1
|
Martínez-Espartosa D, Alegre E, Casero-Ramírez H, Díaz-Garzón J, Fernández-Calle P, Fuentes-Bullejos P, Varo N, González Á. Clinical utility of personalized reference intervals for CEA in the early detection of oncologic disease. Clin Chem Lab Med 2024; 0:cclm-2024-0546. [PMID: 39101454 DOI: 10.1515/cclm-2024-0546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024]
Abstract
OBJECTIVES Personalized reference intervals (prRI) have been proposed as a diagnostic tool for assessing measurands with high individuality. Here, we evaluate clinical performance of prRI using carcinoembryonic antigen (CEA) for cancer detection and compare it with that of reference change values (RCV) and other criteria recommended by clinical guidelines (e.g. 25 % of change between consecutive CEA results (RV25) and the cut-off point of 5 μg/L (CP5)). METHODS Clinical and analytical data from 2,638 patients collected over 19 years were retrospectively evaluated. A total 15,485 CEA results were studied. For each patient, we calculated prRI and RCV using computer algorithms based on the combination of different strategies to assess the number of CEA results needed, consideration of one or two limits of reference interval and the intraindividual biological variation estimate (CVI) used: (a) publicly available (CVI-EU), (b) CVI calculated using an indirect method (CVI-NOO) and (c) within-person BV (CVP). For each new result identified falling outside the prRI, exceeding the RCV interval, RV25 or CP5, we searched for records identifying the presence of tumour at 3 and 12 months after the test. The sensitivity, specificity and predictive power of each strategy were calculated. RESULTS PrRI approaches derived using CVI-EU, and both limits of reference interval achieve the best sensitivity (87.5 %) and NPV (99.3 %) at 3 and 12 months of all evaluated criteria. Only 3 results per patients are enough to calculate prRIs that reach this diagnostic performance. CONCLUSIONS PrRI approaches could be an effective tool to rule out new oncological findings during the active surveillance of patients.
Collapse
Affiliation(s)
| | - Estíbaliz Alegre
- Biochemistry Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | | | - Jorge Díaz-Garzón
- Department of Laboratory Medicine, Hospital Universitario La Paz, Madrid, Spain
| | | | | | - Nerea Varo
- Biochemistry Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - Álvaro González
- Biochemistry Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| |
Collapse
|
2
|
Trapé J, Bérgamo S, González-Fernández C, Rives J, González-García L. Variations in tumor growth, intra-individual biological variability, and the interpretation of changes. Clin Chem Lab Med 2024; 62:1618-1625. [PMID: 38369758 DOI: 10.1515/cclm-2023-0780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/02/2024] [Indexed: 02/20/2024]
Abstract
OBJECTIVES The identification of changes in tumor markers (TMs) in cancer patients that indicate response to treatment, stabilization or disease progression is a challenge for laboratory medicine. Several approaches have been proposed: assessing percentage increases, applying discriminant values, and estimating half-life (t1/2) or doubling time (DT). In all of them it is assumed that the TM is a surrogate of the variation in tumor size. In general this variation is time-dependent, but this is not the case of intraindividual biological variability (CVi), which can range from 6 % in CA15-3 to 22 % in CA125. When decisions are made on the basis of DT or t1/2, these values can be affected by the CVi; if it is very large, the growth rate very slow and the period of time between determinations very short, the result obtained for DT may be due mainly to the CVi. The aim of this study is to establish the relationship between the CVi and temporal variables. METHODS We related equations for calculating DT and t1/2 to the reference change values in tumor markers. RESULTS The application of the formula obtained allows the calculation of the optimal time between measurements to ensure that the influence of the CVi is minimal in different types of tumors and different scenarios. CONCLUSIONS Intraindividual variation affects the calculation of DT and t1/2. It is necessary to establish the minimum time between two measurements to ensure that the CVi does not affect their calculation or lead to misinterpretation.
Collapse
Affiliation(s)
- Jaume Trapé
- Laboratory Medicine Department, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Catalonia, Spain
- Tissue Repair and Regeneration Laboratory (TR2Lab), Centre for Health and Social Care Research (CESS), University of Vic - Central University of Catalonia, Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Vic, Spain
- Faculty of Medicine, University of Vic - Central University of Catalonia, Vic, Spain
| | - Silvia Bérgamo
- Laboratory Medicine Department, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Catalonia, Spain
- Tissue Repair and Regeneration Laboratory (TR2Lab), Centre for Health and Social Care Research (CESS), University of Vic - Central University of Catalonia, Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Vic, Spain
- Doctoral School, University of Vic - Central University of Catalonia, Vic, Spain
| | - Carolina González-Fernández
- Laboratory Medicine Department, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Catalonia, Spain
- Gastrointestinal Oncology, Endoscopy and Surgery research group (GOES), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Vic, Spain
| | - José Rives
- Laboratory Medicine Department, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Catalonia, Spain
| | - Laura González-García
- Laboratory Medicine Department, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Catalonia, Spain
- Tissue Repair and Regeneration Laboratory (TR2Lab), Centre for Health and Social Care Research (CESS), University of Vic - Central University of Catalonia, Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Vic, Spain
| |
Collapse
|
3
|
Cheng Y, Chen B, Fang Q, Zang G, Yao J. Analysis of tumor abnormal protein expression and epidermal growth factor receptor mutation status in non-small cell lung cancer. Discov Oncol 2024; 15:274. [PMID: 38980474 PMCID: PMC11233477 DOI: 10.1007/s12672-024-01094-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND The level of tumor abnormal protein (TAP) level has a significant impact on tumor growth, recurrence, and metastasis. Previous studies have highlighted the influence of the mutations in exons 19 and 21 of the epidermal growth factor receptor (EGFR), particularly the sensitivity displayed by tumor cells to epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) therapy. Our study is centered on exploring the clinical relevance of TAP and EGFR mutations in patients with non-small cell lung cancer (NSCLC). MATERIAL AND METHODS In this study, tissue samples were collected from a total of 176 patients diagnosed with non-small cell lung cancer (NSCLC). Real-time PCR technology was utilized to detect mutations within exons 19 and 21 of the epidermal growth factor receptor (EGFR) gene in these samples. This approach enables precise identification of EGFR mutations associated with NSCLC. Furthermore, the study investigated the impact of various tumor markers, including tumor abnormal protein (TAP) and carcinoembryonic antigen (CEA), on EGFR mutation status. Established assays were employed to evaluate TAP and CEA levels, aiming to ascertain their potential correlation with EGFR mutation in NSCLC patients. RESULTS EGFR exhibited mutation rates of 23.86% and 12.50% in exons 19 and 21, respectively. EGFR mutations were more prevalent in younger women (< 60 years old) and in cases with pleural invasion, vessel invasion, CEA > 6.5 ng/mL, and TAP > 228 µm2 for both genders. Increased TAP levels independently predicted EGFR mutations (P = 0.001 for males; P = 0.000 for females). An area under the curve (AUC) of 0.833 indecated EGFR mutation prediction with sensitivity and specificity of 79.7% and 87.0%, respectively. For females, the sensitivity increased to 89.7% and specificity increased to 93.8%. CONCLUSIONS TAP effectively predicts EGFR mutations in NSCLC patients with moderate accuracy, particularly benefiting diagnosis in females with high sensitivity and specificity. Integrating TAP assessment into EGFR mutation testing can significantly enhance diagnostic precision, especially in female NSCLC cases.
Collapse
Affiliation(s)
- Yuanjun Cheng
- Department of Cardiothoracic Surgery, People's Hospital of Chizhou, No. 3 Baiya Road, Guichi District, Chizhou, 247000, China
| | - Bin Chen
- Department of Cardiothoracic Surgery, People's Hospital of Chizhou, No. 3 Baiya Road, Guichi District, Chizhou, 247000, China
| | - Qianru Fang
- Department of Obstetrics, People's Hospital of Chizhou, Chizhou, 247000, China
| | - Guohui Zang
- Department of Cardiothoracic Surgery, People's Hospital of Chizhou, No. 3 Baiya Road, Guichi District, Chizhou, 247000, China
| | - Jie Yao
- Department of Cardiothoracic Surgery, People's Hospital of Chizhou, No. 3 Baiya Road, Guichi District, Chizhou, 247000, China.
| |
Collapse
|
4
|
Kikuchi Y, Shimada H, Yamasaki F, Yamashita T, Araki K, Horimoto K, Yajima S, Yashiro M, Yokoi K, Cho H, Ehira T, Nakahara K, Yasuda H, Isobe K, Hayashida T, Hatakeyama S, Akakura K, Aoki D, Nomura H, Tada Y, Yoshimatsu Y, Miyachi H, Takebayashi C, Hanamura I, Takahashi H. Clinical practice guidelines for molecular tumor marker, 2nd edition review part 2. Int J Clin Oncol 2024; 29:512-534. [PMID: 38493447 DOI: 10.1007/s10147-024-02497-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 03/19/2024]
Abstract
In recent years, rapid advancement in gene/protein analysis technology has resulted in target molecule identification that may be useful in cancer treatment. Therefore, "Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition" was published in Japan in September 2021. These guidelines were established to align the clinical usefulness of external diagnostic products with the evaluation criteria of the Pharmaceuticals and Medical Devices Agency. The guidelines were scoped for each tumor, and a clinical questionnaire was developed based on a serious clinical problem. This guideline was based on a careful review of the evidence obtained through a literature search, and recommendations were identified following the recommended grades of the Medical Information Network Distribution Services (Minds). Therefore, this guideline can be a tool for cancer treatment in clinical practice. We have already reported the review portion of "Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition" as Part 1. Here, we present the English version of each part of the Clinical Practice Guidelines for Molecular Tumor Marker, Second Edition.
Collapse
Affiliation(s)
| | - Hideaki Shimada
- Department of Clinical Oncology, Toho University, Tokyo, Japan.
- Department of Surgery, Toho University, Tokyo, Japan.
| | - Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Taku Yamashita
- Department of Otorhinolaryngology-Head and Neck Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Koji Araki
- Department of Otorhinolaryngology-Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - Kohei Horimoto
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Keigo Yokoi
- Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Haruhiko Cho
- Department of Surgery, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Takuya Ehira
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazunari Nakahara
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Hiroshi Yasuda
- Department of Gastroenterology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazutoshi Isobe
- Division of Respiratory Medicine, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Tetsu Hayashida
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shingo Hatakeyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | | | - Daisuke Aoki
- International University of Health and Welfare Graduate School, Tokyo, Japan
| | - Hiroyuki Nomura
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Yuji Tada
- Department of Pulmonology, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Yuki Yoshimatsu
- Department of Patient-Derived Cancer Model, Tochigi Cancer Center Research Institute, Tochigi, Japan
| | - Hayato Miyachi
- Faculty of Clinical Laboratory Sciences, Nitobe Bunka College, Tokyo, Japan
| | - Chiaki Takebayashi
- Division of Hematology and Oncology, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Ichiro Hanamura
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Aichi, Japan
| | | |
Collapse
|
5
|
Li X, Hu M, Wang Z, Liu M, Chen Y. Prevalence of diverse colorectal polyps and risk factors for colorectal carcinoma in situ and neoplastic polyps. J Transl Med 2024; 22:361. [PMID: 38632639 PMCID: PMC11022362 DOI: 10.1186/s12967-024-05111-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 03/20/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Most colorectal cancers originate from precancerous polyps. This study aimed to determine the prevalence of colorectal polyps with diverse pathological morphologies and to explore the risk factors for colorectal carcinoma in situ (CCS) and neoplastic polyps. METHODS Inpatients admitted from January 2018 to May 2023 were screened through the hospital information system. Polyps were classified according to pathological morphology. The prevalence of polyps was described by frequency and 95% confidence interval. Univariate and multivariate logistic regression analyses were used to explore the risk factors for CCS and neoplastic polyps. RESULTS In total, 2329 individuals with 3550 polyps were recruited. Among all patients, 76.99% had neoplastic polyps and 44.31% had advanced adenomas. Tubular adenoma had the highest prevalence at 60.15%, and the prevalence of CCS was 3.86%. Patients with a colorectal polyp diameter ≥ 1.0 cm or number ≥ 3 were 8.07 times or 1.98 times more likely to develop CCS than were those with a diameter < 1.0 cm or number < 3, respectively (OR 8.07, 95%CI 4.48-14.55, p < 0.0001; and OR 1.98, 95%CI 1.27-3.09, p = 0.002). The risk of CCS with schistosome egg deposition was also significantly increased (OR 2.70, 95%CI 1.05-6.98). The higher the levels of carbohydrate antigen (CA) 724 (OR 1.01, 95%CI 1.00-1.02) and CA211 (OR 1.16, 95%CI 1.03-1.32) in patients with colorectal polyps were, the greater the risk of CCS. When colorectal neoplastic polyps were analyzed, we discovered that for each 1-year increase in age, the risk of neoplastic polyps increased by 3% (OR 1.03, 95%CI 1.02-1.04), p < 0.0001. Patients with a polyp diameter ≥ 1.0 cm had a 2.11-fold greater risk of neoplastic polyps compared to diameter < 1.0 cm patients (OR 3.11, 95%CI 2.48-3.92), p < 0.0001. In addition, multiple polyps and CA199 levels are risk factors for neoplastic polyps. CONCLUSION More than 3/4 of colorectal polyp patients have neoplastic polyps. Patients are more inclined to develop CCS and neoplastic polyps if they have large polyps (> 1.0 cm) or multifocal polyps. The levels of the tumor markers CA724 and CA211 show some potential usefulness for predicting CCS and may be exploited for early identification of high-risk populations.
