; (M.T.H.); (A.T.P.)
| |
Collapse
|
164
|
Kankeu Fonkoua LA, Serrano Uson Junior PL, Mody K, Mahipal A, Borad MJ, Roberts LR. Novel and emerging targets for cholangiocarcinoma progression: therapeutic implications. Expert Opin Ther Targets 2022; 26:79-92. [PMID: 35034558 DOI: 10.1080/14728222.2022.2029412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Cholangiocarcinoma (CCA) is a heterogeneous group of aggressive biliary malignancies. While surgery and liver transplantation are the only potentially curative modalities for early-stage disease, limited options are available for most patients with incurable-stage disease. Survival outcomes remain dismal. Recent molecular profiling efforts have led to improved understanding of the genomic landscape of CCA and to the identification of subgroups with distinct diagnostic, prognostic, and therapeutic implications. AREAS COVERED : We provide an updated review and future perspectives on features of cholangiocarcinogenesis that can be translated into therapeutic biomarkers and targets. We highlight the critical studies that have established current systemic chemotherapy and targeted therapeutics, while elaborating on novel targeted and immunotherapeutic approaches in development. Relevant literature and clinical studies were identified by searching PubMed and www.ClinicalTrials.gov. EXPERT OPINION : While therapies targeting the various molecular subgroups of CCA are rapidly emerging and changing treatment paradigms, their success has been limited by the genetic heterogeneity of CCA and the plasticity of the targets. Novel strategies aiming to combine immunotherapy, chemotherapy, and molecularly-targeted therapeutics will be required to offer durable clinical benefit and maximize survival.
Collapse
Affiliation(s)
| | | | - Kabir Mody
- Rochester, MN, and Oncology in Jacksonville, FL, Mayo Clinic, USA
| | | | | | | |
Collapse
|
165
|
Validation of Urinary Charged Metabolite Profiles in Colorectal Cancer Using Capillary Electrophoresis-Mass Spectrometry. Metabolites 2022; 12:metabo12010059. [PMID: 35050180 PMCID: PMC8779129 DOI: 10.3390/metabo12010059] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/25/2021] [Accepted: 01/06/2022] [Indexed: 02/04/2023] Open
Abstract
This study aimed to validate and reanalyze urinary biomarkers for detecting colorectal cancers (CRCs). We previously conducted urinary metabolomic analyses using capillary electrophoresis-mass spectrometry and found a significant difference in various metabolites, especially polyamines, between patients with CRC and healthy controls (HC). We analyzed additional samples and confirmed consistency between the newly and previously analyzed data. In total, we included 36 HC, 34 adenoma (AD), and 214 CRC samples, which were used for subsequent analyses. Among the 132 quantified metabolites, 16 exhibited consistent differences in both datasets, which included polyamines, etc. Pathway analyses of the integrated data revealed significant differences in many metabolites, such as glutamine, and metabolites of the TCA (tricarboxylic acid cycle) and urea cycles. The discrimination ability of the combination of multiple metabolites among the three groups was evaluated, which yielded higher sensitivity than tumor markers. The Mann–Whitney test was employed to evaluate the prognosis predictivity of the assessed metabolites and the difference between the patients with or without recurrence, which yielded 16 significantly different metabolites. Among these 16 metabolites, 11 presented significant prognosis predictivity. These data indicated the potential of metabolite-based discrimination of patients with CRC and AD from HC and prognosis predictivity of the monitored metabolites.
Collapse
|
166
|
Clinical significance of controlling nutritional status score (CONUT) in evaluating outcome of postoperative patients with gastric cancer. Sci Rep 2022; 12:93. [PMID: 34997105 PMCID: PMC8742112 DOI: 10.1038/s41598-021-04128-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
The stomach is the main digestive organ in humans. Patients with gastric cancer often develop digestive problems, which result in poor nutrition. Nutritional status is closely related to postoperative complications and quality of life (QoL) in patients with gastric cancer. The controlling nutritional status (CONUT) score is a novel tool to evaluate the nutritional status of patients. However, the relationship of the CONUT score with postoperative complications, QoL, and psychological status in patients with gastric cancer has not been investigated. The present follow-up study was conducted in 106 patients who underwent radical gastrectomy in our hospital between 2014 and 2019. The CONUT score, postoperative complications, psychological status, postoperative QoL scores, and overall survival (OS) of patients with gastric cancer were collected, and the relationship between them was analyzed. A significant correlation was observed between the CONUT score and postoperative complications of gastric cancer (P < 0.001), especially anastomotic leakage (P = 0.037). The multivariate regression analysis exhibited that the CONUT score (P = 0.002) is an independent risk factor for postoperative complications. The CONUT score was correlated with the state anxiety questionnaire (S-AI) for evaluating psychological status (P = 0.032). However, further regression analysis exhibited that the CONUT score was not an independent risk factor for psychological status. Additionally, the CONUT score was associated with postoperative QoL. The multivariate regression analysis exhibited that the CONUT score was an independent risk factor for the global QoL (P = 0.048). Moreover, the efficiency of CONUT score, prognostic nutrition index, and serum albumin in evaluating complications, psychological status, and QoL was compared, and CONUT score was found to outperform the other measures (Area Under Curve, AUC = 0.7368). Furthermore, patients with high CONUT scores exhibited shorter OS than patients with low CONUT scores (P = 0.005). Additionally, the postoperative complications (HR 0.43, 95% CI 0.21–0.92, P = 0.028), pathological stage (HR 2.26, 95% CI 1.26–4.06, P = 0.006), and global QoL (HR 15.24, 95% CI 3.22–72.06, P = 0.001) were associated with OS. The CONUT score can be used to assess the nutritional status of patients undergoing gastric cancer surgery and is associated with the incidence of postoperative complications and QoL.
Collapse
|
167
|
Transcriptomic profiling of adjuvant colorectal cancer identifies three key prognostic biological processes and a disease specific role for granzyme B. PLoS One 2022; 16:e0262198. [PMID: 34972191 PMCID: PMC8719661 DOI: 10.1371/journal.pone.0262198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 11/25/2021] [Indexed: 12/20/2022] Open
Abstract
Background Colorectal cancer (CRC) is a leading cause of cancer-related deaths, with a 5% 5-year survival rate for metastatic disease, yet with limited therapeutic advancements due to insufficient understanding of and inability to accurately capture high-risk CRC patients who are most likely to recur. We aimed to improve high-risk classification by identifying biological pathways associated with outcome in adjuvant stage II/III CRC. Methods and findings We included 1062 patients with stage III or high-risk stage II colon carcinoma from the prospective three-arm randomized phase 3 AVANT trial, and performed expression profiling to identify a prognostic signature. Data from validation cohort GSE39582, The Cancer Genome Atlas, and cell lines were used to further validate the prognostic biology. Our retrospective analysis of the adjuvant AVANT trial uncovered a prognostic signature capturing three biological functions—stromal, proliferative and immune—that outperformed the Consensus Molecular Subtypes (CMS) and recurrence prediction signatures like Oncotype Dx in an independent cohort. Importantly, within the immune component, high granzyme B (GZMB) expression had a significant prognostic impact while other individual T-effector genes were less or not prognostic. In addition, we found GZMB to be endogenously expressed in CMS2 tumor cells and to be prognostic in a T cell independent fashion. A limitation of our study is that these results, although robust and derived from a large dataset, still need to be clinically validated in a prospective study. Conclusions This work furthers our understanding of the underlying biology that propagates stage II/III CRC disease progression and provides scientific rationale for future high-risk stratification and targeted treatment evaluation in biomarker defined subpopulations of resectable high-risk CRC. Our results also shed light on an alternative GZMB source with context-specific implications on the disease’s unique biology.
Collapse
|
168
|
Ambade V, Singh A, Bhatia K, Misra P, Mukherjee B, Negi R. Serum tumor markers: A study on their rational use in various cancers. MEDICAL JOURNAL OF DR. D.Y. PATIL VIDYAPEETH 2022. [DOI: 10.4103/mjdrdypu.mjdrdypu_175_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
169
|
Oldfield L, Evans A, Rao RG, Jenkinson C, Purewal T, Psarelli EE, Menon U, Timms JF, Pereira SP, Ghaneh P, Greenhalf W, Halloran C, Costello E. Blood levels of adiponectin and IL-1Ra distinguish type 3c from type 2 diabetes: Implications for earlier pancreatic cancer detection in new-onset diabetes. EBioMedicine 2022; 75:103802. [PMID: 34990893 PMCID: PMC8741427 DOI: 10.1016/j.ebiom.2021.103802] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Screening for pancreatic ductal adenocarcinoma (PDAC) in populations at high risk is recommended. Individuals with new-onset type 2 diabetes mellitus (NOD) are the largest high-risk group for PDAC. To facilitate screening, we sought biomarkers capable of stratifying NOD subjects into those with type 2 diabetes mellitus (T2DM) and those with the less prevalent PDAC-related diabetes (PDAC-DM), a form of type 3c DM commonly misdiagnosed as T2DM. METHODS Using mass spectrometry- and immunoassay-based methodologies in a multi-stage analysis of independent sample sets (n=443 samples), blood levels of 264 proteins were considered using Ingenuity Pathway Analysis, literature review and targeted training and validation. FINDINGS Of 30 candidate biomarkers evaluated in up to four independent patient sets, 12 showed statistically significant differences in levels between PDAC-DM and T2DM. The combination of adiponectin and interleukin-1 receptor antagonist (IL-1Ra) showed strong diagnostic potential, (AUC of 0.91; 95% CI: 0.84-0.99) for the distinction of T3cDM from T2DM. INTERPRETATION Adiponectin and IL-1Ra warrant further consideration for use in screening for PDAC in individuals newly-diagnosed with T2DM. FUNDING North West Cancer Research, UK, Cancer Research UK, Pancreatic Cancer Action, UK.