Collapse
Affiliation(s)
- Xiaojuan Li
- Department of Gastroenterology, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Mengting Hu
- Department of General Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Zhangjun Wang
- Proprietary Trading Department, Huaan Securities Co., Ltd, Shanghai, 200120, China
| | - Mei Liu
- Department of General Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China.
| | - Ying Chen
- Department of Gastroenterology, Minhang Hospital, Fudan University, Shanghai, 201199, China.
| |
Collapse
|
6
|
Morikawa H, Oba T, Kitazawa A, Iji R, Kiyosawa N, Amitani M, Shimizu T, Kanai T, Uehara T, Ito KI. CA19-9 producing locally advanced papillary thyroid carcinoma: a case report. Surg Case Rep 2024; 10:83. [PMID: 38598167 PMCID: PMC11006640 DOI: 10.1186/s40792-024-01887-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND CA19-9 is a tumor marker for gastrointestinal and biliary-pancreatic adenocarcinomas; however, its association with thyroid cancer is unknown. Here, we report a case of CA19-9 producing locally advanced papillary thyroid carcinoma (PTC). CASE PRESENTATION A 66-year-old woman who was identified with a thyroid tumor after a close examination of an elevated serum CA19-9 level, which was detected at health screening, was referred to our hospital. Ultrasonography revealed a 34 × 31 mm hypoechoic lesion in the lower pole of the left thyroid lobe. Computed tomography revealed a solid thyroid tumor with tracheal invasion without any distant metastases. Bronchoscopy revealed tumor exposure into the tracheal lumen on the left side of the trachea. Fine-needle aspiration cytology led to a diagnosis of papillary thyroid carcinoma (PTC). The patient underwent a total thyroidectomy, tracheal sleeve resection with end-to-end anastomosis, and lymph node dissection in the left cervical and superior mediastinal regions (D3c) with a reversed T-shaped upper sternotomy down to the third intercostal level. Histopathological analysis confirmed the diagnosis of PTC with tracheal invasion and no lymph node metastases (pT4a Ex2 N0). Immunohistochemical staining showed the expression of CA19-9 in cancer cells. Postoperatively, the serum CA19-9 level of the patient decreased to within the normal range. CONCLUSIONS Some PTCs produce CA19-9, although less frequently. When elevated serum CA19-9 levels are observed, PTC should be included in the differential diagnosis for further investigation.
Collapse
Affiliation(s)
- Hiroki Morikawa
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Takaaki Oba
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan.
| | - Ayaka Kitazawa
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Ryoko Iji
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Nami Kiyosawa
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Masatsugu Amitani
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Tadafumi Shimizu
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Toshiharu Kanai
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Takeshi Uehara
- Division of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Ken-Ichi Ito
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| |
Collapse
|
7
|
Kobayashi S, Hiwasa T, Kitamura K, Kano M, Hoshino T, Hirano S, Hashimoto M, Seimiya M, Shimada H, Nomura F, Matsubara H, Matsushita K. Combinational antibody detection approach increases the clinical validity of colorectal cancer screening. J Clin Lab Anal 2023; 37:e24978. [PMID: 37964630 PMCID: PMC10749486 DOI: 10.1002/jcla.24978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/27/2023] [Accepted: 10/15/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND At different stages of the disease, biomarkers can help to determine disease progression and recurrence and provide a personalized indicator of therapeutic effectiveness. The serological identification of antigens by recombinant cDNA expression cloning (SEREX) has identified five SEREX antigens. RESULTS Compared with healthy donors, anti-FIRΔexon2 and anti-SOHLH antibodies (Abs) in the sera of patients with colorectal cancer (CRC) were markedly higher. Furthermore, no correlation was noted between five SEREX antigens and the three tumor markers (CEA, CA19-9, and anti-p53 Abs), indicating that anti-FIRΔexon2 Abs are an independent candidate marker for patients with CRC. Generally, the levels of anti-FIRΔexon2 Abs combined with clinically available tumor markers were determined to be significantly higher compared with CEA, CA19-9. Moreover, in early-stage CRC, the levels of anti-FIRΔexon2 Abs combined with existing tumor markers were higher than those of CEA, CA19-9. CONCLUSION Due to the highly heterogeneous nature of CRC, a single tumor marker is unlikely to become a standalone diagnostic test due to its commonly insufficient sensitivity and/or specificity. Using a combination antibody detection approach of tumor markers for CRC diagnosis has the potential to be an effective approach. Therefore, the use of serum protein biomarker candidates holds promise for the development of inexpensive, noninvasive, and inexpensive tests for the detection of CRC.
Collapse
Affiliation(s)
- Sohei Kobayashi
- Department of Laboratory Medicine & Division of Clinical GeneticsChiba University HospitalChibaJapan
- Department of Medical Technology & Sciences, School of Health Sciences at NaritaInternational University of Health and WelfareChibaJapan
| | - Takaki Hiwasa
- Department of Neurological Surgery, Graduate School of MedicineChiba UniversityChibaJapan
| | - Kouichi Kitamura
- Department of Laboratory Medicine & Division of Clinical GeneticsChiba University HospitalChibaJapan
| | - Masayuki Kano
- Department of Frontier Surgery, Graduate School of MedicineChiba UniversityChibaJapan
| | - Tyuji Hoshino
- Department of Physical Chemistry, Graduate School of Pharmaceutical SciencesChiba UniversityChibaJapan
| | - Sho Hirano
- Department of Medical Technology & Sciences, School of Health Sciences at NaritaInternational University of Health and WelfareChibaJapan
| | - Mayuko Hashimoto
- Department of Medical Technology & Sciences, School of Health Sciences at NaritaInternational University of Health and WelfareChibaJapan
| | - Masanori Seimiya
- Department of Medical Technology & Sciences, School of Health Sciences at NaritaInternational University of Health and WelfareChibaJapan
| | - Hideaki Shimada
- Department of Gastroenterological Surgery, Graduate School of MedicineToho UniversityTokyoJapan
| | - Fumio Nomura
- Department of Laboratory Medicine & Division of Clinical GeneticsChiba University HospitalChibaJapan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of MedicineChiba UniversityChibaJapan
| | - Kazuyuki Matsushita
- Department of Laboratory Medicine & Division of Clinical GeneticsChiba University HospitalChibaJapan
| |
Collapse
|
8
|
Lerch M, Sengul D, Sengul I, Peteja M, Přibylová L, Gawel W, Pelikán A, Tomaskova H, Malý T, Janout V. Revisiting ab initio carcinoembryonic antigen and CA19-9 tumor markers in colorectal carcinoma in association with anatomotopographic location and staging of disease. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230627. [PMID: 37729380 PMCID: PMC10508941 DOI: 10.1590/1806-9282.20230627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/03/2023] [Indexed: 09/22/2023]
Abstract
OBJECTIVE This study purposed to evaluate preoperative two tumor markers, namely, carcinoembryonic antigen and carbohydrate antigen (CA)19-9, in colorectal cancer for anatomotopographic location with disease stage and to assess their utility for diagnostic staging purposes. METHODS The study retrospectively incorporated patients who had undergone surgery for colorectal cancer at our department in 2015-2018 and in whom carcinoembryonic antigen and CA19-9 tumor markers had been preoperatively analyzed. The obtained data were then statistically processed using R-project. RESULTS A total of 155 patients had been incorporated, of whom 96 (62%) were men and 59 (38%) were women. Rectum was the most common location (74 patients, 48%), and the least represented stage was IV (18, 12%). The marker carcinoembryonic antigen was obtained in all 155 cases, while CA19-9 was in 105. The median carcinoembryonic antigen was 3 (0.34-1104.25), and the median CA19-9 was 12 (0.18-840.00). A significance was recognized between median carcinoembryonic antigen and disease stage (p-value=0.016), with stages I, II, and III (medians 2, 3, and 2) different from stage IV (median 13), while no significance for CA19-9 was recognized (p-value=0.343). No significance between either marker and location (carcinoembryonic antigen: p=0.276; CA19-9: p=0.505) was detected. The testing was performed at a significance level of alpha=0.05. CONCLUSION This study revealed a significance between the marker carcinoembryonic antigen, but not CA19-9, and the disease stage, while no relationship of either of these markers with tumor location was found. Herewith, the study confirmed that higher carcinoembryonic antigen values may suggest the finding of more advanced forms of colorectal cancer and thus a worse prognosis of this malignant phenomenon.
Collapse
Affiliation(s)
- Milan Lerch
- Silesian Hospital in Opava, Department of Surgery - Opava, Czech Republic
- University of Ostrava, Faculty of Medicine, Department of Surgery - Ostrava, Czech Republic
| | - Demet Sengul
- Giresun University, Faculty of Medicine, Department of Pathology - Giresun, Turkey
| | - Ilker Sengul
- Giresun University, Faculty of Medicine, Division of Endocrine Surgery - Giresun, Turkey
- Giresun University, Faculty of Medicine, Department of General Surgery - Giresun, Turkey
| | - Matúš Peteja
- Silesian Hospital in Opava, Department of Surgery - Opava, Czech Republic
- University of Ostrava, Faculty of Medicine, Department of Surgery - Ostrava, Czech Republic
| | - Lenka Přibylová
- VŠB-TUO Technical University of Ostrava, Faculty of Electrotechniques and Informatics, Department of Applied Mathematics - Ostrava, Czech Republic
| | - Wladyslav Gawel
- Silesian Hospital in Opava, Department of Surgery - Opava, Czech Republic
| | - Anton Pelikán
- University of Ostrava, Faculty of Medicine, Department of Surgery - Ostrava, Czech Republic
- Tomas Bata University in Zlin, Faculty of Humanities - Zlin, Czech Republic
| | - Hana Tomaskova
- University Hospital Olomouc, 1st Department of Surgery - Olomouc, Czech Republic
| | - Tomáš Malý
- University of Ostrava, Department of Epidemiology and Public Health - Ostrava, Czech Republic
| | - Vladimír Janout
- Palacký University in Olomouc, Faculty of Medicine, Centre for Science and Research - Olomouc, Czech Republic
| |
Collapse
|
9
|
De Nunzio V, Donghia R, Pesole PL, Coletta S, Calò N, Notarnicola M. Serum Cytokine and miRNA Levels Are Differently Expressed in Right- and Left-Sided Colon Cancer. J Clin Med 2023; 12:5986. [PMID: 37762927 PMCID: PMC10532301 DOI: 10.3390/jcm12185986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/21/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
The tumor location in colorectal cancer (right- or left-sided colon cancer) is a key factor in determining disease progression. Right- and left-sided colon tumors are different in their clinical and molecular characteristics. Dysregulation of serum levels of proinflammatory cytokines, such as Transforming Growth Factor β (TGF-β) and Tumor Necrosis Factor-α (TNF-α), and Peroxisome Proliferator Activated Receptor-γ (PPAR-γ), known to be a growth-limiting and differentiation-promoting factor, as well as changes in miRNAs expression, are the major signaling pathways involved in the pathogenesis of this neoplasia. In the serum from 60 colorectal cancer (CRC) patients, we compared the differences in the expression of the levels of TGF-β, TNF-α, and PPAR-γ and in the expression of the main human miRNAs between right and left CRC. A significant over-expression in the TGF-β and TNF-α levels was observed in the serum from right-sided colon cancer patients. For the PPAR-γ, the patients with CRC located on the right-side showed lower levels than those detected in the serum from left-sided CRC subjects. Furthermore, significant differences also existed in the expression of specific circulating miRNAs between right- and left-sided CRC. In particular, the right upregulated miRNAs were all involved in the cell growth and proliferation related pathways. These findings confirm that the analysis of circulating levels of TGF-β, TNF-α, and PPAR-γ, as well as the study of the specific miRNAs in the serum, are able to identify specific characteristics of CRC patients, useful for choosing a personalized treatment protocol.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Notarnicola
- National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (V.D.N.); (R.D.); (P.L.P.); (S.C.); (N.C.)
| |
Collapse
|
10
|
Li C, You R, Liu L, Li Y, Pu H, Lei M, Li Z, You D, Xiong Q, Zhang T. Perioperative changing patterns and longitudinal trajectories of CA242 with colorectal cancer prognosis: a retrospective longitudinal cohort study. Sci Bull (Beijing) 2023; 68:1875-1879. [PMID: 37532589 DOI: 10.1016/j.scib.2023.07.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/19/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Affiliation(s)
- Chunxia Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Institute for Medical Dataology, Shandong University, Jinan 250002, China
| | - Ruimin You
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming 650118, China
| | - Lizhu Liu
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming 650118, China
| | - Yanli Li
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming 650118, 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
| | - Zhenhui Li
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming 650118, China
| | - Dingyun You
- School of Biomedical Engineering Research, Kunming Medical University, Kunming 650500, China.
| | - Qiuxia Xiong
- Yunnan Key Laboratory of Laboratory Medicine, Kunming 650032, China; Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming 650032, China; Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Institute for Medical Dataology, Shandong University, Jinan 250002, China.
| |
Collapse
|
11
|
Wojtalewicz N, Vierbaum L, Kaufmann A, Schellenberg I, Holdenrieder S. Longitudinal Evaluation of AFP and CEA External Proficiency Testing Reveals Need for Method Harmonization. Diagnostics (Basel) 2023; 13:2019. [PMID: 37370914 DOI: 10.3390/diagnostics13122019] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
The glycoproteins alpha-fetoprotein (AFP) and carcinoembryonic antigen (CEA) have long been approved as biomarkers for diagnosing and monitoring tumors. International Reference Preparations (IRPs) have been around since 1975. Nevertheless, manufacturer-dependent differences have been reported, indicating a lack of harmonization. This paper analyzes data from 15 external quality assessment (EQA) surveys conducted worldwide between 2018 and 2022. The aim was to gain insight into the longitudinal development of manufacturer-dependent differences for CEA and AFP. In each survey, participating laboratories received two samples with different tumor marker levels. Inter- and intra-assay variability was analyzed and the mean 80% and 90% of the manufacturer collectives were compared to the evaluation criteria of the German Medical Association (RiliBÄK). The median EQA results for CEA revealed manufacturer-dependent differences between the highest and lowest collective of up to 100%; for AFP, the median differences mostly remained below 40%. The coefficients of variation were predominantly low for both markers. We concluded that the current assays for AFP and CEA detection are better harmonized than previously reported. The assays displayed a good robustness; however, a narrowing of the current assessment limits in EQA schemes could further enhance the quality of laboratory testing.
Collapse
Affiliation(s)
- Nathalie Wojtalewicz
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223 Duesseldorf, Germany
| | - Laura Vierbaum
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223 Duesseldorf, Germany
| | - Anne Kaufmann
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223 Duesseldorf, Germany
| | - Ingo Schellenberg
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223 Duesseldorf, Germany
- Institute of Bioanalytical Sciences (IBAS), Center of Life Sciences, Anhalt University of Applied Sciences, Strenzfelder Allee 28, 06406 Bernburg, Germany
| | - Stefan Holdenrieder
- INSTAND e.V., Society for Promoting Quality Assurance in Medical Laboratories, Ubierstr. 20, 40223 Duesseldorf, Germany
- Institute of Laboratory Medicine, Munich Biomarker Research Center, Deutsches Herzzentrum München, Technische Universität München, 80636 Munich, Germany
| |
Collapse
|
12
|
Kang Q, Guo X, Li T, Yang C, Han J, Jia L, Liu Y, Wang X, Zhang B, Li J, Wen HL, Li H, Li L. Identification of differentially expressed HERV-K(HML-2) loci in colorectal cancer. Front Microbiol 2023; 14:1192900. [PMID: 37342563 PMCID: PMC10277637 DOI: 10.3389/fmicb.2023.1192900] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/04/2023] [Indexed: 06/23/2023] Open
Abstract
Colorectal cancer is one of the malignant tumors with the highest mortality rate in the world. Survival rates vary significantly among patients at various stages of the disease. A biomarker capable of early diagnosis is required to facilitate the early detection and treatment of colorectal cancer. Human endogenous retroviruses (HERVs) are abnormally expressed in various diseases, including cancer, and have been involved in cancer development. Real-time quantitative PCR was used to detect the transcript levels of HERV-K(HML-2) gag, pol, and env in colorectal cancer to systematically investigate the connection between HERV-K(HML-2) and colorectal cancer. The results showed that HERV-K(HML-2) transcript expression was significantly higher than healthy controls and was consistent at the population and cell levels. We also used next-generation sequencing to identify and characterize HERV-K(HML-2) loci that were differentially expressed between colorectal cancer patients and healthy individuals. The analysis revealed that these loci were concentrated in immune response signaling pathways, implying that HERV-K impacts the tumor-associated immune response. Our results indicated that HERV-K might serve as a screening tumor marker and a target for tumor immunotherapy in colorectal cancer.
Collapse
Affiliation(s)
- Qian Kang
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Xin Guo
- Key Laboratory for the Prevention and Control of Infectious Diseases, Department of Microbiological Laboratory Technology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tianfu Li
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Caiqin Yang
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Jingwan Han
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Lei Jia
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yongjian Liu
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Xiaolin Wang
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Bohan Zhang
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Jingyun Li
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Hong-Ling Wen
- Key Laboratory for the Prevention and Control of Infectious Diseases, Department of Microbiological Laboratory Technology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hanping Li
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Lin Li
- State Key Laboratory of Pathogen and Biosecurity, Department of Virology, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Pan M, Xiao T, Xu L, Xie Y, Ge W. UTP18-mediated p21 mRNA instability drives adenoma-carcinoma progression in colorectal cancer. Cell Rep 2023; 42:112423. [PMID: 37086406 DOI: 10.1016/j.celrep.2023.112423] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 01/13/2023] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
Colorectal cancer (CRC) often develops slowly from adenoma, but the underlying mechanism remains unclear, hampering the prevention or treatment of colorectal adenoma-carcinoma progression. In this study, we use in-depth quantitative proteomics combined with survival analysis, revealing that the ribosome protein U3 small nucleolar RNA-associated protein 18 homolog (UTP18) is consistently upregulated in the progression of colorectal adenoma to carcinoma and is associated with adenoma recurrence, effective serodiagnosis, and poor prognosis of CRC. Furthermore, deSUMOylation induces the nucleocytoplasmic transport of UTP18, driving cell-cycle progression and tumorigenesis via mediation of the instability of p21 mRNA. In addition, the growth and ribosome biogenesis of adenoma organoids is found to be promoted by overexpression of UTP18. Thus, UTP18 contributes to multiple roles in adenogenesis and malignancy of CRC, suggesting that it could be a potential biomarker and drug target for colorectal adenoma and cancer.
Collapse
Affiliation(s)
- Meng Pan
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Tixian Xiao
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lai Xu
- Division of Colorectal Surgery, Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Yong Xie
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China; Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100853, China; National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China.
| | - Wei Ge
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
14
|
Srour SA, Akin S. Chimeric Antigen Receptor T-Cell Therapy for Solid Tumors: The Past and the Future. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2023; 6:19-30. [PMID: 36751657 PMCID: PMC9888521 DOI: 10.36401/jipo-22-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 06/18/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is the new standard treatment for various indications in patients with advanced hematologic malignancies. Despite the several preclinical and early phase clinical trials, the overall clinical experience has been disappointing when applying this innovative therapy in solid tumors. The failure of CAR T-cell therapy and its limited antitumor activity in solid tumors have been attributed to several mechanisms, including tumor antigen heterogeneity, the hostile tumor microenvironment and poor trafficking of CAR T cells into tumor sites, and the unacceptable toxicities in some settings, among others. However, remarkable improvements have been made in understanding many of these failure mechanisms for which several emerging novel approaches are being applied to overcome these challenges. In this review, after a brief historic background for immunotherapy in solid tumors, we highlight the recent developments achieved in CAR T-cell designs, summarize completed clinical trials, and discuss current challenges facing CAR T-cell therapy and the suggested strategies to overcome these barriers.
Collapse
Affiliation(s)
- Samer A. Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Serkan Akin
- Department of Medical Oncology, Hacettepe University Cancer Institute, Hacettepe University, Ankara, Turkey
| |
Collapse
|
15
|
Liu HN, Yao C, Wang XF, Zhang NP, Chen YJ, Pan D, Zhao GP, Shen XZ, Wu H, Liu TT. Diagnostic and economic value of carcinoembryonic antigen, carbohydrate antigen 19-9, and carbohydrate antigen 72-4 in gastrointestinal cancers. World J Gastroenterol 2023; 29:706-730. [PMID: 36742169 PMCID: PMC9896613 DOI: 10.3748/wjg.v29.i4.706] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/28/2022] [Accepted: 12/21/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The diagnostic and economic value of carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA19-9) and CA72-4 for gastrointestinal malignant tumors lacked evaluation in a larger scale.
AIM To reassess the diagnostic and economic value of the three tumor biomarkers.
METHODS A retrospective analysis of all 32857 subjects who underwent CEA, CA19-9, CA72-4, gastroscopy and colonoscopy from October 2006 to May 2018 was conducted. Then, we assessed the discrimination and clinical usefulness. Total cost, cost per capita and cost-effectiveness ratios were used to evaluate the economic value of two schemes (gastrointestinal endoscopy for all people without blood tests vs both gastroscopy and colonoscopy when blood tests were positive).
RESULTS The analysis of 32857 subjects showed that CEA was a qualified biomarker for colorectal cancer (CRC), while the diagnostic efficiencies of CA72-4 were catastrophic for all gastrointestinal cancers (GICs). Regarding early diagnosis, only CEA could be used for early CRC. The combination of biomarkers didn’t greatly increase the area under the curve. The economic indicators of CEA were superior to those of CA19-9, CA72-4 and any combination. At the threshold of 1.8 μg/L to 10.4 μg/L, all four indicators of CEA were lower than those in the scheme that conducted gas-trointestinal endoscopy only. Subgroup analysis implied that the health checkup of CEA for people above 65 years old was economically valuable.
CONCLUSION CEA had qualified diagnostic value for CRC and superior economic value for GICs, especially for elderly health checkup subjects. CA72-4 was not suitable as a diagnostic biomarker.
Collapse
Affiliation(s)
- Hai-Ning Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Can Yao
- Department of Gastroenterology and Hepatology, Minhang District Central Hospital, Fudan University, Shanghai 201199, China
| | - Xiao-Fan Wang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ning-Ping Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan-Jie Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Dong Pan
- Department of Internet Technology Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guo-Ping Zhao
- Chinese National Human Genome Center at Shanghai, Zhujiang Hospital, Central Lab, Institute of Plant Physiology and Ecology, Shanghai Institute for Biological Sciences, Shanghai 200032, China
| | - Xi-Zhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hao Wu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Tao-Tao Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Taguchi M, Bouchi R, Fukuda T, Ihana-Sugiyama N, Kodani N, Ohsugi M, Tanabe A, Ueki K, Kajio H. Clinical significance of tumor markers in patients with type 2 diabetes: a retrospective observational study. Diabetol Int 2023; 14:40-50. [PMID: 36636164 PMCID: PMC9829951 DOI: 10.1007/s13340-022-00594-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/24/2022] [Indexed: 01/16/2023]
Abstract
Aim To cross-sectionally and longitudinally investigate the association between tumor markers (Cancer embryonic antigen (CEA) Carbohydrate antigen 19-9 (CA19-9)) and malignancies in type 2 diabetes patients without evidence of malignancy. Materials and Methods The study included 707 patients admitted for the treatment of diabetes from 1 August 2010 to 1 September 2018. Serum CEA and CA19-9 levels were measured for screening of malignancies at admission. Abdominal ultrasonography, computed tomography, and endoscopy were performed for close examination. The percentage of patients diagnosed with malignancy was calculated, and among those without malignancy, the incidence of malignancies was examined after discharge. Results A total of 26 patients (3.7%) were newly diagnosed with malignancy during hospitalization. The optimal cut-off value of CEA and CA19-9 by receiver operating characteristic analysis was 5.0 ng/mL and 75 U/mL, and their positive predictive values (PPV) were 8.7% and 22.5%, respectively. The addition of CA19-9 to age, smoking status, body mass index, and glycated hemoglobin significantly improved classification performance for malignancy using net reclassification improvement (0.682, 95% CI 0.256-1.107) and integrated discrimination improvement (0.150, 95% CI 0.007-0.294). Among 681 patients without malignancies during hospitalization, 30 patients (4.4%) developed malignancies during an average follow-up of 3.9 years. CA19-9 (hazard ratio: 1.005, 95% CI: 1.003-1.008) was associated with the development of malignancies. Conclusions PPV of serum CEA and CA19-9 for detecting malignancy was high in type 2 diabetes patients with poor glycemic control. Measuring CA19-9 was found to be valuable to cross-sectionally and longitudinally detect malignancies. Supplementary Information The online version contains supplementary material available at 10.1007/s13340-022-00594-x.
Collapse
Affiliation(s)
- Maho Taguchi
- Department of Diabetes, Endocrinology and Metabolism, Center Hospital, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
| | - Ryotaro Bouchi
- Department of Diabetes, Endocrinology and Metabolism, Center Hospital, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
- Diabetes and Metabolism Information Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
| | - Tatsuya Fukuda
- Department of Endocrinology and Metabolism, Tokyo Metropolitan Health and Hospitals Corporation Ohkubo Hospital, 2-44-1 Kabuki-cho, Shinjuku-ku, Tokyo, 160-8488 Japan
| | - Noriko Ihana-Sugiyama
- Department of Diabetes, Endocrinology and Metabolism, Center Hospital, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
- Diabetes and Metabolism Information Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
| | - Noriko Kodani
- Department of Diabetes, Endocrinology and Metabolism, Center Hospital, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
| | - Mitsuru Ohsugi
- Department of Diabetes, Endocrinology and Metabolism, Center Hospital, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
- Diabetes and Metabolism Information Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
| | - Akiyo Tanabe
- Department of Diabetes, Endocrinology and Metabolism, Center Hospital, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
| | - Kohjiro Ueki
- Department of Diabetes, Endocrinology and Metabolism, Center Hospital, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
- Diabetes Research Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
| | - Hiroshi Kajio
- Department of Diabetes, Endocrinology and Metabolism, Center Hospital, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655 Japan
| |
Collapse
|
17
|
Lee HG, Lim SB, Lee JL, Kim CW, Yoon YS, Park IJ, Kim JC. Preoperative albumin-bilirubin score as a prognostic indicator in patients with stage III colon cancer. Sci Rep 2022; 12:14910. [PMID: 36050367 PMCID: PMC9437055 DOI: 10.1038/s41598-022-19329-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
The albumin-bilirubin (ALBI) score was created to assess the severity of liver dysfunction and to predict prognosis of hepatocellular carcinoma. Purpose of this study was to investigate the prognostic value of the ALBI score in patients with stage III colon cancer using propensity score matching (PSM) analysis. This study analyzed 510 patients with stage III colon cancer who had surgery between 2014 and 2015. The ALBI score was calculated as follows: (log10 bilirubin (μmol/L) [Formula: see text] 0.66) + (albumin (g/L) [Formula: see text] -0.0852), and the optimal cut-off value was determined using a receiver operating characteristic analysis and the Youden Index. According to the calculated cut-off value, patients were divided into two groups: Group A (ALBI ≤ - 2.54) and Group B (ALBI > - 2.54). The average ALBI score was - 2.68 (from - 3.39 to - 0.69). Group A had a significantly higher 5-year disease-free survival rate (85.5% vs 75.7%, p = 0.02), 5-year cancer-specific survival rate (93.7% vs 84.4%, p = 0.02), and 5-year overall survival rate (90.6% vs 77.4%, p = 0.01) than Group B. High ALBI scores were found to be an independent risk factor for both disease-free survival (HR 1.68, p = 0.048) and cancer-specific survival (HR 2.24, p = 0.028). The preoperative ALBI score was found to be a promising prognostic indicator for predicting recurrence and survival in patients with stage III colon cancer in this study. Because the ALBI score is simple and inexpensive to obtain, it has the potential to be a useful clinical marker for colon cancer patients.
Collapse
Affiliation(s)
- Hyun Gu Lee
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Seok-Byung Lim
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea.
| | - Jong Lyul Lee
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Chan Wook Kim
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Yong Sik Yoon
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - In Ja Park
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Jin Cheon Kim
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| |
Collapse
|
18
|
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
|
19
|
Sanz L, Ibáñez-Pérez R, Guerrero-Ochoa P, Lacadena J, Anel A. Antibody-Based Immunotoxins for Colorectal Cancer Therapy. Biomedicines 2021; 9:1729. [PMID: 34829955 PMCID: PMC8615520 DOI: 10.3390/biomedicines9111729] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 01/21/2023] Open
Abstract
Monoclonal antibodies (mAbs) are included among the treatment options for advanced colorectal cancer (CRC). However, while these mAbs effectively target cancer cells, they may have limited clinical activity. A strategy to improve their therapeutic potential is arming them with a toxic payload. Immunotoxins (ITX) combining the cell-killing ability of a toxin with the specificity of a mAb constitute a promising strategy for CRC therapy. However, several important challenges in optimizing ITX remain, including suboptimal pharmacokinetics and especially the immunogenicity of the toxin moiety. Nonetheless, ongoing research is working to solve these limitations and expand CRC patients' therapeutic armory. In this review, we provide a comprehensive overview of targets and toxins employed in the design of ITX for CRC and highlight a wide selection of ITX tested in CRC patients as well as preclinical candidates.