Collapse
Affiliation(s)
- Lucy Oldfield
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
| | - Anthony Evans
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
| | - Rohith Gopala Rao
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
| | - Claire Jenkinson
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
| | - Tejpal Purewal
- Department of Diabetes and Endocrinology, Royal Liverpool University Hospital, UK
| | - Eftychia E Psarelli
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
| | - Usha Menon
- Institute of Clinical Trials and Methodology, University College London, UK
| | - John F Timms
- Women's Cancer, Institute for Women's Health, University College London, UK
| | - Stephen P Pereira
- Institute for Liver and Digestive Health, University College London, UK
| | - Paula Ghaneh
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
| | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
| | - Christopher Halloran
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, UK.
| |
Collapse
|
170
|
Vanek P, Eid M, Psar R, Zoundjiekpon V, Urban O, Kunovský L. Current trends in the diagnosis of pancreatic cancer. VNITRNI LEKARSTVI 2022; 68:363-370. [PMID: 36316197 DOI: 10.36290/vnl.2022.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a dreaded malignancy with a dismal 5-year survival rate despite maximal efforts on optimizing treatment strategies. Currently, early detection is considered to be the most effective way to improve survival as radical resection is the only potential cure. PDAC is often divided into four categories based on the extent of disease: resectable, borderline resectable, locally advanced, and metastatic. Unfortunately, the majority of patients are diagnosed with locally advanced or metastatic disease, which renders them ineligible for curative resection. This is mainly due to the lack of or vague symptoms while the disease is still localized, although appropriate utilization and prompt availability of adequate diagnostic tools is also critical given the aggressive nature of the disease. A cost-effective biomarker with high specificity and sensitivity allowing early detection of PDAC without the need for advanced or invasive methods is still not available. This leaves the diagnosis dependent on radiodiagnostic methods or endoscopic ultrasound. Here we summarize the latest epidemiological data, risk factors, clinical manifestation, and current diagnostic trends and implications of PDAC focusing on serum biomarkers and imaging modalities. Additionally, up-to-date management and therapeutic algorithms are outlined.
Collapse
|
171
|
Tumour- associated autoantibodies as prognostic cancer biomarkers- a review. Autoimmun Rev 2022; 21:103041. [DOI: 10.1016/j.autrev.2022.103041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 12/12/2022]
|
172
|
Kuligina E, Moiseyenko F, Belukhin S, Stepanova E, Zakharova M, Chernobrivtseva V, Aliev I, Sharabura T, Moiseyenko V, Aleksakhina S, Laidus T, Martianov A, Kholmatov M, Whitehead A, Yanus G, Imyanitov E. Tumor irradiation may facilitate the detection of tumor-specific mutations in plasma. World J Clin Oncol 2021; 12:1215-1226. [PMID: 35070740 PMCID: PMC8716992 DOI: 10.5306/wjco.v12.i12.1215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/26/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The mutation-based analysis of circulating tumor DNA (ctDNA) is a promising diagnostic tool for clinical oncology. However, it has low success rate because many cancer patients do not have detectable ctDNA in the bloodstream.
AIM To evaluate whether preoperative tumor irradiation results in a transient increase of plasma ctDNA concentration due to the induction of apoptosis in radiation-exposed cells.
METHODS This study focused on patients with locally advanced rectal cancer, because preoperative tumor irradiation is a part of their standard treatment plan. Nine subjects, whose tumors contained KRAS, NRAS or BRAF mutations, donated serial blood samples 1 h prior to the first fraction of irradiation (at baseline), immediately after the first fraction (time 0), and 1, 3, 6, 12, 24, 36, 48, 72 and 96 h after the first fraction. The amount of mutated gene copies was measured by droplet digital PCR.
RESULTS Five out of nine patients were mutation-negative by ctDNA test at baseline; two of these subjects demonstrated an emergence of the mutated DNA copies in the bloodstream within the follow-up period. There were 4 patients, who had detectable ctDNA in the plasma at the start of the experiment; three of them showed an evident treatment-induced increase of the content of mutated RAS/RAF alleles.
CONCLUSION Local tumor irradiation may facilitate the detection of tumor-specific DNA in the bloodstream. These data justify further assessment of the clinical feasibility of irradiation-assisted liquid biopsy.
Collapse
Affiliation(s)
- Ekaterina Kuligina
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Fedor Moiseyenko
- Department of Therapy, City Cancer Center, St.-Petersburg 197758, Russia
| | - Sergey Belukhin
- Department of Surgery, City Cancer Center, St.-Petersburg 197758, Russia
| | | | - Maria Zakharova
- Department of Radiology, City Cancer Center, St.-Petersburg 197758, Russia
| | | | - Ikram Aliev
- Department of Surgery, City Cancer Center, St.-Petersburg 197758, Russia
| | - Tatiana Sharabura
- Department of Radiology, City Cancer Center, St.-Petersburg 197758, Russia
| | | | - Svetlana Aleksakhina
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Tatiana Laidus
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
| | - Aleksandr Martianov
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Maksim Kholmatov
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
| | - Aldon Whitehead
- Internal Medicine Residency Program, The University of Illinois College of Medicine, Chicago, IL 60612, United States
| | - Grigoriy Yanus
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Evgeny Imyanitov
- Department of Tumor Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
- Department of Oncology, I.I. Mechnikov Northwestern Medical University, St.-Petersburg 191015, Russia
| |
Collapse
|
173
|
Ju J, Wismans LV, Mustafa DAM, Reinders MJT, van Eijck CHJ, Stubbs AP, Li Y. Robust deep learning model for prognostic stratification of pancreatic ductal adenocarcinoma patients. iScience 2021; 24:103415. [PMID: 34901786 PMCID: PMC8637475 DOI: 10.1016/j.isci.2021.103415] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/27/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023] Open
Abstract
A major challenge for treating patients with pancreatic ductal adenocarcinoma (PDAC) is the unpredictability of their prognoses due to high heterogeneity. We present Multi-Omics DEep Learning for Prognosis-correlated subtyping (MODEL-P) to identify PDAC subtypes and to predict prognoses of new patients. MODEL-P was trained on autoencoder integrated multi-omics of 146 patients with PDAC together with their survival outcome. Using MODEL-P, we identified two PDAC subtypes with distinct survival outcomes (median survival 10.1 and 22.7 months, respectively, log rank p = 1 × 10−6), which correspond to DNA damage repair and immune response. We rigorously validated MODEL-P by stratifying patients in five independent datasets into these two survival groups and achieved significant survival difference, which is superior to current practice and other subtyping schemas. We believe the subtype-specific signatures would facilitate PDAC pathogenesis discovery, and MODEL-P can provide clinicians the prognoses information in the treatment decision-making to better gauge the benefits versus the risks. We developed DL-based MODEL-P to identify prognosis-correlated PDAC subtypes The identified subtypes related to DNA damage repair and immune response processes MODEL-P stratified patients from independent datasets into distinct survival groups MODEL-P could be used in clinics to aid treatment decision-making
Collapse
Affiliation(s)
- Jie Ju
- Department of Pathology & Clinical Bioinformatics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Leonoor V Wismans
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dana A M Mustafa
- Department of Pathology & Clinical Bioinformatics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marcel J T Reinders
- The Delft Bioinformatics Lab, Delft University of Technology, Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Andrew P Stubbs
- Department of Pathology & Clinical Bioinformatics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yunlei Li
- Department of Pathology & Clinical Bioinformatics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
174
|
Song J, Sokoll LJ, Chan DW, Zhang Z. Validation of Serum Biomarkers That Complement CA19-9 in Detecting Early Pancreatic Cancer Using Electrochemiluminescent-Based Multiplex Immunoassays. Biomedicines 2021; 9:biomedicines9121897. [PMID: 34944713 PMCID: PMC8698985 DOI: 10.3390/biomedicines9121897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy; its early detection is critical for improving prognosis. Electrochemiluminescent-based multiplex immunoassays were developed with high analytical performance. All proteins were analyzed in sera of patients diagnosed with PDAC (n = 138), benign pancreatic conditions (111), and healthy controls (70). The clinical performance of these markers was evaluated individually or in combination for their complementarity to CA19-9 in detecting early PDAC. Logistic regression modeling including sex and age as cofactors identified a two-marker panel of CA19-9 and CA-125 that significantly improved the performance of CA19-9 alone in discriminating PDAC (AUC: 0.857 vs. 0.766), as well as early stage PDAC (0.805 vs. 0.702) from intraductal papillary mucinous neoplasm (IPMN). At a fixed specificity of 80%, the panel significantly improved sensitivities (78% vs. 41% or 72% vs. 59%). A two-marker panel of HE4 and CEA significantly outperformed CA19-9 in separating IPMN from chronic pancreatitis (0.841 vs. 0.501). The biomarker panels evaluated by assays demonstrated potential complementarity to CA19-9 in detecting early PDAC, warranting additional clinical validation to determine their role in the early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Jin Song
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- Correspondence: (J.S.); (Z.Z.); Tel.: +1-443-287-6363 (J.S.); +1-410-502-7871 (Z.Z.)
| | - Lori J. Sokoll
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel W. Chan
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zhen Zhang
- Center for Biomarker Discovery and Translation, Department of Pathology, Johns Hopkins University School of Medicine, 419 North Caroline Street, Baltimore, MD 21231, USA; (L.J.S.); (D.W.C.)
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Correspondence: (J.S.); (Z.Z.); Tel.: +1-443-287-6363 (J.S.); +1-410-502-7871 (Z.Z.)
| |
Collapse
|
175
|
Circadian and chemotherapy-related changes in urinary modified nucleosides excretion in patients with metastatic colorectal cancer. Sci Rep 2021; 11:24015. [PMID: 34907230 PMCID: PMC8671418 DOI: 10.1038/s41598-021-03247-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022] Open
Abstract
Urinary levels of modified nucleosides reflect nucleic acids turnover and can serve as non-invasive biomarkers for monitoring tumour circadian dynamics, and treatment responses in patients with metastatic colorectal cancer. In 39 patients, median overnight urinary excretion of LC-HRMS determinations of pseudouridine, was ~ tenfold as large as those of 1-methylguanosine, 1-methyladenosine, or 4-acetylcytidine, and ~ 100-fold as large as those of adenosine and cytidine. An increase in any nucleoside excretion after chemotherapy anticipated plasma carcinoembryonic antigen progression 1–2 months later and was associated with poor survival. Ten fractionated urines were collected over 2-days in 29 patients. The median value of the rhythm-adjusted mean of urinary nucleoside excretion varied from 64.3 for pseudouridine down to 0.61 for cytidine. The rhythm amplitudes relative to the 24-h mean of 6 nucleoside excretions were associated with rest duration, supporting a tight link between nucleosides turnover and the rest-activity rhythm. Moreover, the amplitude of the 1-methylguanosine rhythm was correlated with the rest-activity dichotomy index, a significant predictor of survival outcome in prior studies. In conclusion, urinary excretion dynamics of modified nucleosides appeared useful for the characterization of the circadian control of cellular proliferation and for tracking early responses to treatments in colorectal cancer patients.
Collapse
|
176
|
Current advances in prognostic and diagnostic biomarkers for solid cancers: Detection techniques and future challenges. Biomed Pharmacother 2021; 146:112488. [PMID: 34894516 DOI: 10.1016/j.biopha.2021.112488] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/19/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022] Open
Abstract
Solid cancers are one of the leading causes of cancer related deaths, characterized by rapid growth of tumour, and local and distant metastases. Current advances on multimodality care have substantially improved local control and metastasis-free survival of patients by resection of primary tumour. The major concern in disease prognosis is the timely detection of resectable or metastatic tumour, thus reinforcing the need for identification of biomarkers for premalignant lesions of solid cancer. This ultimately improves the outcome for the patients. Therefore, the purpose of this review is to update the recent advancements on prognostic and diagnostic biomarkers to enhance early detection of common solid cancers including, breast, lung, colorectal, prostate and stomach cancer. We also provide an insight into Food and Drug Administration (FDA)-approved solid cancers biomarkers; various conventional techniques used for detection of prognostic and diagnostic biomarkers and discuss approaches to turn challenges in this field into opportunities.
Collapse
|
177
|
Kudelova E, Holubekova V, Grendar M, Kolkova Z, Samec M, Vanova B, Mikolajcik P, Smolar M, Kudela E, Laca L, Lasabova Z. Circulating miRNA expression over the course of colorectal cancer treatment. Oncol Lett 2021; 23:18. [PMID: 34868358 PMCID: PMC8630815 DOI: 10.3892/ol.2021.13136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/20/2021] [Indexed: 11/06/2022] Open
Abstract
Colorectal cancer (CRC) is the third-most common cancer type in males and the second-most common cancer type in females, and has the second-highest overall mortality rate worldwide. Approximately 50% of patients in stage I–III develop metastases, mostly localized to the liver. All physiological conditions occurring in the organism are also reflected in the levels of circulating microRNAs (miRNAs/miRs) in patients. miRNAs are a class of small, non-coding, single-stranded RNAs consisting of 18–25 nucleotides, which have important roles in various cellular processes. The aim of the present study was to evaluate a panel of seven circulating miRNAs (miR-106a-5p, miR-210-5p, miR-155-5p, miR-21-5p, miR-103a-3p, miR-191-5p and miR-16-5p) as biomarkers for monitoring patients undergoing adjuvant treatment of CRC. Total RNA was extracted from the plasma of patients with CRC prior to surgery, in the early post-operative period (n=60) and 3 months after surgery (n=14). The levels of the selected circulating miRNAs were measured with the miRCURY LNA miRNA PCR system and fold changes were calculated using the standard ∆∆Cq method. DIANA-miRPath analysis was used to evaluate the role of significantly deregulated miRNAs. The results indicated significant upregulation of miR-155-5p, miR-21-5p and miR-191-5p, and downregulation of miR-16-5p directly after the surgery. In paired follow-up samples, the most significant upregulation was detected for miR-106a-5p and miR-16-5p, and the most significant downregulation was for miR-21-5p. Pathway analysis outlined the role of the differentially expressed miRNAs in cancer development, but the same pathways are also involved in wound healing and regeneration of intestinal epithelium. It may be suggested that these processes should also be considered in studies investigating sensitive and easily detectable circulating biomarkers for recurrence in patients.
Collapse
Affiliation(s)
- Eva Kudelova
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Veronika Holubekova
- Biomedical Center in Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Marian Grendar
- Biomedical Center in Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Zuzana Kolkova
- Biomedical Center in Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Marek Samec
- Clinic of Gynecology and Obstetrics, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Barbora Vanova
- Biomedical Center in Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Peter Mikolajcik
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Marek Smolar
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Erik Kudela
- Clinic of Gynecology and Obstetrics, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Ludovit Laca
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| | - Zora Lasabova
- Department of Molecular Biology and Genomics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin SK-03601, Slovak Republic
| |
Collapse
|
178
|
Application of droplet digital polymerase chain reaction of plasma methylated septin 9 on detection and early monitoring of colorectal cancer. Sci Rep 2021; 11:23446. [PMID: 34873218 PMCID: PMC8648834 DOI: 10.1038/s41598-021-02879-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023] Open
Abstract
Methylated septin 9 (SEPT9) has been approved for non-invasive screening of colorectal cancer (CRC), but data on monitoring of CRC is sparse. Droplet digital polymerase chain reaction (ddPCR), with higher detection precision and simpler quantification than conventional PCR, has not been applied in SEPT9 detection. We explored the role of SEPT9 ddPCR for CRC detection and to measure serial SEPT9 levels in blood samples of CRC patients before and 3-month after surgery. SEPT9 methylated ratio, methylated abundance, and CEA levels were all higher in CRC patients than normal controls (all P < 0.05). The area under the curve (AUC) for methylated ratio and abundance to detect CRC was 0.707 and 0.710, respectively. There was an increasing trend for SEPT9 methylated abundance from proximal to distal cancers (P = 0.017). At 3-month after surgery, both methylated abundance and ratio decreased (P = 0.005 and 0.053, respectively), especially methylated abundance in stage III and distal cancer (both P < 0.01). We have developed a ddPCR platform for the quantitative detection of plasma SEPT9 in CRC patients. SEPT9 methylated abundance had an early post-operative decline, which may be useful in monitoring of treatment response.
Collapse
|
179
|
Xie X, Lin H, Zhang X, Song P, He X, Zhong J, Shi J. Overexpression of GDP dissociation inhibitor 1 gene associates with the invasiveness and poor outcomes of colorectal cancer. Bioengineered 2021; 12:5595-5606. [PMID: 34515625 PMCID: PMC8806759 DOI: 10.1080/21655979.2021.1967031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022] Open
Abstract
GDP dissociation inhibitor (GDI) regulates the GDP/GTP exchange reaction of most Rab proteins by inhibiting GDP dissociation. This study evaluated the potential prognostic and predictive value of GDI1 in colorectal cancer (CRC). To address the prognostic power of GDI1, we performed individual and pooled survival analyses on six independent CRC microarray gene expression datasets. GDI1-enriched signatures were also analyzed. Kaplan-Meier and Cox proportional analyses were employed for survival analysis. An immunohistochemistry (IHC) analysis was performed to validate the clinical relevance and prognostic significance of the GDI1 protein level in CRC tissue samples. The results revealed that GDI1 mRNA level was significantly linked with the aggressiveness of CRC, which is compatible with gene set enrichment analysis. A meta-analysis and pooled analysis demonstrated that a higher mRNA GDI1 expression was dramatically correlated with a worse survival in a dose-dependent manner in CRC patients. Further IHC analysis validated that the protein expression of GDI1 in both cytoplasm and membrane also significantly impacted the outcome of CRC patients. In CRC patients with stage III, chemotherapy significantly reduced the relative risk of death in low-GDI1 subgroup (hazard ratio (HR) = 0.22; 95% confidence interval (95% CI) 0.09-0.56, p = 0.0003), but not in high-GDI1 subgroup (HR = 0.63; 95% CI 0.35-1.14, p = 0.1137). Therefore, both high mRNA and protein levels of GDI1 were significantly related to poor outcomes in CRC patients. GD11 may serve as a prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Xiao Xie
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Huajiang Lin
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Xiaolei Zhang
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Pengtao Song
- Department of Pathology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Xiangyi He
- Department of Pathology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Jing Zhong
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Jiemin Shi
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| |
Collapse
|
180
|
Chen D, Yang X, Liu M, Zhang Z, Xing E. Roles of miRNA dysregulation in the pathogenesis of multiple myeloma. Cancer Gene Ther 2021; 28:1256-1268. [PMID: 33402729 PMCID: PMC8636266 DOI: 10.1038/s41417-020-00291-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/03/2020] [Accepted: 12/11/2020] [Indexed: 01/29/2023]
Abstract
Multiple myeloma (MM) is a malignant disease of plasma cells with complex pathology, causing significant morbidity due to its end-organ destruction. The outcomes of patients with myeloma have significantly improved in the past couple of decades with the introduction of novel agents, such as proteasome inhibitors, immunomodulators, and monoclonal antibodies. However, MM remains incurable and presents considerable individual heterogeneity. MicroRNAs (miRNAs) are short, endogenous noncoding RNAs of 19-22 nucleotides that regulate gene expression at the posttranscriptional level. Numerous studies have shown that miRNA deregulation is closely related to MM pathology, including tumor initiation, progression, metastasis, prognosis, and drug response, which make the complicated miRNA network an attractive and marvelous area of investigation for novel anti-MM therapeutic approaches. Herein, we mainly summarized the current knowledge on the roles of miRNAs, which are of great significance in regulating pathological factors involved in MM progressions, such as bone marrow microenvironment, methylation, immune regulation, genomic instability, and drug resistance. Meanwhile, their potential as novel prognostic biomarkers and therapeutic targets was also discussed.
Collapse
Affiliation(s)
- Dan Chen
- Department of Central Laboratory, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Xinhong Yang
- Department of Hematology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Min Liu
- Department of Hematology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Zhihua Zhang
- Department of Hematology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China.
| | - Enhong Xing
- Department of Central Laboratory, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China.