Collapse
Affiliation(s)
- Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute, Hospital Universitario Puerta de Hierro, 28222 Madrid, Spain
| | - Raquel Ibáñez-Pérez
- Apoptosis, Immunity and Cancer Group, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, 50009 Zaragoza, Spain; (R.I.-P.); (P.G.-O.)
| | - Patricia Guerrero-Ochoa
- Apoptosis, Immunity and Cancer Group, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, 50009 Zaragoza, Spain; (R.I.-P.); (P.G.-O.)
| | - Javier Lacadena
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University, 28040 Madrid, Spain
| | - Alberto Anel
- Apoptosis, Immunity and Cancer Group, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, 50009 Zaragoza, Spain; (R.I.-P.); (P.G.-O.)
| |
Collapse
|
20
|
Sefrioui D, Beaussire L, Gillibert A, Blanchard F, Toure E, Bazille C, Perdrix A, Ziegler F, Gangloff A, Hassine M, Elie C, Bignon AL, Parzy A, Gomez P, Thill C, Clatot F, Sabourin JC, Frebourg T, Benichou J, Bouhier-Leporrier K, Gallais MP, Sarafan-Vasseur N, Michel P, Di Fiore F. CEA, CA19-9, circulating DNA and circulating tumour cell kinetics in patients treated for metastatic colorectal cancer (mCRC). Br J Cancer 2021; 125:725-733. [PMID: 34112948 PMCID: PMC8405627 DOI: 10.1038/s41416-021-01431-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 04/17/2021] [Accepted: 04/28/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND We previously reported that CEA kinetics are a marker of progressive disease (PD) in metastatic colorectal cancer (mCRC). This study was specifically designed to confirm CEA kinetics for predicting PD and to evaluate CA19-9, cell-free DNA (cfDNA), circulating tumour DNA (ctDNA) and circulating tumour cell (CTC) kinetics. METHODS Patients starting a chemotherapy (CT) with pre-treatment CEA > 5 ng/mL and/or CA19.9 > 30 UI/mL were prospectively included. Samples were collected from baseline to cycle 4 for CEA and CA19-9 and at baseline and the sixth week for other markers. CEA kinetics were calculated from the first to the third or fourth CT cycle. RESULTS A total of 192 mCRC patients were included. CEA kinetics based on the previously identified >0.05 threshold was significantly associated with PD (p < 0.0001). By dichotomising by the median value, cfDNA, ctDNA and CA19-9 were associated with PD, PFS and OS in multivariate analysis. A circulating scoring system (CSS) combining CEA kinetics and baseline CA19-9 and cfDNA values classified patients based on high (n = 58) and low risk (n = 113) of PD and was independently associated with PD (ORa = 4.6, p < 0.0001), PFS (HRa = 2.07, p < 0.0001) and OS (HRa = 2.55, p < 0.0001). CONCLUSIONS CEA kinetics alone or combined with baseline CA19-9 and cfDNA are clinically relevant for predicting outcomes in mCRC. TRIAL REGISTRATION NUMBER NCT01212510.
Collapse
Affiliation(s)
- David Sefrioui
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Ludivine Beaussire
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - André Gillibert
- grid.41724.34Department of Biostatistics, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
| | - France Blanchard
- grid.41724.34Department of Pathology, Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Emmanuel Toure
- grid.41724.34Department of Pathology, Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Céline Bazille
- grid.411149.80000 0004 0472 0160Department of Pathology, Caen University Hospital, Caen, France
| | - Anne Perdrix
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Biopathology, Henri Becquerel Centre, Rouen, France
| | - Frédéric Ziegler
- grid.7429.80000000121866389Normandie Univ, UNIROUEN, INSERM U1073, Rouen University Hospital and General Biochemistry Laboratory, Institute of Clinical Biology, Rouen, France
| | - Alice Gangloff
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Mélanie Hassine
- Department of Hepatogastroenterology, Elbeuf Hospital, Elbeuf, France
| | - Caroline Elie
- Department of Hepatogastroenterology, Elbeuf Hospital, Elbeuf, France
| | - Anne-Laure Bignon
- grid.411149.80000 0004 0472 0160Department of Hepatogastroenterology, Caen University Hospital, Caen, France
| | - Aurélie Parzy
- Department of Hepatogastroenterology, Francois Baclesse Centre, Caen, France
| | - Philippe Gomez
- Department of Medical Oncology, Frédéric Joliot Centre, Rouen, France
| | - Caroline Thill
- grid.41724.34Department of Biostatistics, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
| | - Florian Clatot
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Medical Oncology, Henri Becquerel Centre, Rouen, France
| | - Jean-Christophe Sabourin
- grid.41724.34Department of Pathology, Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Thierry Frebourg
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Genetics, Rouen, France
| | - Jacques Benichou
- grid.41724.34Department of Biostatistics, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
| | - Karine Bouhier-Leporrier
- grid.411149.80000 0004 0472 0160Department of Hepatogastroenterology, Caen University Hospital, Caen, France
| | | | - Nasrin Sarafan-Vasseur
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Pierre Michel
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Frédéric Di Fiore
- grid.41724.34Department of Hepatogastroenterology and Department of Medical Oncology, Henri Becquerel Centre, Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| |
Collapse
|
21
|
Jiang X, Wang J, Wang M, Xuan M, Han S, Li C, Li M, Sun XF, Yu W, Zhao Z. ITGB4 as a novel serum diagnosis biomarker and potential therapeutic target for colorectal cancer. Cancer Med 2021; 10:6823-6834. [PMID: 34414684 PMCID: PMC8495272 DOI: 10.1002/cam4.4216] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/17/2022] Open
Abstract
Purpose To develop new and effective biomarkers for the diagnosis of colorectal cancer (CRC). Experimental design The serum expression of ITGB4 (49 CRC and 367 HC) was detected by enzyme‐linked immunosorbent assay (ELISA), and its diagnostic value was analyzed using the receiver operating characteristic (ROC) curve. The sensitivity and specificity of ITGB4 in CRC diagnosis were calculated through statistical analysis. The optimal clinical cutoff value was calculated using the Youden index, and diagnostic efficacy was analyzed in a larger serum sample (98 CRC and 1631 non‐CRC). The expression of ITGB4 was measured by CyTOF (cell experimental technology) at the single‐cell level, and characteristics were analyzed using viSNE and SPADE TREE. Results Serum ITGB4 and CEA levels were significantly higher in CRC patients than in HC and non‐CRC patients. The use of serum ITGB4 levels for the diagnosis of CRC has a high sensitivity (79%) but not high specificity when the clinical cutoff value was 0.70 ng/mL. However, the optimal cutoff value was 1.6 ng/mL with 86.2% specificity and 52.0% sensitivity, and the diagnostic efficacy was greatly improved with high specificity (82.0%) and sensitivity (71.4%) when combined with CEA. ITGB4 expression characteristics were measured and related to the expression of EpCAM, Ck8/18, and perforin at the single‐cell level. Single‐cell analysis showed that cell clusters with low expression of CK8/18 and ITGB4 were more sensitive to 5FU and radiotherapy (RT). Conclusions ITGB4 is an effective diagnostic serum biomarker and a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Xia Jiang
- Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China.,Department of General Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Jia Wang
- Department of Internal Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Mengyu Wang
- Department of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Mingda Xuan
- Department of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Shuangshuang Han
- Department of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Chao Li
- Department of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Meng Li
- Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China.,The First Department of Colorectal Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiao-Feng Sun
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Weifang Yu
- Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China.,Department of Endoscopy Center, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Zengren Zhao
- Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China.,Department of General Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| |
Collapse
|
22
|
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
|
23
|
Park J, Park JS, Huang CH, Jo A, Cook K, Wang R, Lin HY, Deun JV, Li H, Min J, Wang L, Yoon G, Carter BS, Balaj L, Choi GS, Castro CM, Weissleder R, Lee H. An integrated magneto-electrochemical device for the rapid profiling of tumour extracellular vesicles from blood plasma. Nat Biomed Eng 2021; 5:678-689. [PMID: 34183802 PMCID: PMC8437135 DOI: 10.1038/s41551-021-00752-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/18/2021] [Indexed: 02/02/2023]
Abstract
Assays for cancer diagnosis via the analysis of biomarkers on circulating extracellular vesicles (EVs) typically have lengthy sample workups, limited throughput or insufficient sensitivity, or do not use clinically validated biomarkers. Here we report the development and performance of a 96-well assay that integrates the enrichment of EVs by antibody-coated magnetic beads and the electrochemical detection, in less than one hour of total assay time, of EV-bound proteins after enzymatic amplification. By using the assay with a combination of antibodies for clinically relevant tumour biomarkers (EGFR, EpCAM, CD24 and GPA33) of colorectal cancer (CRC), we classified plasma samples from 102 patients with CRC and 40 non-CRC controls with accuracies of more than 96%, prospectively assessed a cohort of 90 patients, for whom the burden of tumour EVs was predictive of five-year disease-free survival, and longitudinally analysed plasma from 11 patients, for whom the EV burden declined after surgery and increased on relapse. Rapid assays for the detection of combinations of tumour biomarkers in plasma EVs may aid cancer detection and patient monitoring.
Collapse
Affiliation(s)
- Jongmin Park
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Chemistry, Kangwon National University, Chuncheon 24341, Korea
| | - Jun Seok Park
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Surgery, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Chen-Han Huang
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Biomedical Sciences and Engineering, National Central University, Taoyuan 32001, Taiwan
| | - Ala Jo
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kaitlyn Cook
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Rui Wang
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA,Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Hsing-Ying Lin
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jan Van Deun
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Huiyan Li
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jouha Min
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lan Wang
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ghilsuk Yoon
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Gyu-Seog Choi
- Department of Surgery, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Cesar M. Castro
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA,Correspondence and requests for materials should be addressed to ;
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Correspondence and requests for materials should be addressed to ;
| |
Collapse
|
24
|
Non-Invasive Biomarkers for Earlier Detection of Pancreatic Cancer-A Comprehensive Review. Cancers (Basel) 2021; 13:cancers13112722. [PMID: 34072842 PMCID: PMC8198035 DOI: 10.3390/cancers13112722] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC), which represents approximately 90% of all pancreatic cancers, is an extremely aggressive and lethal disease. It is considered a silent killer due to a largely asymptomatic course and late clinical presentation. Earlier detection of the disease would likely have a great impact on changing the currently poor survival figures for this malignancy. In this comprehensive review, we assessed over 4000 reports on non-invasive PDAC biomarkers in the last decade. Applying the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool, we selected and reviewed in more detail 49 relevant studies reporting on the most promising candidate biomarkers. In addition, we also highlight the present challenges and complexities of translating novel biomarkers into clinical use. Abstract Pancreatic ductal adenocarcinoma (PDAC) carries a deadly diagnosis, due in large part to delayed presentation when the disease is already at an advanced stage. CA19-9 is currently the most commonly utilized biomarker for PDAC; however, it lacks the necessary accuracy to detect precursor lesions or stage I PDAC. Novel biomarkers that could detect this malignancy with improved sensitivity (SN) and specificity (SP) would likely result in more curative resections and more effective therapeutic interventions, changing thus the present dismal survival figures. The aim of this study was to systematically and comprehensively review the scientific literature on non-invasive biomarkers in biofluids such as blood, urine and saliva that were attempting earlier PDAC detection. The search performed covered a period of 10 years (January 2010—August 2020). Data were extracted using keywords search in the three databases: MEDLINE, Web of Science and Embase. The Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool was applied for study selection based on establishing the risk of bias and applicability concerns in Patient Selection, Index test (biomarker assay) and Reference Standard (standard-of-care diagnostic test). Out of initially over 4000 published reports, 49 relevant studies were selected and reviewed in more detail. In addition, we discuss the present challenges and complexities in the path of translating the discovered biomarkers into the clinical setting. Our systematic review highlighted several promising biomarkers that could, either alone or in combination with CA19-9, potentially improve earlier detection of PDAC. Overall, reviewed biomarker studies should aim to improve methodological and reporting quality, and novel candidate biomarkers should be investigated further in order to demonstrate their clinical usefulness. However, challenges and complexities in the path of translating the discovered biomarkers from the research laboratory to the clinical setting remain and would have to be addressed before a more realistic breakthrough in earlier detection of PDAC is achieved.
Collapse
|
25
|
Mahmood RD, Shaw D, Descamps T, Zhou C, Morgan RD, Mullamitha S, Saunders M, Mescallado N, Backen A, Morris K, Little RA, Cheung S, Watson Y, O'Connor JPB, Jackson A, Parker GJM, Dive C, Jayson GC. Effect of oxaliplatin plus 5-fluorouracil or capecitabine on circulating and imaging biomarkers in patients with metastatic colorectal cancer: a prospective biomarker study. BMC Cancer 2021; 21:354. [PMID: 33794823 PMCID: PMC8017714 DOI: 10.1186/s12885-021-08097-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 03/24/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Patients with metastatic colorectal cancer are treated with cytotoxic chemotherapy supplemented by molecularly targeted therapies. There is a critical need to define biomarkers that can optimise the use of these therapies to maximise efficacy and avoid unnecessary toxicity. However, it is important to first define the changes in potential biomarkers following cytotoxic chemotherapy alone. This study reports the impact of standard cytotoxic chemotherapy across a range of circulating and imaging biomarkers. METHODS A single-centre, prospective, biomarker-driven study. Eligible patients included those diagnosed with colorectal cancer with liver metastases that were planned to receive first line oxaliplatin plus 5-fluorouracil or capecitabine. Patients underwent paired blood sampling and magnetic resonance imaging (MRI), and biomarkers were associated with progression-free survival (PFS) and overall survival (OS). RESULTS Twenty patients were recruited to the study. Data showed that chemotherapy significantly reduced the number of circulating tumour cells as well as the circulating concentrations of Ang1, Ang2, VEGF-A, VEGF-C and VEGF-D from pre-treatment to cycle 2 day 2. The changes in circulating concentrations were not associated with PFS or OS. On average, the MRI perfusion/permeability parameter, Ktrans, increased in response to cytotoxic chemotherapy from pre-treatment to cycle 2 day 2 and this increase was associated with worse OS (HR 1.099, 95%CI 1.01-1.20, p = 0.025). CONCLUSIONS In patients diagnosed with colorectal cancer with liver metastases, treatment with standard chemotherapy changes cell- and protein-based biomarkers, although these changes are not associated with survival outcomes. In contrast, the imaging biomarker, Ktrans, offers promise to direct molecularly targeted therapies such as anti-angiogenic agents.