| |
Collapse
|
181
|
Barros AG, Pulido CF, Machado M, Brito MJ, Couto N, Sousa O, Melo SA, Mansinho H. Treatment optimization of locally advanced and metastatic pancreatic cancer (Review). Int J Oncol 2021; 59:110. [PMID: 34859257 PMCID: PMC8651228 DOI: 10.3892/ijo.2021.5290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignant tumor types, being the sixth leading cause of mortality worldwide and the fourth in Europe. Globally, it has a mortality/incidence ratio of 98%, and the 5‑year survival rate in Europe is only 3%. Although risk factors, such as obesity, diabetes mellitus, smoking, alcohol consumption and genetic factors, have been identified, the causes of PDAC remain elusive. Additionally, the only curative treatment for PDAC is surgery with negative margins. However, upon diagnosis, ~30% of the patients already present with locally advanced disease. In these cases, a multidisciplinary approach is required to improve disease‑related symptoms and prolong patient survival. In the present article, a comprehensive review of PDAC epidemiology, physiology and treatment is provided. Moreover, guidelines on patient treatment are suggested. Among the different available therapeutic options for the treatment of advanced PDAC, results are modest, most likely due to the complexity of the disease, and so the prognostic remains poor. Molecular approaches based on multi‑omics research are promising and will contribute to groundbreaking personalized medicine. Thus, economic investment that promotes research of pancreatic cancer will be critical to the development of more efficient diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Anabela G. Barros
- Department of Medical Oncology, University Hospital of Coimbra, 3004-561 Coimbra, Portugal
| | - Catarina F. Pulido
- Department of Medical Oncology, Luz Lisbon Hospital, 1500-650 Lisbon, Portugal
| | - Manuela Machado
- Department of Medical Oncology, Entre o Douro e Vouga Hospital Center (CHEDV), 4520-211 Santa Maria da Feira, Portugal
| | - Maria José Brito
- Pathologic Anatomy Department, Garcia de Orta Hospital, 2805-267 Almada, Portugal
| | - Nuno Couto
- Digestive Unit, Champalimaud Clinical Centre, 4200-135 Porto, Portugal
- Champalimaud Research Centre, 1400-038 Lisbon, 4200-135 Porto, Portugal
| | - Olga Sousa
- Radiotherapy Department, Portuguese Institute of Oncology, 4200-072 Porto, 4200-135 Porto, Portugal
| | - Sónia A. Melo
- i3S-Institute for Research and Innovation in Health of University of Porto, 4200-135 Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Hélder Mansinho
- Hemato-Oncology Department, Garcia de Orta Hospital, 2805-267 Almada, Portugal
| |
Collapse
|
182
|
Nakao T, Shimada M, Yoshikawa K, Tokunaga T, Nishi M, Kashihara H, Takasu C, Wada Y, Yoshimoto T, Yamashita S, Iwakawa Y. The correlation of immunoscore and frailty in colorectal cancer. Int J Clin Oncol 2021; 27:528-537. [PMID: 34846645 DOI: 10.1007/s10147-021-02096-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/23/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND The Immunoscore is a useful prognostic and predictive factor for colorectal cancer. Frailty predicts overall and recurrence-free survival following resection of colorectal cancer, and the immunosuppressive state of frailty might affect tumor progression. This study investigated the relationship between the Immunoscore and frailty in colorectal cancer. METHODS This retrospective study included patients who underwent radical surgery for stage II and III colorectal cancer (n = 108). Patients were divided into frail (n = 11) and non-frail (n = 97) groups, and low (IS0-2) (n = 70) and high (IS3-4) Immunoscore groups (n = 38), for comparison. RESULTS American Society of Anesthesiologists physical status was higher, tumor diameter was larger, number of well-differentiated tumors was higher, albumin was lower, 5-year overall survival (OS) was lower (frail group: 50.0%, non-frail group: 90.5%, p < 0.001) and 5-year disease-free survival (DFS) was lower (frail group: 36.4%, non-frail group: 75.2%, p = 0.024) in the frail than in the non-frail group. Left colon cancer was more, 5-year OS was lower (IS0-2 group: 82.0%, IS3-4 group: 96.7%, p = 0.040) and 5-year DFS was lower (IS0-2 group: 66.3%, IS3-4 group: 83.3%, p = 0.043) in the IS0-2 than in the IS3-4 group. The Immunoscore was lower in the frail than in the non-frail group. CONCLUSION Immunoscore and frailty are prognostic and predictive factors in colorectal cancer, and they are correlated with each other. The immunosuppressive state from frailty might affect this correlation.
Collapse
Affiliation(s)
- Toshihiro Nakao
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan.
| | - Mitsuo Shimada
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| | - Kozo Yoshikawa
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| | - Takuya Tokunaga
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| | - Masaaki Nishi
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| | - Hideya Kashihara
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| | - Chie Takasu
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| | - Yuma Wada
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| | - Toshiaki Yoshimoto
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| | - Syoko Yamashita
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| | - Yosuke Iwakawa
- Department of Digestive and Transplant Surgery, Tokushima University Hospital, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 7708503, Japan
| |
Collapse
|
183
|
Li C, Zhang D, Pang X, Pu H, Lei M, Fan B, Lv J, You D, Li Z, Zhang T. Trajectories of Perioperative Serum Tumor Markers and Colorectal Cancer Outcomes: A Retrospective, Multicenter Longitudinal Cohort Study. EBioMedicine 2021; 74:103706. [PMID: 34801967 PMCID: PMC8605392 DOI: 10.1016/j.ebiom.2021.103706] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 01/06/2023] Open
Abstract
Background The dynamic monitoring of perioperative carcinoembryonic antigen (CEA) is recommended by current colorectal cancer (CRC) guidelines, while the benefits of additional measurements of carbohydrate antigen 19-9 (CA19-9) and carbohydrate antigen 125 (CA125) have remained controversial. Methods This retrospective longitudinal cohort included 3539 CRC patients who underwent curative resection. Distinct trajectory groups were identified by the latent class growth mixed model. Patients were grouped into subgroups jointly by CEA, CA19-9, and CA125 according to preoperative levels and longitudinal trajectories, respectively. The end points were overall survival (OS) and recurrence-free survival (RFS). Findings Three distinct trajectory groups were characterized for serum CEA, CA19-9, and CA125: low-stable, early-rising, and later-rising. Jointly, patients were grouped into six preoperative (trajectory) joint groups. Compared with the three-low group, the adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) associated with death were 1.87 (1.29-2.70), 3.82 (2.37-6.17), 1.87 (0.97-3.61), 2.81 (1.93-4.11), and 4.99 (2.80-8.86) for the CEA-high, CA19-9-high, CA125-high, two-high, and three-high group, respectively. And compared with the three-stable trajectory group, the corresponding HRs (95% CIs) were 1.59 (1.10-2.30), 1.55 (0.77-3.10), 6.25 (4.02-9.70), 4.05 (2.73-6.02), and 12.40 (5.77-26.70) for the five rising trajectory groups, respectively. Similar associations between joint groups and RFS were observed. Notably, the trajectory joint group still had prognostic significance after adjusting for preoperative levels. The CA19-9-high group (HR: 3.82, 95% CI: 2.37-6.17) was associated with higher risk of death than the two-high group (HR: 2.81, 95% CI: 1.93-4.11). Likewise, for the CA125-rising trajectory group and two-rising trajectory group, the HRs (95% CIs) were 6.13 (3.75-10.00) and 3.99 (2.63-6.05) for death, and 3.08 (2.07-4.58) and 2.10 (1.52-2.90) for recurrence. Interpretation In addition to CEA, the dynamic measurements of CA19-9 and CA125 are recommended to monitor the prognosis of CRC patients. Funding National Natural Science Foundation of China [81973147, 82001986, 81960592, 82073569, 81660545].
Collapse
Affiliation(s)
- Chunxia Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Institute for Medical Dataology, Shandong University, Jinan, 250002, China
| | - Dafu Zhang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Xiaolin Pang
- Department of Radiotherapy, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Hongjiang Pu
- Department of Colorectal Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Ming Lei
- Department of Clinical Laboratory Medicine, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Bingbing Fan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Institute for Medical Dataology, Shandong University, Jinan, 250002, China
| | - Jiali Lv
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Institute for Medical Dataology, Shandong University, Jinan, 250002, China
| | - Dingyun You
- School of Public Health, Kunming Medical University, Kunming, Yunnan, 650500, China.
| | - Zhenhui Li
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China; Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Institute for Medical Dataology, Shandong University, Jinan, 250002, China.
| |
Collapse
|
184
|
Tieng FYF, Abu N, Nasir SN, Lee LH, Ab Mutalib NS. Liquid Biopsy-Based Colorectal Cancer Screening via Surface Markers of Circulating Tumor Cells. Diagnostics (Basel) 2021; 11:2136. [PMID: 34829483 PMCID: PMC8618170 DOI: 10.3390/diagnostics11112136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is ranked second for cancer-related deaths worldwide with approximately half of the patients being diagnosed at the late stages. The untimely detection of CRC results in advancement to the metastatic stage and nearly 90% of cancer-related deaths. The early detection of CRC is crucial to decrease its overall incidence and mortality rates. The recent introduction of circulating tumor cells (CTCs) has enabled a less invasive sampling method from liquid biopsies, besides revealing key information toward CRC metastasis. The current gold standard for CTC identification is the CellSearch® system (Veridex). This first-generation instrumentation relies on a single cell surface marker (CSM) to capture and count CTCs. Detection of CTCs allows the identification of patients at risk for metastasis, whereas CTC enumeration could improve risk assessment, monitoring of systemic therapy, and detection of therapy resistance in advanced metastatic CRC. In this review, we compared the pros and cons between single CSM-based CTC enrichment techniques and multi-marker-based systems. We also highlighted the challenges faced in the routine implementation of CSM-dependent CTC detection methods in CRC screening, prediction, prognosis, disease monitoring, and therapy selection toward precision medicine, as well as the dwelling on post-CTC analysis and characterization methods.
Collapse
Affiliation(s)
- Francis Yew Fu Tieng
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (F.Y.F.T.); (N.A.); (S.N.N.)
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (F.Y.F.T.); (N.A.); (S.N.N.)
| | - Siti Nurmi Nasir
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (F.Y.F.T.); (N.A.); (S.N.N.)
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University of Malaysia, Subang Jaya 47500, Selangor, Malaysia
| | - Nurul-Syakima Ab Mutalib
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (F.Y.F.T.); (N.A.); (S.N.N.)
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University of Malaysia, Subang Jaya 47500, Selangor, Malaysia
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
185
|
Kim HS, Han Y, Kang JS, Kang YH, Lee M, Sohn HJ, Kim H, Kwon W, Jang JY. Serum carcinoembryonic antigen and carbohydrate antigen 19-9 as preoperative diagnostic biomarkers of extrahepatic bile duct cancer. BJS Open 2021; 5:zrab127. [PMID: 34935900 PMCID: PMC8693162 DOI: 10.1093/bjsopen/zrab127] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/07/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Serum carcinoembryonic antigen (CEA) and carbohydrate antigen (CA) 19-9 have been proposed as useful preoperative biomarkers of extrahepatic bile duct cancer (EBDC). This study investigated the accuracy of CEA and CA19-9 for preoperative diagnosis of EBDC. METHODS Patients who underwent surgery for EBDC at a tertiary centre between 1995 and 2018 were studied, and those with concurrent hepatobiliary diseases (including gallbladder cancer, intraductal papillary mucinous neoplasms of pancreas), which could affect CEA or CA19-9 levels, were excluded. The control group included patients who underwent cholecystectomy for benign gallbladder diseases during the same period. Diagnostic accuracy was determined using sensitivity, specificity and area under the receiver operating characteristic curve (AUC). RESULTS After excluding 23 patients, 687 patients (488 men and 199 women, mean age 65.8 years) were compared with the control group of 2310 patients. Median CEA and CA19-9 levels were 1.8 μg/l and 47.0 kU/l in patients with EBDC. CEA (cut-off 5.0 μg/l) showed AUC of 0.541, sensitivity 9.0 per cent and specificity 99.2 per cent, whereas CA19-9 (cut-off 37.0 kU/l) showed AUC of 0.753, sensitivity 56.2 per cent and specificity 94.5 per cent. Sensitivity of CA19-9 was lower in early (T stages 0-II) than advanced (T stages III and IV) cancer (47.0 versus 64.9 per cent), and also lower in N0 stage cancer than lymph node metastasis (50.1 versus 68.8 per cent). CONCLUSION Serum CEA and CA19-9 showed low sensitivity limiting their usefulness as diagnostic biomarkers of EBDC.
Collapse
Affiliation(s)
- Hyeong Seok Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jae Seung Kang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon Hyung Kang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Mirang Lee
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hee Ju Sohn
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hongbeom Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
186
|
Sun Z, Ji S, Wu J, Tian J, Quan W, Shang A, Ji P, Xiao W, Liu D, Wang X, Li D. Proteomics-Based Identification of Candidate Exosomal Glycoprotein Biomarkers and Their Value for Diagnosing Colorectal Cancer. Front Oncol 2021; 11:725211. [PMID: 34737948 PMCID: PMC8560707 DOI: 10.3389/fonc.2021.725211] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/29/2021] [Indexed: 01/21/2023] Open
Abstract
Early diagnosis and treatment of colorectal cancer (CRC) significantly improves the survival rate and quality of life. Here we screened for differences in glycoproteins associated with tumor-derived exosomes and validated their clinical value to serve as liquid biopsy biomarkers to diagnosed early CRC. Exosomes were extracted from paracancerous tissues, cancer tissues, and plasma. LC-MS/MS proteomic and glycoproteomics analyses were performed using an LTQ-Orbitrap Elite mass spectrometer. The differences in glycoproteins associated with exosomes of paracancerous tissues and cancer tissue were determined, and their levels in plasma exosomes were determined. Statistical analysis was performed to evaluate the diagnostic efficacy of exosome-associated glycoproteins for CRC. We found that the levels of fibrinogen beta chain (FGB) and beta-2-glycoprotein 1 (β2-GP1) in the exosome of CRC tissue were significantly higher compared with those of paracancerous tissues exosome. The areas under the receiver operating characteristic (ROC) curves of plasma exosomal FGB and β2-GP1 as biomarkers for CRC were 0.871 (95% CI = 0.786–0.914) and 0.834 (95% CI = 0.734–0.901), respectively, compared with those of the concentrations of carcinoembryonic antigen concentration [0.723 (95% CI = 0.679–0.853)] and carbohydrate antigen19-9 concentration [0.614 (95% CI = 0.543–0.715)]. Comprehensive proteomics analyses of plasma exosomal biomarkers in CRC identified biomarkers with significant diagnostic efficacy for early CRC, which can be measured using relatively non-invasive techniques.
Collapse
Affiliation(s)
- Zujun Sun
- Department of Clinical Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shurong Ji
- Department of General Surgery, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junlu Wu
- Department of Clinical Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiale Tian
- Department of Clinical Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenqiang Quan
- Department of Clinical Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anquan Shang
- Department of Clinical Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ping Ji
- Department of Clinical Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weidong Xiao
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ding Liu
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Xuan Wang
- Department of Pharmacy, Shanghai Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dong Li
- Department of Clinical Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
187
|
Fackche N, Schmocker RK, Kubi B, Cloyd JM, Ahmed A, Grotz T, Leiting J, Fournier K, Lee AJ, Powers B, Dineen S, Veerapong J, Baumgartner JM, Clarke C, Gamblin TC, Patel SH, Dhar V, Hendrix RJ, Lambert L, Abbott DE, Pokrzywa C, Lafaro K, Lee B, Zaidi MY, Maithel SK, Johnston FM, Greer JB. The Utility of Preoperative Tumor Markers in Peritoneal Carcinomatosis from Primary Appendiceal Adenocarcinoma: an Analysis from the US HIPEC Collaborative. J Gastrointest Surg 2021; 25:2908-2919. [PMID: 33634422 DOI: 10.1007/s11605-021-04953-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 01/31/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND Prognostication based on preoperative clinical factors is lacking in patients undergoing cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC). This study aims to determine the value of preoperative tumor markers as predictors of progression-free survival (PFS) and overall survival (OS) for patients with peritoneal carcinomatosis from a primary mucinous adenocarcinoma of the appendix (MACA). METHODS We queried the United States HIPEC Collaborative, a database of patients with peritoneal carcinomatosis treated with CRS/HIPEC at twelve institutions between 2000 and 2017, identifying 409 patients with MACA. Multivariate analysis was used to identify independent predictors of disease progression. Subgroup analysis was conducted to evaluate the impact of tumor grade on the predictive value of tumor markers. RESULTS CA19-9 [HR 2.44, CI 1.2-3.4] emerged as an independent predictor of PFS while CEA [HR 4.98, CI 1.06-23.46] was independently predictive of OS (p <0.01). Tumor differentiation was the most potent predictor of both PFS (poorly differentiated vs well, [HR 4.5 CI 2.01-9.94]) and OS ([poorly differentiated vs well-differentiated: [HR 13.5, CI 3.16-57.78]), p <0.05. Among patients with combined CA19-9 elevation and poorly differentiated histology, 86% recurred within a year of CRS/HIPEC (p < 0.01). Similarly, the coexistence of CEA elevation and unfavorable histology led to the lowest survival rate at two years [36%, p < 0.01]. CA-125 was not predictive of PFS or OS. CONCLUSION Elevated preoperative CA19-9 portends worse PFS, while elevated CEA predicts worse OS after CRS/HIPEC in patients with MACA. This study provides additional evidence that CA19-9 and CEA levels should be collected during standard preoperative bloodwork, while CA-125 can likely be omitted. Tumor differentiation, when added to preoperative tumor marker levels, provides powerful prognostic information. Prospective studies are required to confirm this association.
Collapse
Affiliation(s)
- Nadege Fackche
- Division of Surgical Oncology, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Ryan K Schmocker
- Division of Surgical Oncology, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Boateng Kubi
- Division of Surgical Oncology, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan M Cloyd
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ahmed Ahmed
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Travis Grotz
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jennifer Leiting
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN, USA
| | - Keith Fournier
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Lee
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Benjamin Powers
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Department of Oncologic Sciences, Morsani College of Medicine, Tampa, FL, USA
| | - Sean Dineen
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Department of Oncologic Sciences, Morsani College of Medicine, Tampa, FL, USA
| | - Jula Veerapong
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Joel M Baumgartner
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Callisia Clarke
- Division of Surgical Oncology, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - T Clark Gamblin
- Division of Surgical Oncology, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sameer H Patel
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Vikrom Dhar
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ryan J Hendrix
- Division of Surgical Oncology, Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - Laura Lambert
- Division of Surgical Oncology, Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - Daniel E Abbott
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin, Madison, WI, USA
| | - Courtney Pokrzywa
- Division of Surgical Oncology, Department of Surgery, University of Wisconsin, Madison, WI, USA
| | - Kelly Lafaro
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, South Pasadena, CA, USA
| | - Byrne Lee
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, South Pasadena, CA, USA
| | - Mohammad Y Zaidi
- Division of Surgical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Shishir K Maithel
- Division of Surgical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Fabian M Johnston
- Division of Surgical Oncology, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Jonathan B Greer
- Division of Surgical Oncology, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
188
|
Zhou S, Sheng N, Ren J, He Q, Zhang Y, Gong J, Wang Z. Clinical Significance of and Predictive Risk Factors for the Postoperative Elevation of Carcinoembryonic Antigen in Patients With Non-Metastatic Colorectal Cancer. Front Oncol 2021; 11:741309. [PMID: 34692522 PMCID: PMC8529031 DOI: 10.3389/fonc.2021.741309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022] Open
Abstract
Background Recently, a few researches focus on the correlation between postoperative carcinoembryonic antigen (post-CEA) and the outcome of colorectal cancer (CRC), but none investigates the predictive value of post-CEA in a prognostic model. Besides, current recommendations on the frequency of post-CEA surveillance are not individualized and well followed. There is an absence of identification of patients who are more likely to have abnormal post-CEA levels and need more frequent CEA measurements. Methods Consecutive CRC patients who underwent curative surgery were enrolled and randomly divided into the discovery (n=352) and testing cohort (n=233). Impacts of preoperative CEA (pre-CEA) and post-CEA on prognosis were assessed. Cox regression model was applied to develop prognostic nomograms, which were validated by the concordance index (C-index), calibration curve, and receiver operating characteristic curve (ROC) analysis. And prediction improvement of the nomograms was assessed with net reclassification improvement (NRI) and integrated discrimination improvement (IDI). Logistic regression was used to identify predictive risk factors and construct the prediction model for post-CEA elevation. Results Post-CEA independently predicted overall survival (OS) and disease-free survival (DFS), while pre-CEA did not. Post-CEA elevation represented higher risks in patients with normal pre-CEA, compared to those with persistent elevated CEA. The nomograms for OS and DFS were established with body mass index, tumor differentiation, N stage, lymphocyte-to-monocyte ratio, and post-CEA. The nomograms showed good calibration and superior discrimination than pTNM stage, with the C-index of 0.783 and 0.759 in the discovery set and 0.712 and 0.774 in the testing set for OS and DFS, respectively. Comparisons between models using IDI and NRI implied that the nomograms performed better than pTNM stage and the predictive power could be improved with the addition of post-CEA. The prediction model for post-CEA elevation was established with age, platelet-to-lymphocyte ratio, preoperative CA19-9, and pre-CEA. The AUC of the model in the two cohorts was 0.802 and 0.764, respectively. Conclusions Elevated post-CEA was a strong indicator of poor prognosis. The addition of post-CEA significantly enhanced the performance of prognostic nomograms. And the prediction model for post-CEA elevation may help identify patients who ought to reasonably receive more intensive postoperative surveillance of CEA levels.
Collapse
Affiliation(s)
- Siyu Zhou
- Department of Gastrointestinal Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Nengquan Sheng
- Department of Gastrointestinal Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiazi Ren
- Department of Gastrointestinal Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qian He
- College of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaya Zhang
- College of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfeng Gong
- Department of Gastrointestinal Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhigang Wang
- Department of Gastrointestinal Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
189
|
Singh D, Dheer D, Samykutty A, Shankar R. Antibody drug conjugates in gastrointestinal cancer: From lab to clinical development. J Control Release 2021; 340:1-34. [PMID: 34673122 DOI: 10.1016/j.jconrel.2021.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022]
Abstract
The antibody-drug conjugates (ADCs) are one the fastest growing biotherapeutics in oncology and are still in their infancy in gastrointestinal (GI) cancer for clinical applications to improve patient survival. The ADC based approach is developed with tumor specific antigen, antibody carrying cytotoxic agents to precisely target and deliver chemotherapeutics at the tumor site. To date, 11 ADCs have been approved by US-FDA, and more than 80 are in the clinical development phase for different oncological indications. However, The ADCs based therapies in GI cancers are still far from having high-efficient clinical outcomes. The limited success of these ADCs and lessons learned from the past are now being used to develop a newer generation of ADC against GI cancers. In this review, we did a comprehensive assessment of the key components of ADCs, including tumor marker, antibody, cytotoxic payload, and linkage strategy, with a focus on technical improvement and some future trends in the pipeline for clinical translation. The various preclinical and clinical ADCs used in gastrointestinal malignancies, their target, composition and bioconjugation, along with preclinical and clinical outcomes, are discussed. The emphasis is also given to new generation ADCs employing novel mAb, payload, linker, and bioconjugation methods are also included.