Collapse
Affiliation(s)
- Reem D Mahmood
- Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK.
| | - Danielle Shaw
- The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, UK
| | - Tine Descamps
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Alderley Park, Macclesfield, UK
| | - Cong Zhou
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Alderley Park, Macclesfield, UK
| | - Robert D Morgan
- Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
- Division of Cancer Sciences, School of Medicine, University of Manchester, Manchester, UK
| | - Saifee Mullamitha
- Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Mark Saunders
- Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Nerissa Mescallado
- Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Alison Backen
- Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
- Division of Cancer Sciences, School of Medicine, University of Manchester, Manchester, UK
| | - Karen Morris
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Alderley Park, Macclesfield, UK
| | - Ross A Little
- Division of Cancer Sciences, School of Medicine, University of Manchester, Manchester, UK
| | - Susan Cheung
- Division of Cancer Sciences, School of Medicine, University of Manchester, Manchester, UK
| | - Yvonne Watson
- Division of Cancer Sciences, School of Medicine, University of Manchester, Manchester, UK
| | - James P B O'Connor
- Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
- Division of Cancer Sciences, School of Medicine, University of Manchester, Manchester, UK
| | - Alan Jackson
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, University of Manchester, Manchester, UK
| | - Geoff J M Parker
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, University of Manchester, Manchester, UK
- Bioxydyn Limited, Manchester, UK
- Department of Computer Science, Centre for Medical Image Computing, University College London, London, UK
| | - Caroline Dive
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Alderley Park, Macclesfield, UK
| | - Gordon C Jayson
- Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
- Division of Cancer Sciences, School of Medicine, University of Manchester, Manchester, UK
| |
Collapse
|
26
|
Cha SE, Kujawski M, J Yazaki P, Brown C, Shively JE. Tumor regression and immunity in combination therapy with anti-CEA chimeric antigen receptor T cells and anti-CEA-IL2 immunocytokine. Oncoimmunology 2021; 10:1899469. [PMID: 33796409 PMCID: PMC7993151 DOI: 10.1080/2162402x.2021.1899469] [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] [Indexed: 02/07/2023] Open
Abstract
Targeted immunotherapy of solid cancers with chimeric antigen receptor (CAR) T cells and immunocytokines are attractive options in that they both rely on the specificity of tumor-targeted antibodies. Since carcinoembryonic antigen (CEA) expression in both colon and breast cancers is correlated with poor prognosis, it was chosen as a model tumor target in immunocompetent CEA transgenic (CEATg) mice. A second-generation anti-CEA CAR derived from CEA-specific antibody T84.66 was used to treat murine MC38 colon or E0771 breast carcinomas transfected with CEA. Anti-CEA CAR vs. mock transduced T cells exhibited a CEA-specific cytotoxic and IFNγ dose response to both CEA transfected cell lines vs. their CEA-negative controls. Anti-CEA CAR vs. mock transduced T cells delayed the median survival of CEA transfected s.c. MC38 or orthotopic E0771 tumor-bearing CEATg mice by 2 days. With the addition of one-day prior cyclophosphamide (CY) lymphodepletion, anti-CEA CAR T cell treatment delayed the median survival of MC38/CEA and E0771/CEA tumor-bearing CEATg mice by ten and 3 days, respectively. Since CAR T cells require IL2 for survival and expansion, anti-CEA-IL2 immunocytokine (ICK) treatment was performed post CAR T cell therapy. Single ICK treatment 1 day after CY plus anti-CEA CAR T cell therapy in the MC38/CEA model, and two ICK treatments every 3 days after CY plus anti-CEA CAR T cell therapy in the E0771/CEA model were ineffective, while four ICK treatments every 3 days after CY plus anti-CEA CAR T cell therapy completely eradicated MC38/CEA tumor growth and induced tumor immunity when the mice were re-challenged with tumor. These studies show the therapeutic potential of anti-CEA CAR T cells combined with ICK to treat CEA-positive tumors. Abbreviations: CAR: Chimeric antigen receptor, CEA: Carcinoembryonic antigen, CEACAM5, ICK: Immunocytokine, CY: Cyclophosphamide, CEATg mouse: transgenic CEA mouse, TDLN: Tumor-draining lymph node
Collapse
Affiliation(s)
- Seung E Cha
- Department of Immunology and Theranostics, City of Hope, Duarte, USA.,Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, USA
| | - Maciej Kujawski
- Department of Immunology and Theranostics, City of Hope, Duarte, USA
| | - Paul J Yazaki
- Department of Immunology and Theranostics, City of Hope, Duarte, USA
| | - Christine Brown
- Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, USA.,Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, USA.,T Cell Therapeutics Research Laboratory, City of Hope, Duarte, USA
| | - John E Shively
- Department of Immunology and Theranostics, City of Hope, Duarte, USA.,Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, USA
| |
Collapse
|
27
|
Zhao C, Ma C, Li W, Song Y, Hong C, Qi Y. Differences in Performance of Immunosensors Constructed Based on CeO2-Simulating Auxiliary Enzymes. ACS Biomater Sci Eng 2021; 7:1058-1064. [DOI: 10.1021/acsbiomaterials.0c01680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Chulei Zhao
- School of Chemistry and Chemical Engineering, Key Laboratory for Green Processing of Chemical Engineering of Xinjiang Bingtuan, Shihezi University, No. 221, Beisi Road, Xinjiang Uygur Autonomous Region, Shihezi 832000, China
| | - Chaoyun Ma
- School of Chemistry and Chemical Engineering, Key Laboratory for Green Processing of Chemical Engineering of Xinjiang Bingtuan, Shihezi University, No. 221, Beisi Road, Xinjiang Uygur Autonomous Region, Shihezi 832000, China
| | - Wenjun Li
- School of Chemistry and Chemical Engineering, Key Laboratory for Green Processing of Chemical Engineering of Xinjiang Bingtuan, Shihezi University, No. 221, Beisi Road, Xinjiang Uygur Autonomous Region, Shihezi 832000, China
| | - Yiju Song
- School of Chemistry and Chemical Engineering, Key Laboratory for Green Processing of Chemical Engineering of Xinjiang Bingtuan, Shihezi University, No. 221, Beisi Road, Xinjiang Uygur Autonomous Region, Shihezi 832000, China
| | - Chenglin Hong
- School of Chemistry and Chemical Engineering, Key Laboratory for Green Processing of Chemical Engineering of Xinjiang Bingtuan, Shihezi University, No. 221, Beisi Road, Xinjiang Uygur Autonomous Region, Shihezi 832000, China
| | - Yu Qi
- School of Chemistry and Chemical Engineering, Key Laboratory for Green Processing of Chemical Engineering of Xinjiang Bingtuan, Shihezi University, No. 221, Beisi Road, Xinjiang Uygur Autonomous Region, Shihezi 832000, China
| |
Collapse
|
28
|
Lee T, Teng TZJ, Shelat VG. Carbohydrate antigen 19-9 - tumor marker: Past, present, and future. World J Gastrointest Surg 2020; 12:468-490. [PMID: 33437400 PMCID: PMC7769746 DOI: 10.4240/wjgs.v12.i12.468] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/06/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Carbohydrate antigen 19-9 (CA 19-9) is a cell surface glycoprotein complex most commonly associated with pancreatic ductal adenocarcinoma (PDAC). Koprowski first described it in 1979 using a mouse monoclonal antibody in a colorectal carcinoma cell line. Historically, it is one of the most commonly used tumor markers for diagnosing, managing, and prognosticating PDAC. Additionally, elevated CA 19-9 levels are used as an indication for surgery in suspected benign pancreatic conditions. Another common application of CA 19-9 in the biliary tract includes its use as an adjunct in diagnosing cholangiocarcinoma. However, its clinical value is not limited to the hepatopancreatobiliary system. The reality is that the advancing literature has broadened the clinical value of CA 19-9. The potential value of CA 19-9 in patients' workup extends its reach to gastrointestinal cancers - such as colorectal and oesophageal cancer - and further beyond the gastrointestinal tract - including urological, gynecological, pulmonary, and thyroid pathologies. Apart from its role in investigations, CA 19-9 presents a potential therapeutic target in PDAC and acute pancreatitis. In a bid to consolidate its broad utility, we appraised and reviewed the biomarker's current utility and limitations in investigations and management, while discussing the potential applications for CA 19-9 in the works for the future.
Collapse
Affiliation(s)
- Tsinrong Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Thomas Zheng Jie Teng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Vishal G Shelat
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Department of General Surgery, Tan Tock Seng Hospital, Singapore 308433, Singapore
| |
Collapse
|
29
|
Fan J, Liu Y, Cai X, Wang J, Guo R, Ji Y, Li C, Xu Y, Li X, Zhang C, Zhang R, Zhu J, Cai S. A Novel Prognostic Model Incorporating Carcinoembryonic Antigen in 3-Week or Longer Postoperative Period for Stage III Colon Cancer: A Multicenter Retrospective Study. Front Oncol 2020; 10:566784. [PMID: 33335852 PMCID: PMC7736239 DOI: 10.3389/fonc.2020.566784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/02/2020] [Indexed: 01/21/2023] Open
Abstract
Background The prognostic stratification of colon cancer using only the tumor-node-metastasis (TNM) stage has some limitations. We sought to increase the accuracy of stratifying patients with stage III colon cancer by constructing a prognostic model combining carcinoembryonic antigen (CEA) with TNM. Methods We retrospectively analyzed the data generated from stage III colon cancer patients who had early postoperative CEA measurement from 21 to 100 days after surgery from 2006 to 2017. CEA value was processed using restricted cubic splines (RCS) method. The prognostic model was developed using cox proportional hazards regression. Results The time later than 20 days after surgery was optimal for measuring CEA, which was determined by comparing the prognostic value for preoperative and postoperative CEA (N = 2,049), and by evaluating the relationship between the hazard ratio (HR) and postoperative CEA measuring time. Postoperative CEA, T stage and N stage were selected into the final model, and the mean integrated-AUC (iAUC) was 0.78 with 1,000 × bootstrap resampling, which was higher than the model using only T and N stages (TN model; mean iAUC, 0.66). The net reclassification improvement (NRI) was 15% when compared with TN model. Patients could be divided into high and low risk groups by the model, and 3-year disease-free survival (DFS) were 53.7% and 87.0%, respectively (HR, 4.30; 95% CI, 2.65 to 6.96; P < 0.001). Similar results were found in the validation set. Conclusions Stage III colon cancer could be stratified more accurately using the new prognostic model combining postoperative CEA with T and N stage.
Collapse
Affiliation(s)
- Jin Fan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yanlong Liu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Cai
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jingwen Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Rui Guo
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yuan Ji
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, United States
| | - Chao Li
- Department of Radiation Oncology, Huashan Hospital Fudan University, Shanghai, China
| | - Ye Xu
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.,Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xinxiang Li
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.,Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chundong Zhang
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.,Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Rui Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Ji Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Sanjun Cai
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.,Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
30
|
Sekiguchi M, Matsuda T. Limited usefulness of serum carcinoembryonic antigen and carbohydrate antigen 19-9 levels for gastrointestinal and whole-body cancer screening. Sci Rep 2020; 10:18202. [PMID: 33097814 PMCID: PMC7585432 DOI: 10.1038/s41598-020-75319-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/14/2020] [Indexed: 12/24/2022] Open
Abstract
The diagnostic performance of serum carcinoembryonic antigen (CEA) and carbohydrate antigen (CA) 19-9 levels for multiple-organ cancer screening has not been fully elucidated. However, they are widely used for real-world opportunistic screening of multiple-organ cancers. This study aimed to examine the diagnostic performance of these serum markers in multiple-organ cancer screening. Data from asymptomatic individuals subjected to opportunistic cancer screening were analyzed. The diagnostic performance of CEA and CA 19-9 was assessed for (A) upper/lower gastrointestinal cancers and (B) whole-body cancers (including both gastrointestinal and other organ cancers) using the results of upper/lower gastrointestinal endoscopy and whole-body imaging as reference. Data from 12,349 and 7616 screened individuals were used to assess the diagnostic performance of CEA and CA 19-9 for (A) and (B), respectively. For (A), the sensitivity and positive predictive value (PPV) of CEA (cut-off: 5 ng/mL) were 7.8% and 3.7%, respectively; those of CA19-9 (cut-off: 37 U/mL) were 7.4% and 2.7%, respectively. For (B), the sensitivity and PPV of CEA were 6.6% and 4.1%, respectively, and those of CA19-9 were 10.8% and 5.8%, respectively. Considering even multiple cancers, the sensitivity and PPV of CEA and CA 19-9 were low, thus confirming their limited usefulness in multiple-organ cancer screening.
Collapse
Affiliation(s)
- Masau Sekiguchi
- Cancer Screening Center, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan. .,Division of Screening Technology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan. .,Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan.
| | - Takahisa Matsuda
- Cancer Screening Center, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.,Division of Screening Technology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan.,Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
31
|
Argilés G, Tabernero J, Labianca R, Hochhauser D, Salazar R, Iveson T, Laurent-Puig P, Quirke P, Yoshino T, Taieb J, Martinelli E, Arnold D. Localised colon cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2020; 31:1291-1305. [PMID: 32702383 DOI: 10.1016/j.annonc.2020.06.022] [Citation(s) in RCA: 664] [Impact Index Per Article: 132.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- G Argilés
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Spain
| | - J Tabernero
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), UVic-UCC, IOB-Quiron, Barcelona, Spain
| | - R Labianca
- Department Oncology, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | | | - R Salazar
- Department of Medical Oncology, Catalan Institute of Oncology, Oncobell Program (IDIBELL), CIBERONC, Hospitalet de Llobregat, Barcelona, Spain
| | - T Iveson
- University Hospital Southampton, NHS Foundation Trust, Southampton, UK
| | - P Laurent-Puig
- Assitance Publique-Hôpitaux de Paris AP-HP Paris Centre, Paris, France; Paris Cancer Institute CARPEM, Centre de Recherche des Cordeliers, Paris Sorbonne University, Paris University, Paris, France; INSERM, CNRS, Paris, France
| | - P Quirke
- Pathology and Data Analytics, School of Medicine, University of Leeds, Leeds, UK
| | - T Yoshino
- National Cancer Centre Hospital East, Kashiwa, Japan
| | - J Taieb
- Assitance Publique-Hôpitaux de Paris AP-HP Paris Centre, Paris, France; Paris Cancer Institute CARPEM, Centre de Recherche des Cordeliers, Paris Sorbonne University, Paris University, Paris, France; INSERM, CNRS, Paris, France; Department of Gastroenterology and GI Oncology, Georges Pompidou European Hospital, Paris Descartes University, Paris, France
| | - E Martinelli
- Università degli Studi della Campania Luigi Vanvitelli, Department of Precision Medicine, Naples, Italy
| | - D Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Hamburg, Germany
| |
Collapse
|
32
|
Yu H, Pan R, Gao T, Wu D, Ying J, Duan S. FANCF hypomethylation is associated with colorectal cancer in Han Chinese. TURKISH JOURNAL OF GASTROENTEROLOGY 2020; 31:558-565. [PMID: 32915143 DOI: 10.5152/tjg.2020.19394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND/AIMS Fanconi anemia complement group F (FANCF) is known to be involved in DNA repair, and the overexpression of FANCF protein leads to cell proliferation and ultimately to cancer. The purpose of this study was to assess whether FANCF methylation was associated with colorectal cancer (CRC). MATERIALS AND METHODS A case-control experiment was conducted to study the association between FANCF methylation and CRC. We used quantitative methylation-specific PCR to measure the FANCF promoter methylation, and the percentage of methylation reference (PMR) to quantify the FANCF promoter methylation level. To investigate the effect of the selected FANCF fragment on gene expression regulation, we also performed a dual-luciferase reporter gene assay. RESULTS The results indicated that FANCF methylation in CRC tumor tissues was significantly lower than that in the nontumor tissues (median PMR: 44.86% vs. 65.77%, p=0.00001). Analysis of receiver-operating characteristic curves showed that FANCF hypomethylation had a diagnostic value for CRC (area under curve [AUC]: 0.670, sensitivity: 55.8%, specificity: 71.7%, p=0.00001). The dual-luciferase reporter assay showed that the FANCF fragment upregulated gene expression (fold change: 1.93, p=0.002). CONCLUSION Research demonstrates for the first time that FANCF hypomethylation is significantly associated with CRC risk. FANCF hypomethylation may ultimately increase the risk of CRC by upregulating the expression of FANCF.