Collapse
Affiliation(s)
- Davinder Singh
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Dheer
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Abhilash Samykutty
- Stephenson Comprehensive Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA.
| | - Ravi Shankar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
190
|
Wang Q, Li F, Jiang Q, Sun Y, Liao Q, An H, Li Y, Li Z, Fan L, Guo F, Xu Q, Wo Y, Ren W, Yue J, Meng B, Liu W, Zhou X. Gene Expression Profiling for Differential Diagnosis of Liver Metastases: A Multicenter, Retrospective Cohort Study. Front Oncol 2021; 11:725988. [PMID: 34631555 PMCID: PMC8493028 DOI: 10.3389/fonc.2021.725988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/18/2021] [Indexed: 02/05/2023] Open
Abstract
Background Liver metastases (LM) are the most common tumors encountered in the liver and continue to be a significant cause of morbidity and mortality. Identification of the primary tumor of any LM is crucial for the implementation of effective and tailored treatment approaches, which still represents a difficult problem in clinical practice. Methods The resection or biopsy specimens and associated clinicopathologic data were archived from seven independent centers between January 2017 and December 2020. The primary tumor sites of liver tumors were verified through evaluation of available medical records, pathological and imaging information. The performance of a 90-gene expression assay for the determination of the site of tumor origin was assessed. Result A total of 130 LM covering 15 tumor types and 16 primary liver tumor specimens that met all quality control criteria were analyzed by the 90-gene expression assay. Among 130 LM cases, tumors were most frequently located in the colorectum, ovary and breast. Overall, the analysis of the 90-gene signature showed 93.1% and 100% agreement rates with the reference diagnosis in LM and primary liver tumor, respectively. For the common primary tumor types, the concordance rate was 100%, 95.7%, 100%, 93.8%, 87.5% for classifying the LM from the ovary, colorectum, breast, neuroendocrine, and pancreas, respectively. Conclusion The overall accuracy of 93.8% demonstrates encouraging performance of the 90-gene expression assay in identifying the primary sites of liver tumors. Future incorporation of the 90-gene expression assay in clinical diagnosis will aid oncologists in applying precise treatments, leading to improved care and outcomes for LM patients.
Collapse
Affiliation(s)
- Qifeng Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China.,The Cancer of Unknown Primary Group of Pathology Committee, Chinese Research Hospital Association, Shanghai, China
| | - Fen Li
- Department of Pathology, Chengdu Second People's Hospital, Chengdu, China
| | - Qingming Jiang
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yifeng Sun
- The Canhelp Genomics Research Center, Canhelp Genomics Co., Ltd., Hangzhou, China
| | - Qiong Liao
- The Cancer of Unknown Primary Group of Pathology Committee, Chinese Research Hospital Association, Shanghai, China.,Department of Pathology, Sichuan Cancer Hospital, Chengdu, China
| | - Huimin An
- The Cancer of Unknown Primary Group of Pathology Committee, Chinese Research Hospital Association, Shanghai, China.,Department of Pathology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yunzhu Li
- Department of Pathology, Sichuan Cancer Hospital, Chengdu, China
| | - Zhenyu Li
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China
| | - Lifang Fan
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Guo
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinghua Xu
- The Cancer of Unknown Primary Group of Pathology Committee, Chinese Research Hospital Association, Shanghai, China.,The Canhelp Genomics Research Center, Canhelp Genomics Co., Ltd., Hangzhou, China.,The Institute of Machine Learning and Systems Biology, College of Electronics and Information Engineering, Tongji University, Shanghai, China.,Xuzhou Engineering Research Center of Medical Genetics and Transformation, Department of Genetics, Xuzhou Medical University, Xuzhou, China
| | - Yixin Wo
- The Canhelp Genomics Research Center, Canhelp Genomics Co., Ltd., Hangzhou, China
| | - Wanli Ren
- The Canhelp Genomics Research Center, Canhelp Genomics Co., Ltd., Hangzhou, China
| | - Junqiu Yue
- The Cancer of Unknown Primary Group of Pathology Committee, Chinese Research Hospital Association, Shanghai, China.,Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Meng
- The Cancer of Unknown Primary Group of Pathology Committee, Chinese Research Hospital Association, Shanghai, China.,Department of Pathology, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Weiping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China.,The Cancer of Unknown Primary Group of Pathology Committee, Chinese Research Hospital Association, Shanghai, China
| |
Collapse
|
191
|
Li L, Xing S, Wu M, Ao Y, Zheng X, Cai R, Han R, Li J, Li X, Zeng Q. Fecal CEA Has an Advantage in the Diagnosis of Colorectal Cancer at Early Stage. Cancer Control 2021; 28:10732748211048292. [PMID: 34615391 PMCID: PMC8504688 DOI: 10.1177/10732748211048292] [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] [Indexed: 02/06/2023] Open
Abstract
Purpose Serum carcinoembryonic antigen (SCEA) level is often measured in patients with CRC but suffers from poor sensitivity and specificity as a diagnostic biomarker. CEA is more abundant in stool than in serum, but it has not been widely studied. This study aimed to elucidate the efficacy of fecal CEA (FCEA) as a potential non-invasive biomarker for early diagnosis of CRC. Materials and Methods We retrospectively analyzed the determination of FCEA and SCEA levels by electrochemiluminescence. We evaluated the diagnostic accuracy of FCEA and SCEA levels in early-stage CRC patients and healthy controls using ROC curve. Results A total of 298 people were included: 115 patients with CRC, 35 patients with adenomatous polyp (APC), 46 patients with non-gastrointestinal cancer (NGC), and 102 healthy controls (HC). The FCEA concentrations in CRC and APC patients were significantly higher than that of NGC and HC, and this is different from SCEA expression in APC and NGC. As a diagnostic biomarker of CRC, FCEA had significantly larger AUC compared with SCEA (.802 vs .735, P < .001). For identifying early-stage colorectal cancer, FCEA showed better diagnostic efficacy (AUC: .831) than SCEA (AUC: .750), and the combination of the 2 biomarkers was even higher (AUC: .896). The sensitivity of FCEA was higher than that of SCEA (78.7% vs 29.8%). When SCEA was negative, 80.3% of CRC and 54.6% of APC cases could be identified by FCEA. Conclusion Compared with SCEA, FCEA has more advantages in the diagnosis of the early stage of colorectal cancer and adenomatous polyps.
Collapse
Affiliation(s)
- Linfang Li
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shan Xing
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Miantao Wu
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yufeng Ao
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Zheng
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rongzeng Cai
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Runkun Han
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jingcong Li
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaohui Li
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiuyao Zeng
- Department of Clinical Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 71067Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
192
|
Tanţău A, Leucuţa DC, Tanţău M, Boţan E, Zaharie R, Mândruţiu A, Tomuleasa IC. Inflammation, Tumoral Markers and Interleukin-17, -10, and -6 Profiles in Pancreatic Adenocarcinoma and Chronic Pancreatitis. Dig Dis Sci 2021; 66:3427-3438. [PMID: 33184795 DOI: 10.1007/s10620-020-06700-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Interleukin profiles can be used as biochemical markers regarding the early diagnosis of pancreatic cancer. AIMS To assess CRP, CA 19-9, CEA levels, and interleukin-6, -10, and -17 profiles in pancreatic ductal adenocarcinoma, chronic pancreatitis was compared with a control group, and the correlation with pancreatic cancer survival. METHODS A total of 87 patients were prospective divided in pancreatic cancer (n = 53), chronic pancreatitis (n = 22) ,and control group (n = 12). The diagnosis of PDAC was made histologically. The diagnosis of chronic pancreatitis was based on medical history, imaging methods, and endoscopic ultrasound. Systemic concentrations of interleukins were measured using ELISA kits. The patients were followed at 1, 3, and 6 months. RESULTS CRP, CA 19-9, and CEA were higher in the pancreatic cancer group (p < 0.001). Interleukin-10 was significantly higher in the pancreatic cancer and chronic pancreatitis groups (p < 0.001). Interleukin-17 was statistically higher in the pancreatic cancer group (p < 0.0001). The cut-off of interleukin-17 of 0.273 had a sensitivity of 90.9 and a specificity of 80.9 with a curve under ROC of 0.80 in order to differentiate between pancreatic cancer and chronic pancreatitis. The serum levels of interleukins are not correlated with the stage of the disease. CRP, CA 19-9, CEA, and interleukin-6, -10, and -17 were lower in patients with survival more than 6 months. CONCLUSIONS We detected high levels of interleukin-6, -10, and -17 in chronic pancreatitis and pancreatic cancer. Serum interleukin-17 levels can discriminate between pancreatic cancer and chronic pancreatitis. The prognostic role of interleukins needs to be established.
Collapse
Affiliation(s)
- Alina Tanţău
- The 4th Medical Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400015, Cluj-Napoca City, Cluj, Romania
- Department of Gastroenterology and Hepatology Medical Center, 400132, Cluj-Napoca City, Cluj, Romania
| | - Daniel-Corneliu Leucuţa
- Medical Informatics and Biostatistics Department, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400012, Cluj-Napoca City, Cluj, Romania.
| | - Marcel Tanţău
- The 3rd Medical Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400012, Cluj-Napoca City, Cluj, Romania
- Department of Internal Medicine and Gastroenterology, "Prof. Dr. Octavian Fodor" Regional Institute of Gastroenterology and Hepatology, 400158, Cluj-Napoca City, Cluj, Romania
| | - Emil Boţan
- Anatomopathology Department, "Regina Maria" Medical Center, 400117, Cluj-Napoca City, Cluj, Romania
| | - Roxana Zaharie
- The 3rd Medical Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400012, Cluj-Napoca City, Cluj, Romania
- Department of Internal Medicine and Gastroenterology, "Prof. Dr. Octavian Fodor" Regional Institute of Gastroenterology and Hepatology, 400158, Cluj-Napoca City, Cluj, Romania
| | - Alina Mândruţiu
- Department of Gastroenterology and Hepatology Medical Center, 400132, Cluj-Napoca City, Cluj, Romania
| | - Ionuţ-Ciprian Tomuleasa
- Hematology Department, "Prof. Dr. Ion Chiricuţă" Institute of Oncology Cluj-Napoca, 400015, Cluj-Napoca City, Cluj, Romania
| |
Collapse
|
193
|
Henry KE, Shaffer TM, Mack KN, Ring J, Ogirala A, Klein-Scory S, Eilert-Micus C, Schmiegel W, Bracht T, Sitek B, Clyne M, Reid CJ, Sipos B, Lewis JS, Kalthoff H, Grimm J. Exploiting the MUC5AC Antigen for Noninvasive Identification of Pancreatic Cancer. J Nucl Med 2021; 62:1384-1390. [PMID: 33712530 PMCID: PMC8724889 DOI: 10.2967/jnumed.120.256776] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/13/2021] [Indexed: 12/31/2022] Open
Abstract
Pancreatic cancer (PC) remains the fourth leading cause of cancer death; therefore, there is a clinically unmet need for novel therapeutics and diagnostic markers to treat this devastating disease. Physicians often rely on biopsy or CT for diagnosis, but more specific protein biomarkers are highly desired to assess the stage and severity of PC in a noninvasive manner. Serum biomarkers such as carbohydrate antigen 19-9 are of particular interest as they are commonly elevated in PC but have exhibited suboptimal performance in the clinic. MUC5AC has emerged as a useful serum biomarker that is specific for PC versus inflammation. We developed RA96, an anti-MUC5AC antibody, to gauge its utility in PC diagnosis through immunohistochemical analysis and whole-body PET in PC. Methods: In this study, extensive biochemical characterization determined MUC5AC as the antigen for RA96. We then determined the utility of RA96 for MUC5AC immunohistochemistry on clinical PC and preclinical PC. Finally, we radiolabeled RA96 with 89Zr to assess its application as a whole-body PET radiotracer for MUC5AC quantification in PC. Results: Immunohistochemical staining with RA96 distinguished chronic pancreatitis, pancreatic intraepithelial neoplasia, and varying grades of pancreatic ductal adenocarcinoma in clinical samples. 89Zr-desferrioxamine-RA96 was able to detect MUC5AC with high specificity in mice bearing capan-2 xenografts. Conclusion: Our study demonstrated that RA96 can differentiate between inflammation and PC, improving the fidelity of PC diagnosis. Our immuno-PET tracer 89Zr-desferrioxamine-RA96 shows specific detection of MUC5AC-positive tumors in vivo, highlighting the utility of MUC5AC targeting for diagnosis of PC.
Collapse
Affiliation(s)
- Kelly E Henry
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
| | - Travis M Shaffer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Radiology, Stanford University, Stanford, California
| | - Kyeara N Mack
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Janine Ring
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anuja Ogirala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | - Wolff Schmiegel
- Department of Medicine, Ruhr University Bochum, Bochum, Germany
| | - Thilo Bracht
- Medical Proteome Center, Ruhr University Bochum, Bochum, Germany
| | - Barbara Sitek
- Medical Proteome Center, Ruhr University Bochum, Bochum, Germany
| | - Marguerite Clyne
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
| | - Colm J Reid
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
| | - Bence Sipos
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Departments of Pharmacology and Radiology, Weill Cornell Medical College, New York, New York
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, Christian-Albrechts University, Kiel, Germany
| | - Jan Grimm
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Departments of Pharmacology and Radiology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
194
|
Dou R, He S, Deng Y, Wang J. Comparison of guidelines on rectal cancer: exception proves the rule? Gastroenterol Rep (Oxf) 2021; 9:290-298. [PMID: 34567560 PMCID: PMC8460091 DOI: 10.1093/gastro/goab034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/14/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022] Open
Abstract
The standard of care for early or locally advanced rectal cancer is promoted by multiple clinical practice guidelines globally, but the considerable differences between the guidelines may cause confusion. We compared the latest updated clinical practice guidelines from five professional societies/authorities: National Comprehensive Cancer Network, American Society of Colorectal Surgeons, European Society of Medical Oncology, Chinese National Health Commission, and Chinese Society of Clinical Oncology. Key evidence is discussed for a better understanding of some seemingly contradictory recommendations.
Collapse
Affiliation(s)
- Ruoxu Dou
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Siqi He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yanhong Deng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Jianping Wang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Department of Gastrointestinal Surgery, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
195
|
Shu Z, Mao D, Song Q, Xu Y, Pang P, Zhang Y. Multiparameter MRI-based radiomics for preoperative prediction of extramural venous invasion in rectal cancer. Eur Radiol 2021; 32:1002-1013. [PMID: 34482429 DOI: 10.1007/s00330-021-08242-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVES To compare multiparameter MRI-based radiomics for preoperative prediction of extramural venous invasion (EMVI) in rectal cancer using different machine learning algorithms and to develop and validate the best diagnostic model. METHODS We retrospectively analyzed 317 patients with rectal cancer. Of these, 114 were EMVI positive and 203 were EMVI negative. Radiomics features were extracted from T2-weighted imaging, T1-weighted imaging, diffusion-weighted imaging, and enhanced T1-weighted imaging of rectal cancer, followed by the dimension reduction of the features. Logistic regression, support vector machine, Bayes, K-nearest neighbor, and random forests algorithms were trained to obtain the radiomics signatures. The area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of each radiomics signature. The best radiomics signature was selected and combined with clinical and radiological characteristics to construct a joint model for predicting EMVI. Finally, the predictive performance of the joint model was assessed. RESULTS The Bayes-based radiomics signature performed well in both the training set and the test set, with the AUCs of 0.744 and 0.738, sensitivities of 0.754 and 0.728, and specificities of 0.887 and 0.918, respectively. The joint model performed best in both the training set and the test set, with the AUCs of 0.839 and 0.835, sensitivities of 0.633 and 0.714, and specificities of 0.901 and 0.885, respectively. CONCLUSIONS The joint model demonstrated the best diagnostic performance for the preoperative prediction of EMVI in patients with rectal cancer. Hence, it can be used as a key tool for clinical individualized EMVI prediction. KEY POINTS • Radiomics features from magnetic resonance imaging can be used to predict extramural venous invasion (EMVI) in rectal cancer. • Machine learning can improve the accuracy of predicting EMVI in rectal cancer. • Radiomics can serve as a noninvasive biomarker to monitor the status of EMVI.
Collapse
Affiliation(s)
- Zhenyu Shu
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dewang Mao
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiaowei Song
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuyun Xu
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Peipei Pang
- Department of Pharmaceuticals Diagnosis, GE Healthcare, Hangzhou, China
| | - Yang Zhang
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
196
|
Cortés-Hernández LE, Eslami-S Z, Costa-Silva B, Alix-Panabières C. Current Applications and Discoveries Related to the Membrane Components of Circulating Tumor Cells and Extracellular Vesicles. Cells 2021; 10:2221. [PMID: 34571870 PMCID: PMC8465935 DOI: 10.3390/cells10092221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022] Open
Abstract
In cancer, many analytes can be investigated through liquid biopsy. They play fundamental roles in the biological mechanisms underpinning the metastatic cascade and provide clinical information that can be monitored in real time during the natural course of cancer. Some of these analytes (circulating tumor cells and extracellular vesicles) share a key feature: the presence of a phospholipid membrane that includes proteins, lipids and possibly nucleic acids. Most cell-to-cell and cell-to-matrix interactions are modulated by the cell membrane composition. To understand cancer progression, it is essential to describe how proteins, lipids and nucleic acids in the membrane influence these interactions in cancer cells. Therefore, assessing such interactions and the phospholipid membrane composition in different liquid biopsy analytes might be important for future diagnostic and therapeutic strategies. In this review, we briefly describe some of the most important surface components of circulating tumor cells and extracellular vesicles as well as their interactions, putting an emphasis on how they are involved in the different steps of the metastatic cascade and how they can be exploited by the different liquid biopsy technologies.
Collapse
Affiliation(s)
- Luis Enrique Cortés-Hernández
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, CEDEX 5, 34295 Montpellier, France; (L.E.C.-H.); (Z.E.-S.)
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, CNRS, IRD, 34000 Montpellier, France
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, CEDEX 5, 34295 Montpellier, France; (L.E.C.-H.); (Z.E.-S.)
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, CNRS, IRD, 34000 Montpellier, France
| | - Bruno Costa-Silva
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal;
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, CEDEX 5, 34295 Montpellier, France; (L.E.C.-H.); (Z.E.-S.)
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, CNRS, IRD, 34000 Montpellier, France
| |
Collapse
|
197
|
Contursi A, Schiavone S, Dovizio M, Hinz C, Fullone R, Tacconelli S, Tyrrell VJ, Grande R, Lanuti P, Marchisio M, Zucchelli M, Ballerini P, Lanas A, O'Donnell VB, Patrignani P. Platelets induce free and phospholipid-esterified 12-hydroxyeicosatetraenoic acid generation in colon cancer cells by delivering 12-lipoxygenase. J Lipid Res 2021; 62:100109. [PMID: 34428433 PMCID: PMC8456051 DOI: 10.1016/j.jlr.2021.100109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/26/2021] [Accepted: 08/07/2021] [Indexed: 12/15/2022] Open
Abstract
Platelets promote tumor metastasis by inducing promalignant phenotypes in cancer cells and directly contributing to cancer-related thrombotic complications. Platelet-derived extracellular vesicles (EVs) can promote epithelial-mesenchymal transition (EMT) in cancer cells, which confers high-grade malignancy. 12S-hydroxyeicosatetraenoic acid (12-HETE) generated by platelet-type 12-lipoxygenase (12-LOX) is considered a key modulator of cancer metastasis through unknown mechanisms. In platelets, 12-HETE can be esterified into plasma membrane phospholipids (PLs), which drive thrombosis. Using cocultures of human platelets and human colon adenocarcinoma cells (line HT29) and LC-MS/MS, we investigated the impact of platelets on cancer cell biosynthesis of 12S-HETE and its esterification into PLs and whether platelet ability to transfer its molecular cargo might play a role. To this aim, we performed coculture experiments with CFSE[5-(and-6)-carboxyfluorescein diacetate, succinimidyl ester]-loaded platelets. HT29 cells did not generate 12S-HETE or express 12-LOX. However, they acquired the capacity to produce 12S-HETE mainly esterified in plasmalogen phospholipid forms following the uptake of platelet-derived medium-sized EVs (mEVs) expressing 12-LOX. 12-LOX was detected in plasma mEV of patients with adenomas/adenocarcinomas, implying their potential to deliver the protein to cancer cells in vivo. In cancer cells exposed to platelets, endogenous but not exogenous 12S-HETE contributed to changes in EMT gene expression, mitigated by three structurally unrelated 12-LOX inhibitors. In conclusion, we showed that platelets induce the generation of primarily esterified 12-HETE in colon cancer cells following mEV-mediated delivery of 12-LOX. The modification of cancer cell phospholipids by 12-HETE may functionally impact cancer cell biology and represent a novel target for anticancer agent development.