Collapse
Affiliation(s)
- Hang Yu
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Ranran Pan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Tong Gao
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Dongping Wu
- Department of Medical Oncology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Zhejiang, China
| | - Jieer Ying
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Shiwei Duan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
33
|
Engel RM, Oliva K, Koulis C, Yap R, McMurrick PJ. Predictive factors of complete pathological response in patients with locally advanced rectal cancer. Int J Colorectal Dis 2020; 35:1759-1767. [PMID: 32474708 DOI: 10.1007/s00384-020-03633-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Patients with locally advanced rectal cancer who achieve pathologic complete response (pCR) following neoadjuvant therapy have better long-term outcomes and could be spared from the perioperative and long-term morbidity of rectal resection. The aim of this study was to identify factors that predict the ability to achieve pCR at completion of conventional neoadjuvant therapy, therefore determining their suitability for non-surgical management. METHODS A retrospective analysis was performed on data obtained from a prospectively maintained colorectal neoplasia database. Patients treated for biopsy-proven primary rectal adenocarcinoma between January 1, 2010, and February 28, 2018, who received neoadjuvant radiotherapy or chemoradiotherapy and had undergone surgical resection, were included in this study. Five-year oncologic outcome data was also obtained for 144 patients. Clinicopathological tumour characteristics and treatment regimens were analysed for correlation to clinical outcome. RESULTS Three hundred fifty-four patients met inclusion criteria for this study. We identified significant differences between patients achieving a pCR and those that did not for tumour type (adenocarcinoma vs. mucinous/signet ring; p = 0.008), pre-treatment serum CEA level (</≥ 2.5 μg/L; p = 0.003), neoadjuvant therapy type (short-course radiotherapy and long-course chemoradiotherapy; p = 0.008) and preoperative lymph node status (node-negative versus node-positive disease; p = 0.031). Additionally, this is the first report to our knowledge to identify a significant correlation with pCR and the degree of tumour fixity (mobile vs. fixed/tethered; p = 0.038). CONCLUSIONS This retrospective analysis identified factors that significantly impact a patients' ability to achieve a pCR, which may prove useful for prospectively selecting patients suitable for non-surgical management of disease.
Collapse
Affiliation(s)
- Rebekah M Engel
- Cabrini Monash University Department of Surgery, Cabrini Health, Malvern, VIC, 3144, Australia.
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.
- Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia.
| | - Karen Oliva
- Cabrini Monash University Department of Surgery, Cabrini Health, Malvern, VIC, 3144, Australia
| | - Christine Koulis
- Cabrini Monash University Department of Surgery, Cabrini Health, Malvern, VIC, 3144, Australia
| | - Raymond Yap
- Cabrini Monash University Department of Surgery, Cabrini Health, Malvern, VIC, 3144, Australia
| | - Paul J McMurrick
- Cabrini Monash University Department of Surgery, Cabrini Health, Malvern, VIC, 3144, Australia
| |
Collapse
|
34
|
Establishment and validation of a novel nomogram incorporating clinicopathological parameters into the TNM staging system to predict prognosis for stage II colorectal cancer. Cancer Cell Int 2020; 20:285. [PMID: 32655317 PMCID: PMC7339452 DOI: 10.1186/s12935-020-01382-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 06/26/2020] [Indexed: 12/18/2022] Open
Abstract
Background Survival outcomes are significantly different in stage II colorectal cancer (CRC) patients with diverse clinicopathological features. The objective of this study is to establish a credible prognostic nomogram incorporating easily obtained parameters for stage II CRC patients. Methods A total of 1708 stage II CRC patients seen at Fudan University Shanghai Cancer Center (FUSCC) from 2008 to 2013 were retrospectively analyzed in this study. Cases were randomly separated into a training set (n = 1084) and a validation set (n = 624). Univariate and multivariate Cox regression analyses were used to identify independent prognostic factors that were subsequently incorporated into a nomogram. The performance of the nomogram was evaluated by the predicted concordance index (C-index) and ROC curve to calculate the area under the curve (AUC). The clinical utility of the nomogram was evaluated using decision curve analysis (DCA). Results In univariate and multivariate analyses, eight parameters were correlated with disease-free survival (DFS), which were subsequently selected to generate a prognostic nomogram based on DFS. For DFS predictions, the C-index values of the nomogram were 0.842 (95% confidence interval (CI) 0.710–0.980), and 0.701 (95% CI 0.610–0.770) for the training and validation sets, respectively. The AUC values of the ROC curves for the nomogram to predicted 1, 3 and 5-year survival were 0.869, 0.858, and 0.777 (training group) and 0.673, 0.714, and 0.706 (validation group), respectively. The recurrence probability calibration curve showed good consistency between actual observations and nomogram-based predictions. DCA showed better clinical application value for the nomogram than the TNM staging system. Conclusion A novel nomogram was established and validated in a large population, and the nomogram is a simple-to-use tool for physicians to facilitate postoperative personalized prognostic evaluation and determine therapeutic strategies for stage II CRC patients.
Collapse
|
35
|
Crisafulli G, Mussolin B, Cassingena A, Montone M, Bartolini A, Barault L, Martinetti A, Morano F, Pietrantonio F, Sartore-Bianchi A, Siena S, Di Nicolantonio F, Marsoni S, Bardelli A, Siravegna G. Whole exome sequencing analysis of urine trans-renal tumour DNA in metastatic colorectal cancer patients. ESMO Open 2020; 4:S2059-7029(20)30089-2. [PMID: 32149725 PMCID: PMC7001107 DOI: 10.1136/esmoopen-2019-000572] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/19/2019] [Accepted: 09/21/2019] [Indexed: 12/22/2022] Open
Abstract
Background The analysis of circulating free tumour DNA (ctDNA) in blood, commonly referred as liquid biopsy, is being used to characterise patients with solid cancers. Tumour-specific genetic variants can also be present in DNA isolated from other body fluids, such as urine. Unlike blood, urine sampling is non-invasive, can be self-performed, and allows recurrent longitudinal monitoring. The features of tumour DNA that clears from the glomerular filtration barrier, named trans-renal tumour DNA (trtDNA), are largely unexplored. Patients and methods Specimens were collected from 24 patients with KRAS or BRAF mutant metastatic colorectal cancer (mCRC). Driver mutations were assessed by droplet digital PCR (ddPCR) in ctDNA from plasma and trtDNA from urine. Whole exome sequencing (WES) was performed in DNA isolated from tissue, plasma and urine. Results Out of the 24 CRC cases, only four had sufficient DNA to allow WES analyses in urine and plasma. We found that tumour alterations primarily reside in low molecular weight fragments (less than 112 bp). In patients whose trtDNA was more than 2.69% of the urine derived DNA, cancer-specific molecular alterations, mutational signatures and copy number profiles identified in urine DNA are comparable with those detected in plasma ctDNA. Conclusions With current technologies, WES analysis of trtDNA is feasible in a small fraction of mCRC patients. Tumour-related genetic information is mainly present in low molecular weight DNA fragments. Although the limited amounts of trtDNA poses analytical challenges, enrichment of low molecular weight DNAs and optimised computational tools can improve the detection of tumour-specific genetic information in urine.
Collapse
Affiliation(s)
- Giovanni Crisafulli
- University of Turin, Department of Oncology, Candiolo (TO), Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo TO, Italy
| | | | - Andrea Cassingena
- Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Monica Montone
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo TO, Italy
| | | | - Ludovic Barault
- University of Turin, Department of Oncology, Candiolo (TO), Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo TO, Italy
| | | | - Federica Morano
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | - Salvatore Siena
- Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Di Nicolantonio
- University of Turin, Department of Oncology, Candiolo (TO), Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo TO, Italy
| | - Silvia Marsoni
- IFOM - the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Alberto Bardelli
- University of Turin, Department of Oncology, Candiolo (TO), Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo TO, Italy
| | - Giulia Siravegna
- University of Turin, Department of Oncology, Candiolo (TO), Italy .,Candiolo Cancer Institute, FPO - IRCCS, Candiolo TO, Italy
| |
Collapse
|
36
|
Identification of salivary volatile organic compounds as potential markers of stomach and colorectal cancer: A pilot study. J Oral Biosci 2020; 62:212-221. [PMID: 32474113 DOI: 10.1016/j.job.2020.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The purpose of the pilot study was to determine the potential diagnostic capabilities for the analysis of oxygen-containing salivary volatile organic compounds (VOCs) in stomach and colorectal cancer. METHODS Saliva samples of 11 patients with stomach cancer, 18 patients with colorectal cancer, and 16 healthy volunteers were analyzed through capillary gas chromatography. The levels of lipid peroxidation products and catalase activity were determined in all samples. To assess saliva diagnostic potential, we constructed a Classification and Regression Tree (CART). RESULTS It was shown that the use of a combination of saliva VOCs (acetaldehyde, acetone, propanol-2, and ethanol) allowed classification into Cancer/Control groups with a sensitivity and specificity of 95.7 and 90.9%, respectively. To clarify the location of the tumor, it was necessary to add a methanol level; in this case, the sensitivity for detecting stomach and colorectal cancer was 80.0% and 92.3%, respectively, while the specificity in both cases was 100%. When the lipid peroxidation product content was added to the VOC indicators, they were selected as the main factors for constructing the decision tree. For classification into Cancer/Control groups, only the triene conjugate and Schiff base content in saliva was sufficient. The combination of VOCs in saliva and lipid peroxidation indices improved the sensitivity and specificity for classification to 100%. CONCLUSION Preliminary data were obtained on the sensitivity and specificity of the diagnosis of stomach and colorectal cancer, which confirmed the promise of further studies on saliva VOCs for the purpose of clinical laboratory diagnostics.
Collapse
|
37
|
Rigatti SJ, Stout R. Association of Carcinoembryonic Antigen with Mortality in an Insurance Applicant Population. J Insur Med 2020; 48:24-35. [PMID: 31747325 DOI: 10.17849/insm-48-1-24-35.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Objectives.- To quantify the mortality risks associated with elevated levels of carcinoembryonic antigen (CEA). Background.- Carcinoembryonic antigen is cell surface glycoprotein and has been associated with the presence of high grade or metastatic cancers of the colon as well as other malignant and non-malignant disease. Prior publications have demonstrated the utility of CEA levels in the determination of mortality risk in life insurance applicants. The aim of this paper is to further characterize this risk with a larger set of data containing additional person-years of follow-up, more outcomes, and additional variables potentially associated with occult malignancy. Methods.- By use of the Social Security Death Index, mortality was examined in 321,574 insurance applicants age 50 years and older, who submitted blood samples to Clinical Reference Laboratories for testing including CEA. Results were stratified by age group and by CEA level (<5 ng/mL, 5 to 9.9 ng/mL, 10+ ng/mL), though other thresholds were tested. Mortality comparisons were carried out using Cox models and tabular methods with the 2015 smoker-distinct Valuation Basic Tables as a comparator. Results.- Relative mortality is increased at CEA levels above 4.0 ng/mL in both smokers and non-smokers. This association is persistent in Cox models when albumin, BMI and cholesterol are included as covariates. The strongest association with mortality risk occurred in the first 3-4 durations. The 3-year cumulative mortality ratio when using the 2015 VBT as baseline was 6.51 when comparing the group with CEA levels of 10+ ng/mL, compared to those with levels below 5.0 ng/mL. Conclusion.- This study shows that CEA is strongly associated with the risk of early excess mortality in life insurance applicants, and this risk appears not to be mitigated by consideration of other markers thought to be associated with occult malignancy.
Collapse
Affiliation(s)
- Steven J Rigatti
- Rigatti - Founder, Rigatti Risk Analytics, LLC, Consultant Medical Director, Clinical Reference Laboratories, Lenexa KS. Stout - Chief Scientific Officer/Laboratory Director, Clinical Reference Laboratories, Lenexa KS
| | - Robert Stout
- Rigatti - Founder, Rigatti Risk Analytics, LLC, Consultant Medical Director, Clinical Reference Laboratories, Lenexa KS. Stout - Chief Scientific Officer/Laboratory Director, Clinical Reference Laboratories, Lenexa KS
| |
Collapse
|
38
|
Liu H, Ye D, Chen A, Tan D, Zhang W, Jiang W, Wang M, Zhang X. A pilot study of new promising non-coding RNA diagnostic biomarkers for early-stage colorectal cancers. Clin Chem Lab Med 2020; 57:1073-1083. [PMID: 30978169 DOI: 10.1515/cclm-2019-0052] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/12/2019] [Indexed: 12/11/2022]
Abstract
Background Diagnostic biomarkers for the detection of colorectal cancers (CRCs) are lacking. Recent studies have demonstrated that circulating long non-coding RNAs have the potential to serve as biomarkers for the detection of cancers. We analyzed the significance of lncRNAs 91H, PVT-1 and MEG3 in the detection of CRC. Methods We examined the expression levels of 13 candidate lncRNAs in the plasma of 18 CRC patients and 20 non-cancerous controls. Then, we validated our findings by determining the expression levels of six promising lncRNAs in CRC tissues and normal colorectal tissues. Finally, we evaluated the clinical relevance of lncRNAs 91H, PVT-1 and MEG3 in the plasma of 58 CRC patients and 56 non-cancerous controls. Results Our data revealed that the expression levels of lncRNAs 91H, PVT-1 and MEG3 were significantly higher in plasma samples from CRC patients than in those from non-cancerous controls. The combination of 91H, PVT-1 and MEG3 could discriminate CRC patients from non-cancerous controls with an area under the receiver-operating curve (AUC) of 0.877 at a cut-off value of 0.3816, with a sensitivity of 82.76% and 78.57% specificity. More importantly, the combination of lncRNAs shows more sensitivity in the detection of early-stage CRC than the combination of CEA and CA19-9, biomarkers currently used for CRC detection (p < 0.0001). Conclusions lncRNAs 91H, PVT-1 and MEG3 are promising diagnostic biomarkers for early-stage CRC.