Collapse
Affiliation(s)
- Annalisa Contursi
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy
| | - Simone Schiavone
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy
| | - Melania Dovizio
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy
| | - Christine Hinz
- Systems Immunity Research Institute and Division of Infection and Immunity, School of Medicine Cardiff University, Cardiff, United Kingdom
| | - Rosa Fullone
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy
| | - Stefania Tacconelli
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy
| | - Victoria J Tyrrell
- Systems Immunity Research Institute and Division of Infection and Immunity, School of Medicine Cardiff University, Cardiff, United Kingdom
| | - Rosalia Grande
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Medicine and Aging Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Marco Marchisio
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Medicine and Aging Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Mirco Zucchelli
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy
| | - Patrizia Ballerini
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G. d'Annunzio" University, Chieti, Italy
| | - Angel Lanas
- University Hospital LB, Aragon Health Research Institute (IISAragon), CIBERehd, University of Zaragoza, Zaragoza, Spain
| | - Valerie B O'Donnell
- Systems Immunity Research Institute and Division of Infection and Immunity, School of Medicine Cardiff University, Cardiff, United Kingdom
| | - Paola Patrignani
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy.
| |
Collapse
|
198
|
Marliot F, Chen X, Kirilovsky A, Sbarrato T, El Sissy C, Batista L, Van den Eynde M, Haicheur-Adjouri N, Anitei MG, Musina AM, Scripcariu V, Lagorce-Pagès C, Hermitte F, Galon J, Fieschi J, Pagès F. Analytical validation of the Immunoscore and its associated prognostic value in patients with colon cancer. J Immunother Cancer 2021; 8:jitc-2019-000272. [PMID: 32448799 PMCID: PMC7253006 DOI: 10.1136/jitc-2019-000272] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND New and fully validated tests need to be brought into clinical practice to improve the estimation of recurrence risk in patients with colon cancer. The aim of this study was to assess the analytical performances of the Immunoscore (IS) and show its contribution to prognosis prediction. METHODS Immunohistochemical staining of CD3+ and CD8+ T cells on adjacent sections of colon cancer tissues were quantified in the core of the tumor and its invasive margin with dedicated IS modules integrated into digital pathology software. Staining intensity across samples collected between 1989 and 2016 (n=595) was measured. The accuracy of the IS workflow was established by comparing optical and automatic counts. Analytical precision of the IS was evaluated within individual tumor block on distant sections and between eligible blocks. The IS interlaboratory reproducibility (n=100) and overall assay precision were assessed (n=3). Contribution of the IS to prediction of recurrence based on clinical and molecular parameters was determined (n=538). RESULTS Optical and automatic counts for CD3+ or CD8+ were strongly correlated (r=0.94, p<0.001 and r=0.92, p<0.001, respectively). CD3 and CD8 staining intensities were not altered by the age of the tumor block over a period of 30 years. Neither the position of tested tissue sections within a tumor block nor the selection of the tissue blocks affected the IS. Reproducibility of the IS was not affected by multiple variables (eg, antibody lots, DAB revelation kits, immunohistochemistry automates and operators). Interassay repeatability of the IS was 100% and interlaboratory reproducibility between two testing centers was 93%. Finally, in a case series of patients with stage II-III colon cancer, the relative proportion of variance for time to recurrence was greatest for the IS (53% of prognostic variability) in a model that included IS, T-stage, microsatellite instability status and total number of lymph nodes. CONCLUSION IS is a robust and validated clinical assay leveraging immune scoring to predict recurrence risk of patient with localized colon cancer. The strong and independent prognostic value of IS should pave the way for it use in clinical practice.
Collapse
Affiliation(s)
- Florence Marliot
- Laboratory of Integrative Immunology and cancerology, INSERM, University of Paris, Cordeliers Research centre, Immunomonitoring Platform, Laboratory of Immunology, AP-HP (Assistance Publique-Hôpitaux de Paris) Hôpital Européen Georges Pompidou, Paris, France
| | - Xiaoyi Chen
- Laboratory of Information Sciences to Support Personalized Medicine, Cordeliers Research Centre, Paris, France
| | - Amos Kirilovsky
- Laboratory of Integrative Immunology and cancerology, INSERM, University of Paris, Cordeliers Research centre, Immunomonitoring Platform, Laboratory of Immunology, AP-HP (Assistance Publique-Hôpitaux de Paris) Hôpital Européen Georges Pompidou, Paris, France
| | | | - Carine El Sissy
- Laboratory of Integrative Immunology and cancerology, INSERM, University of Paris, Cordeliers Research centre, Immunomonitoring Platform, Laboratory of Immunology, AP-HP (Assistance Publique-Hôpitaux de Paris) Hôpital Européen Georges Pompidou, Paris, France
| | | | - Marc Van den Eynde
- Department of clinical and medical oncology, Cliniques universitaires Saint-Luc Institut Roi Albert II, Bruxelles, Belgium
| | - Nacilla Haicheur-Adjouri
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP (Assistance Publique-Hôpitaux de Paris) Hôpital Européen Georges Pompidou, Paris, France
| | - Maria-Gabriela Anitei
- Department of Surgery, Grigore T Popa University of Medicine and Pharmacy Faculty of Medicine, Iasi, Romania
| | - Ana-Maria Musina
- Department of Surgery, Grigore T Popa University of Medicine and Pharmacy Faculty of Medicine, Iasi, Romania
| | - Viorel Scripcariu
- Department of Surgery, Grigore T Popa University of Medicine and Pharmacy Faculty of Medicine, Iasi, Romania
| | - Christine Lagorce-Pagès
- Laboratory of Integrative Immunology and cancerology, INSERM, University of Paris, Cordeliers Research centre, Immunomonitoring Platform, Laboratory of Immunology, AP-HP (Assistance Publique-Hôpitaux de Paris) Hôpital Européen Georges Pompidou, Paris, France
| | | | - Jérôme Galon
- Laboratory of Integrative Immunology and cancerology, INSERM, University of Paris, Cordeliers Research centre, Immunomonitoring Platform, Laboratory of Immunology, AP-HP (Assistance Publique-Hôpitaux de Paris) Hôpital Européen Georges Pompidou, Paris, France
| | | | - Franck Pagès
- Laboratory of Integrative Immunology and cancerology, INSERM, University of Paris, Cordeliers Research centre, Immunomonitoring Platform, Laboratory of Immunology, AP-HP (Assistance Publique-Hôpitaux de Paris) Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
199
|
Dai C, Zhang X, Ma Y, Chen Z, Chen S, Zhang Y, Li M. Serum macrophage inhibitory cytokine-1 serves as a novel diagnostic biomarker of early-stage colorectal cancer. Biomarkers 2021; 26:598-605. [PMID: 34266319 DOI: 10.1080/1354750x.2021.1950209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Patients with colorectal cancer usually have a poor prognosis because of the absence of suitable biomarkers for diagnosing asymptomatic patients. Here we determined the ability of MIC-1 to detect precancerous lesions and CRC in an asymptomatic cohort from CRC Screening Program. METHODS We screened 2759 subjects with risk factors. Endoscopic and histopathological analyses revealed that 19 and 47 subjects had CRC or precancerous lesions. We randomly selected 24 subjects with normal colonoscopies as healthy controls. We used receiver operating characteristic curve analysis to evaluate the diagnostic efficacy of MIC-1 for CRC and precancerous lesions. RESULTS The optimal thresholds of MIC-1 levels with precancerous lesions or CRC were 314.12 pg/mL (sensitivity, 91.50%; specificity, 54.20%) and 357.64 pg/mL (sensitivity, 82.40%; specificity, 70.80%). Moreover, MIC-1 levels distinguished precancerous lesions better than CEA, CA19-9, or CA24-2 (AUC: 0.760 vs. 0.529, 0.624, and 0.585) or CRC (AUCs: 0.821 vs. 0.743, 0.657, and 0.688) from the healthy controls. The combination of MIC-1, CEA, CA19-9, and CA24-2 showed the highest in sensitivity and specificity for CRC diagnosis (sensitivity, 94.10%; specificity, 87.50%). CONCLUSIONS Serum MIC-1 levels increased the sensitivity of detection of precancerous colorectal lesions and CRC and can be used to improve screening.
Collapse
Affiliation(s)
- Chunyang Dai
- Department of Laboratory Diagnostics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaolei Zhang
- Department of Laboratory Diagnostics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yanling Ma
- Office for Cancer Prevention and Control, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhaowu Chen
- Department of Laboratory Diagnostics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shaohua Chen
- Department of Laboratory Diagnostics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yang Zhang
- Department of Laboratory Diagnostics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ming Li
- Department of Laboratory Diagnostics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
200
|
CEA increase as a marker of disease progression after first-line induction therapy in metastatic colorectal cancer patients. A pooled analysis of TRIBE and TRIBE2 studies. Br J Cancer 2021; 125:839-845. [PMID: 34253871 DOI: 10.1038/s41416-021-01483-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 06/05/2021] [Accepted: 06/30/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND In mCRC, CEA is used to monitor response to systemic therapy together with imaging. After the end of induction, no major improvement in tumour shrinkage is expected, and the availability of a marker able to predict progressive disease (PD) versus no-PD might allow avoiding CT scans. METHODS We pooled data from patients with baseline CEA ≥ 10 ng/mL included in TRIBE and TRIBE2 studies with the aim of identifying a threshold for percent increase of CEA from nadir able to predict PD after the end of the induction therapy. RESULTS In total, 1178 paired CEA and radiological assessments from 434 patients were included. According to the optimal cut-off determined by ROC, a CEA increase of at least 120% from nadir differentiated between PD and no-PD with a sensitivity of 74% and a specificity of 78%, excluding PD in the 92% of radiological assessments and allowing to avoid the 67% of CT scans. However, CEA cut-off of 120% was not able to detect radiological PD in 26% of cases. In order to mitigate this issue, a different clinically relevant threshold was evaluated based on the best sensitivity cut-off. Therefore, using any CEA increase from nadir as a threshold, the sensitivity grew to 93% and only in the 7% of cases the radiological PD was not detected. CONCLUSIONS In mCRC with baseline CEA ≥ 10 ng/mL, CEA values can accurately predict PD versus no-PD after the end of the first-line induction therapy.
Collapse
|