Collapse
Affiliation(s)
- Hanshao Liu
- Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, P.R. China.,General Surgery Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Deji Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Aijun Chen
- General Surgery Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Dan Tan
- General Surgery Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Wenpeng Zhang
- General Surgery Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Wenxia Jiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Mingliang Wang
- General Surgery Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Xiaoren Zhang
- Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, P.R. China.,General Surgery Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| |
Collapse
|
39
|
Chen Y, Chang W, Ren L, Chen J, Tang W, Liu T, Jian M, Liu Y, Wei Y, Xu J. Comprehensive Evaluation of Relapse Risk (CERR) Score for Colorectal Liver Metastases: Development and Validation. Oncologist 2020; 25:e1031-e1041. [PMID: 32181531 DOI: 10.1634/theoncologist.2019-0797] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/04/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The calculation of the tumor burden score (TBS) is not perfect because the bilobar spread of colorectal liver metastasis (CRLM) is neglected. The identification of an ideal prognostic scoring system for CRLM remains controversial. MATERIALS AND METHODS Patients who underwent curative intent liver resection for CRLM from one medical center were enrolled in cohort 1 (787 patients) and cohort 2 (162 patients). Tumor relapse-free survival (RFS) was the main outcome. A Cox regression model was used to identify independent predictors of prognosis. The time-dependent area under the curve, calibration curve, and C-index were employed to validate the predictive ability of the survival model. RESULTS Modified TBS (mTBS) was established by a mathematical equation with parameters including CRLM size, CRLM number, and unilobar or bilobar metastasis. Five preoperative predictors of worse RFS were identified in cohort 1 and incorporated into the Comprehensive Evaluation of Relapse Risk (CERR) score: KRAS/NRAS/BRAF-mutated tumor (1 point); node-positive primary (1 point); extrahepatic disease (1 point); carcinoembryonic antigen level > 200 ng/mL or carbohydrate antigen 19-9 (CA19-9) >200 U/mL (1 point); and mTBS between 5 and 11 (1 point) or 12 and over (2 points). Patients in cohort 1 were stratified by their CERR score into risk groups: the high-risk group (CERR score 4 or more), the medium-risk group (CERR score 2-3), and the low-risk group (CERR score 0-1). Importantly, internal validation in cohort 1 and further validation in cohort 2 both showed the superior discriminatory capacity of the CERR score. CONCLUSION mTBS should be promoted. The CERR score is a powerful prognostic tool that can help determine optimal clinical management strategies. IMPLICATIONS FOR PRACTICE This work resulted in the successful modification of the tumor burden score and development of a comprehensive and practical prognostic scoring system-the Comprehensive Evaluation of Relapse Risk (CERR) score. The CERR score, with a better prognostic discriminatory ability, outperformed the Fong score. Perhaps more importantly, the CERR score is a powerful prognostic tool because it unified the most consistently reported prognostic factors. Therefore, the CERR score can assist doctors in determining optimal clinical management strategies.
Collapse
Affiliation(s)
- Yijiao Chen
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Wenju Chang
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, People's Republic of China
| | - Li Ren
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, People's Republic of China
| | - Jingwen Chen
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, People's Republic of China
| | - Wentao Tang
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, People's Republic of China
| | - Tianyu Liu
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Mi Jian
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yu Liu
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Ye Wei
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, People's Republic of China
| | - Jianmin Xu
- Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive, Shanghai, People's Republic of China
| |
Collapse
|
40
|
Liu Q, Luo D, Cai S, Li Q, Li X. Circulating basophil count as a prognostic marker of tumor aggressiveness and survival outcomes in colorectal cancer. Clin Transl Med 2020; 9:6. [PMID: 32037496 PMCID: PMC7008108 DOI: 10.1186/s40169-019-0255-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/26/2019] [Indexed: 12/31/2022] Open
Abstract
Background Accumulating evidence demonstrated immune/inflammation-related implications of basophils in affecting tissue microenvironment that surrounded a tumor, and this study aimed to elucidate the clinical value of serum basophil count level. Methods Between December 2007 and September 2013, 1029 patients diagnosed with stage I–III CRC in Fudan University Shanghai Cancer Center meeting the essential criteria were identified. The Kaplan–Meier method was used to construct the survival curves. Several Cox proportional hazard models were constructed to assess the prognostic factors. A simple predictor (CB classifier) was generated by combining serum basophil count and serum carcinoembryonic antigen (CEA) level which had long been accepted as the most important and reliable prognostic factor in CRC. Results The preoperative basophils count < 0.025*109/L was strongly associated with higher T stage, higher N stage, venous invasion, perineural invasion, elevated serum CEA level, and thus poor survival (P < 0.05). Moreover, multivariate Cox analysis showed that patients with low level of preoperative basophils count had an evidently poorer DFS [Hazard ratio (HR) = 2.197, 95% CI 1.868–2.585]. Conclusions As a common immune/inflammation-related biomarker available from the blood routine examination, low level of preoperative serum basophil count was associated with aggressive biology and indicated evidently poor survival. Preoperative serum basophil count would be a useful and simple marker for the management of CRC patients.
Collapse
Affiliation(s)
- Qi Liu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, #270 Dongan Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dakui Luo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, #270 Dongan Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, #270 Dongan Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, #270 Dongan Road, Xuhui District, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, #270 Dongan Road, Xuhui District, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
41
|
Mo S, Zhou Z, Dai W, Xiang W, Han L, Zhang L, Wang R, Cai S, Li Q, Cai G. Development and external validation of a predictive scoring system associated with metastasis of T1-2 colorectal tumors to lymph nodes. Clin Transl Med 2020; 10:275-287. [PMID: 32508061 PMCID: PMC7240869 DOI: 10.1002/ctm2.30] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND It is critical for determining the optimum therapeutic solutions for T1-2 colorectal cancer (CRC) to accurately predict lymph node metastasis (LNM) status. The purpose of the present study is to establish and verify a nomogram to predict LNM status in T1-2 CRCs. METHODS A total of 16 600 T1-2 CRC patients were enrolled and classified into the training, internal validation, and external validation cohorts. The independent predictive parameters were determined by univariate and multivariate analyses to develop a nomogram to predict the probability of LNM status. The calibration curve, the area under the receiver operating characteristic curve (AUROC), and decision curve analysis (DCA) were used to evaluate the performance of the nomogram, and an external verification cohort was to verify the applicability of the nomogram. RESULTS Seven independent predictors of LNM in T1-2 CRC were identified in the multivariable analysis, including age, tumor site, tumor grade, perineural invasion, preoperative carcinoembryonic antigen, clinical assessment of LNM, and T stage. A nomogram incorporating the seven predictors was constructed. The nomogram yielded good discrimination and calibration, with AUROCs of 0.72 (95% confidence interval [CI]: 0.70-0.75), 0.70 (95% CI: 0.67-0.74), and 0.74 (95% CI: 0.71-0.79) in the training, internal validation, and external validation cohorts, respectively. DCA showed that the predictive scoring system had high clinical application value. CONCLUSIONS We proposed a novel predictive model for LNM in T1-2 CRC patients to assist physicians in making treatment decisions. The nomogram is advantageous for tailoring therapy in T1-2 CRC patients.
Collapse
Affiliation(s)
- Shaobo Mo
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zheng Zhou
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Weixing Dai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wenqiang Xiang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Lingyu Han
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Long Zhang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Cancer InstituteFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
| | - Renjie Wang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Sanjun Cai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qingguo Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Guoxiang Cai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
42
|
Lin YH, Wu CH, Fu HC, Chen YJ, Chen YY, Ou YC, Lin H. Prognostic significance of elevated pretreatment serum levels of CEA and CA-125 in epithelial ovarian cancer. Cancer Biomark 2020; 28:285-292. [PMID: 32390605 DOI: 10.3233/cbm-201455] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Epithelial ovarian cancer is a highly lethal gynecological malignancy. Accurate and cost-effective predictive tools to estimate the prognosis of patients with epithelial ovarian cancer before treatment are currently lacking. OBJECTIVE The purpose of this study was to evaluate the prognostic significance of pretreatment serum levels of carcinoembryonic antigen (CEA) and carbohydrate antigen-125 (CA-125) in primary epithelial ovarian cancer. METHODS Between 2008 and 2016, 326 patients with a diagnosis of primary epithelial ovarian cancer were retrospectively reviewed. We attempted to identify an optimal cut-off value of CEA to predict survival using ROC curve analysis. Cox regression univariate and multivariate analyses were used to evaluate prognostic factors. RESULTS The optimal cut-off value of CEA was 2.6 ng/mL. In univariate and multivariate analyses, FIGO stage and pretreatment CA-125 and CEA levels significantly predicted progression-free and overall survival. The 5-year progression-free survival rate for patients with both a CEA level < 2.6 ng/mL and CA-125 level < 35 U/mL was 84%, compared to only 33% for the patients with higher levels of both markers (p< 0.001). The 5-year cancer specific survival rate was 94% in those with a CEA level < 2.6 ng/mL and CA-125 level < 35 U/mL, and only 39% for those with higher levels of both markers (p< 0.001). CONCLUSIONS In addition to traditional prognostic factors, a pretreatment serum CEA level ⩾ 2.6 ng/mL and CA-125 level ⩾ 35 U/mL were also independent prognostic factors for epithelial ovarian cancer. Patients with an elevated CEA and/or CA-125 level before treatment should be considered to be at high-risk of recurrence and death.
Collapse
Affiliation(s)
- Yu-Han Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chen-Hsuan Wu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Lin-Kou, Taiwan
| | - Hung-Chun Fu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Jen Chen
- Department of Obstetrics and Gynecology, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Yin-Yi Chen
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Che Ou
- Department of Obstetrics and Gynecology, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Hao Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
43
|
Mo S, Cai X, Zhou Z, Li Y, Hu X, Ma X, Zhang L, Cai S, Peng J. Nomograms for predicting specific distant metastatic sites and overall survival of colorectal cancer patients: A large population-based real-world study. Clin Transl Med 2020; 10:169-181. [PMID: 32508027 PMCID: PMC7240852 DOI: 10.1002/ctm2.20] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND This study aims to develop functional nomograms to predict specific distant metastatic sites and overall survival (OS) of colorectal cancer (CRC) patients. METHODS CRC case data were retrospectively recruited from a large population-based public dataset. Nomograms were developed to predict the probabilities of specific distant metastatic sites and OS of CRC patients. The performance of nomogram was evaluated with the concordance index (C-index), calibration curves, area under the curve (AUC), and decision curve analysis (DCA). RESULTS A total of 142 343 cases were included in the current study. On the basis of univariate and multivariate analyses, clinicopathological features were correlated with specific distant metastatic sites and survival outcomes and were used to establish nomograms. The nomograms showed excellent accuracy in predicting specific distant metastatic sites. The C-indexes for the prediction of liver, lung, bone, and brain metastases were 0.82 (95% confidence interval (CI), 0.81-0.83), 0.80 (95% CI, 0.78-0.81), 0.83 (95% CI, 0.79-0.86), and 0.73 (95% CI, 0.72-0.84), respectively. Then, a prognostic nomogram integrating clinicopathological features and specific distant metastatic sites was established to predict 1-, 3-, and 5-year OS of CRC, with AUCs of 0.764 (95% CI, 0.741-0.783), 0.762 (95% CI, 0.745-0.781), and 0.745 (95% CI, 0.730-0.761), respectively. DCA showed that the prognostic nomogram had a better clinical application value than current TNM staging system. CONCLUSIONS Based on clinicopathological features, original nomograms were constructed for clinicians to predict specific distant metastatic sites and OS of CRC patients. These models could help to support the postoperative personalized assessment.
Collapse
Affiliation(s)
- Shaobo Mo
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xin Cai
- Department of Radiation OncologyShanghai Proton and Heavy Ion CenterShanghaiChina
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation TherapyShanghaiChina
| | - Zheng Zhou
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yaqi Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xiang Hu
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xiaoji Ma
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Long Zhang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Cancer InstituteFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
| | - Sanjun Cai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Junjie Peng
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
44
|
ÜÇÜNCÜ MZ. Kolorektal Kanserlerin Tanı ve Prognostik Takibinde Eski ve Yeni Serum Biyobelirteçleri: Sistematik İnceleme ve Meta-Analiz. İSTANBUL GELIŞIM ÜNIVERSITESI SAĞLIK BILIMLERI DERGISI 2019. [DOI: 10.38079/igusabder.592956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
45
|
Zhou Z, Mo S, Dai W, Xiang W, Han L, Li Q, Wang R, Liu L, Zhang L, Cai S, Cai G. Prognostic nomograms for predicting cause-specific survival and overall survival of stage I-III colon cancer patients: a large population-based study. Cancer Cell Int 2019; 19:355. [PMID: 31889907 PMCID: PMC6935115 DOI: 10.1186/s12935-019-1079-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Background The purpose of this study was to build functional nomograms based on significant clinicopathological features to predict cause-specific survival (CSS) and overall survival (OS) in patients with stage I–III colon cancer. Methods Data on patients diagnosed with stage I–III colon cancer between 2010 and 2015 were downloaded from the Surveillance, Epidemiology, and End Results (SEER) database. Univariate and multivariate Cox analyses were used to identify independent prognostic factors, which were used to construct nomograms to predict the probabilities of CSS and OS. The performance of the nomogram was assessed by C-indexes, receiver operating characteristic (ROC) curves and calibration curves. Decision curve analysis (DCA) was used to compare clinical usage between the nomogram and the tumor–node–metastasis (TNM) staging system. Results Based on the univariate and multivariate analyses, features that correlated with survival outcomes were used to establish nomograms for CSS and OS prediction. The nomograms showed favorable sensitivity at predicting 1-, 3-, and 5-year CSS and OS, with a C-index of 0.78 (95% confidence interval (CI) 0.77–0.80) for CSS and 0.74 (95% CI 0.73–0.75) for OS. Calibration curves and ROC curves revealed excellent predictive accuracy. The clinically and statistically significant prognostic performance of the nomogram generated with the entire group of patients and risk scores was validated by a stratified analysis. DCA showed that the nomograms were more clinically useful than TNM stage. Conclusion Novel nomograms based on significant clinicopathological characteristics were developed and can be used as a tool for clinicians to predict CSS and OS in stage I–III colon cancer patients. These models could help facilitate a personalized postoperative evaluation.
Collapse
Affiliation(s)
- Zheng Zhou
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032 China
| | - Shaobo Mo
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032 China
| | - Weixing Dai
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032 China
| | - Wenqiang Xiang
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032 China
| | - Lingyu Han
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032 China
| | - Qingguo Li
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032 China
| | - Renjie Wang
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032 China
| | - Lu Liu
- 4School of Foreign Languages and Cultures, Chongqing University, Chongqing, 401331 China
| | - Long Zhang
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,Department of Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032 China
| | - Sanjun Cai
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032 China
| | - Guoxiang Cai
- 1Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032 China
| |
Collapse
|
46
|
Locke WJ, Guanzon D, Ma C, Liew YJ, Duesing KR, Fung KYC, Ross JP. DNA Methylation Cancer Biomarkers: Translation to the Clinic. Front Genet 2019; 10:1150. [PMID: 31803237 PMCID: PMC6870840 DOI: 10.3389/fgene.2019.01150] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/22/2019] [Indexed: 12/23/2022] Open
Abstract
Carcinogenesis is accompanied by widespread DNA methylation changes within the cell. These changes are characterized by a globally hypomethylated genome with focal hypermethylation of numerous 5’-cytosine-phosphate-guanine-3’ (CpG) islands, often spanning gene promoters and first exons. Many of these epigenetic changes occur early in tumorigenesis and are highly pervasive across a tumor type. This allows DNA methylation cancer biomarkers to be suitable for early detection and also to have utility across a range of areas relevant to cancer detection and treatment. Such tests are also simple in construction, as only one or a few loci need to be targeted for good test coverage. These properties make cancer-associated DNA methylation changes very attractive for development of cancer biomarker tests with substantive clinical utility. Across the patient journey from initial detection, to treatment and then monitoring, there are several points where DNA methylation assays can inform clinical practice. Assays on surgically removed tumor tissue are useful to determine indicators of treatment resistance, prognostication of outcome, or to molecularly characterize, classify, and determine the tissue of origin of a tumor. Cancer-associated DNA methylation changes can also be detected with accuracy in the cell-free DNA present in blood, stool, urine, and other biosamples. Such tests hold great promise for the development of simple, economical, and highly specific cancer detection tests suitable for population-wide screening, with several successfully translated examples already. The ability of circulating tumor DNA liquid biopsy assays to monitor cancer in situ also allows for the ability to monitor response to therapy, to detect minimal residual disease and as an early biomarker for cancer recurrence. This review will summarize existing DNA methylation cancer biomarkers used in clinical practice across the application domains above, discuss what makes a suitable DNA methylation cancer biomarker, and identify barriers to translation. We discuss technical factors such as the analytical performance and product-market fit, factors that contribute to successful downstream investment, including geography, and how this impacts intellectual property, regulatory hurdles, and the future of the marketplace and healthcare system.
Collapse
Affiliation(s)
- Warwick J Locke
- Molecular Diagnostics Solutions, CSIRO Health and Biosecurity, North Ryde, NSW, Australia.,Probing Biosystems Future Science Platform, CSIRO Health and Biosecurity, Canberra, ACT, Australia
| | - Dominic Guanzon
- Molecular Diagnostics Solutions, CSIRO Health and Biosecurity, North Ryde, NSW, Australia.,Probing Biosystems Future Science Platform, CSIRO Health and Biosecurity, Canberra, ACT, Australia
| | - Chenkai Ma
- Molecular Diagnostics Solutions, CSIRO Health and Biosecurity, North Ryde, NSW, Australia
| | - Yi Jin Liew
- Molecular Diagnostics Solutions, CSIRO Health and Biosecurity, North Ryde, NSW, Australia.,Probing Biosystems Future Science Platform, CSIRO Health and Biosecurity, Canberra, ACT, Australia
| | - Konsta R Duesing
- Molecular Diagnostics Solutions, CSIRO Health and Biosecurity, North Ryde, NSW, Australia
| | - Kim Y C Fung
- Molecular Diagnostics Solutions, CSIRO Health and Biosecurity, North Ryde, NSW, Australia.,Probing Biosystems Future Science Platform, CSIRO Health and Biosecurity, Canberra, ACT, Australia
| | - Jason P Ross
- Molecular Diagnostics Solutions, CSIRO Health and Biosecurity, North Ryde, NSW, Australia.,Probing Biosystems Future Science Platform, CSIRO Health and Biosecurity, Canberra, ACT, Australia
| |
Collapse
|
47
|
Lin J, Cai D, Li W, Yu T, Mao H, Jiang S, Xiao B. Plasma circular RNA panel acts as a novel diagnostic biomarker for colorectal cancer. Clin Biochem 2019; 74:60-68. [PMID: 31669510 DOI: 10.1016/j.clinbiochem.2019.10.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common cancers worldwide, and emerging lines of evidence have implicated circular RNAs (circRNAs), a novel class of endogenous noncoding RNAs, in CRC development. However, whether plasma circRNAs might be novel diagnostic biomarkers for CRC remains unclear. METHODS We investigated the plasma levels of selected circRNAs by quantitative real-time PCR (qRT-PCR). The presence of the candidate circRNAs was confirmed through RNase R assays, qRT-PCR and DNA sequencing, and their diagnostic value was evaluated using a receiver operating characteristic (ROC) curve. RESULTS The plasma levels of three circRNAs (circ-CCDC66, circ-ABCC1 and circ-STIL) were significantly decreased in CRC patients (n = 45) compared with healthy controls (n = 61). The ROC curve analysis showed that the area under the ROC curve (AUC) of the three-circRNA panel was 0.780, which is higher than that of traditional protein biomarkers, such as carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9). Combining the circRNA panel with CEA and CA19-9 might improve the ability to diagnose CRC (AUC = 0.855). In addition, the plasma circ-ABCC1 level was related to tumor growth and progression, and the plasma circ-CCDC66 and circ-ABCC1 levels were decreased in precursor lesions of CRC, including colon adenomas and adenomatous polyps. More importantly, circ-CCDC66 and circ-STIL were found to be useful for diagnosing early-stage CRC, and the three-circRNA panel improved the ability to diagnose CEA-negative and CA19-9-negative CRC. CONCLUSION Our study provides the first identification of a panel of three plasma circRNAs that could serve as a novel and independent diagnostic biomarker for CRC.
Collapse
Affiliation(s)
- Jie Lin
- Department of Clinical Laboratory, The 904th Hospital of The People's Liberation Army, Wuxi 214044, PR China.
| | - Dongping Cai
- Department of Clinical Laboratory, The 904th Hospital of The People's Liberation Army, Wuxi 214044, PR China
| | - Wei Li
- Department of Pharmacy, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Ting Yu
- Department of Clinical Laboratory, The 89th Hospital of The People's Liberation Army, Weifang 261000, PR China
| | - Hui Mao
- Department of Clinical Laboratory, The 904th Hospital of The People's Liberation Army, Wuxi 214044, PR China
| | - Shan Jiang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, PR China.
| | - Bin Xiao
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
48
|
Ye SB, Cheng YK, Zhang L, Wang XP, Wang L, Lan P. Prognostic value of estrogen receptor-α and progesterone receptor in curatively resected colorectal cancer: a retrospective analysis with independent validations. BMC Cancer 2019; 19:933. [PMID: 31590647 PMCID: PMC6781392 DOI: 10.1186/s12885-019-5918-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/10/2019] [Indexed: 12/13/2022] Open
Abstract
Background Prognostic assessment is crucial for optimal treatment. The aim of our study was to investigate the potential impact of estrogen receptor-α (ER-α) and progesterone receptor (PR) on the prognosis of colorectal cancer (CRC) patients who received curative resection. Methods Retrospective evaluation of two independent cohorts of CRC patients maintained prospectively in 2009–2010 (training set) (n = 148) and 2007–2009 (internal validation set) (n = 485). Furthermore, we used an external independent CRC cohort from The Cancer Genome Atlas (TCGA) (n = 511) for further validation. ER-α and PR expression as well as other potential prognostic factors were retrospectively evaluated in training set with respect to overall survival (OS), local relapse free survival (LRFS) and distant metastasis free survival (DMFS). The prognostic factors found in training set will be validated in two validation cohorts. Results On univariate analysis for the training set, OS, LRFS and DMFS were not associated with PR expression. While patients with ER-αexpression were found to have poor prognosis. In addition, multivariate analysis showed that ER-αexpression maintained significance with respect to OS (HR, 5.06; p = 0.002), LRFS (HR, 8.81; p = 0.002) and DMFS (HR, 8.07; p = 0.004). Similarly, ER-α expression showed prognostic significance with respect to OS with hazard ratios (HRs) of 1.572 (95% CI: 1.001–2.467, p = 0.049) and 1.624 (95% CI: 1.047–2.520, p = 0.031) for the internal and external validation cohort, respectively. Conclusion ER-α expression was a biomarker of poor prognosis and it might inform treatment decision for high risk CRC patients. However, PR expression was not associated with survival outcomes. Electronic supplementary material The online version of this article (10.1186/s12885-019-5918-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shu-Biao Ye
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yi-Kan Cheng
- Department of Radiation Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, People's Republic of China
| | - Lin Zhang
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,State Key Laboratory of Oncology in South China, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People's Republic of China
| | - Xue-Ping Wang
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,State Key Laboratory of Oncology in South China, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People's Republic of China
| | - Lei Wang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China. .,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
49
|
Chen Y, Zhang M, Min KA, Wang H, Shin MC, Li F, Yang VC, Huang Y. Improved Protein Toxin Delivery Based on ATTEMPTS Systems. Curr Drug Targets 2019; 19:380-392. [PMID: 28260497 DOI: 10.2174/1389450118666170302094758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 03/26/2016] [Accepted: 08/16/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Ribosome-inactivating proteins (RIPs) are wildly found in multiple species of plants, bacteria and fungi. As a special family of protein toxins, RIPs can inhibit protein synthesis and induce cell death via inactivating ribosome in eukaryotic cells. Thus, RIPs have been applied for anti-tumor therapy in the past two decades. However, because of poor cell permeability, nonselective mode of action for tumor cells, poor pharmacokinetic profiles and immunogenicity, their clinical application has been severely constrained. As an effort to overcome these obstacles, tumor-specific monoclonal antibodies (mAb) have been conjugated to RIPs (forming so called "immunotoxins") specifically to increase their cytotoxicity and provide tumor targeting. Nevertheless, immunotoxins yet have not fully resolved all the issues and critical challenges still remain, such as immunogenicity and inability to penetrate into the deep site of tumor. OBJECTIVE To overcome the constrain of immunotoxins, the novel cell-penetrating peptide (CPP)- modified ATTEMPTS systems based on combination of CPP-mediated penetration and antibodymediated tumor targeting, with triggerable drug release function, were developed to achieve effective and safe delivery of protein toxin. RESULTS The CPP-modified ATTEMPTS systems showed effective protamine-triggered CPP-toxin release and thus enhanced CPP-mediated cellular uptake and cytotoxicity. It also showed antibodymediated in vivo tumor targeting and significantly increased in vivo tumor growth suppression with limited systematic toxicity. CONCLUSION The CPP-modified ATTEMPTS systems were developed and demonstrated as a proof-ofconcept for CPP-based protein toxin delivery with triggerable antibody targeting to improve the druggability of protein toxin drugs. The systems showed the potential application of protein toxin clinical translation in anticancer treatment.
Collapse
Affiliation(s)
- Yingzhi Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Meng Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Kyoung Ah Min
- Inje University College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Gimhae, Gyeongnam, China
| | - Huiyuan Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Meong Cheol Shin
- Gyeongsang National University College of Pharmacy and Research Institute of Pharmaceutical Sciences, Jinju, Gyeongnam, Korea.,University of Michigan College of Pharmacy, Ann Arbor, MI, United States
| | - Feng Li
- Hampton University School of Pharmacy, Hampton, VA, United States
| | - Victor C Yang
- University of Michigan College of Pharmacy, Ann Arbor, MI, United States
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
50
|
Ibáñez-Pérez R, Guerrero-Ochoa P, Al-Wasaby S, Navarro R, Tapia-Galisteo A, De Miguel D, Gonzalo O, Conde B, Martínez-Lostao L, Hurtado-Guerrero R, Sanz L, Anel A. Anti-tumoral potential of a human granulysin-based, CEA-targeted cytolytic immunotoxin. Oncoimmunology 2019; 8:1641392. [PMID: 31646080 DOI: 10.1080/2162402x.2019.1641392] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
Granulysin is a protein present in the granules of human cytotoxic T lymphocytes (CTL) and natural killer (NK) cells, with cytolytic activity against microbes and tumors. Previous work demonstrated the therapeutic effect of intratumoral injection of recombinant granulysin using in vivo models of breast cancer and multiple myeloma. In the present work we have developed a granulysin gene fusion to the anti-carcinoembryonic antigen (CEA/CEACAM5) single chain Fv antibody fragment MFE23. Both granulysin and the granulysin-based immunotoxin were expressed in Pichia pastoris. The immunotoxin specifically recognized CEA, purified or expressed on the cell surface. Moreover, the bioactivity of the immunotoxin against several CEA+ cell lines was higher than that of granulysin alone. Granulysin and the immunotoxin were tested as a treatment in in vivo xenograft models in athymic mice. When injected intratumorally, both granulysin and the immunotoxin were able to inhibit tumor growth. Furthermore, systemic administration of the immunotoxin demonstrated a decrease in tumor growth in a CEA+ tumor-bearing mouse model, whereas granulysin did not exhibit a therapeutic effect. This is the first granulysin-based immunotoxin and the present work constitutes the proof of concept of its therapeutic potential.
Collapse
Affiliation(s)
- Raquel Ibáñez-Pérez
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Patricia Guerrero-Ochoa
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Sameer Al-Wasaby
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Rocío Navarro
- Molecular Immunology Unit, "Puerta de Hierro" University Hospital, Majadahonda, Madrid, Spain
| | - Antonio Tapia-Galisteo
- Molecular Immunology Unit, "Puerta de Hierro" University Hospital, Majadahonda, Madrid, Spain
| | - Diego De Miguel
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Oscar Gonzalo
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Blanca Conde
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| | - Luis Martínez-Lostao
- Immunology Department, "Lozano Blesa" University Clinical Hospital, Zaragoza, Spain
| | - Ramón Hurtado-Guerrero
- Biocomputation and Physics of Complex Systems Institute (BIFI), University of Zaragoza, Zaragoza, Spain.,ARAID Foundation, Zaragoza, Spain
| | - Laura Sanz
- Molecular Immunology Unit, "Puerta de Hierro" University Hospital, Majadahonda, Madrid, Spain
| | - Alberto Anel
- Apoptosis, Immunity and Cancer Group, University of Zaragoza/Aragón Health Research Institute (IIS-Aragón), Zaragoza, Spain
| |
Collapse
|