1
|
Qiao Y, Zhu J, Han T, Jiang X, Wang K, Chen R, Du Y, Li J, Sun L. Finding the minimum number of retrieved lymph nodes in node-negative colorectal cancer using Real-world Data and the SEER database. Int J Surg 2023; 109:4173-4184. [PMID: 37755374 PMCID: PMC10720778 DOI: 10.1097/js9.0000000000000746] [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: 04/19/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Current clinical guidelines recommend the removal of at least 12 lymph nodes (LNs) in resectable colorectal cancer (CRC). With advancements in lymphadenectomy technologies, the number of retrieved lymph nodes (rLNs) has markedly increased. This study aimed to investigate the lowest number of rLNs in node-negative patients. MATERIALS AND METHODS A total of 1103 N0 and 208 N1a stage patients were enrolled in our cohort, while 8503 N0 and 1276 N1a patients from the Surveillance, Epidemiology, and End Results CRC database were included. Propensity score matching and multivariate Cox regression analyses were performed to mitigate the influence of selection bias and control for potential confounding variables. RESULTS The median number of rLNs in N0 patients increased from 13.5 (interquartile range [IQR]: 9-18) in 2013 to 17 (IQR: 15-20) in 2019. The restrictive cubic spline illustrated a nonlinear relationship between rLNs and prognosis (nonlinearity, P =0.009), with a threshold ( N =16) influencing clinical outcomes. Patients at either N0 or N1a stage with sufficient rLNs (≥16) demonstrated superior prognoses to those with a limited rLNs (<16). After adjusting for clinical confounders, similar prognoses were observed in N0 limited and N1a adequate populations. Furthermore, Kaplan-Meier curves revealed that N0 limited patients who received chemotherapy exhibited better outcomes than those who did not. CONCLUSIONS Among patients with node-negative CRC, it is crucial to remove 16 or more LNs effectively. Fewer than 16 rLNs should be regarded as an independent risk factor, implying the need for adjuvant chemotherapy.
Collapse
Affiliation(s)
- Yihuan Qiao
- Department of Digestive Surgery, Honghui Hospital, Xi’an Jiaotong University
| | - Jun Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Air Force Medical University
- Department of General Surgery, The Southern Theater Air Force Hospital, Guangzhou, People’s Republic of China
| | - Tenghui Han
- Department of Neurology, Airborne Army Hospital, Wuhan
| | - Xunliang Jiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Air Force Medical University
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, Air Force Medical University, Shaanxi
| | - Ke Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Air Force Medical University
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, Air Force Medical University, Shaanxi
| | - Rujie Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Air Force Medical University
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, Air Force Medical University, Shaanxi
| | - Yongtao Du
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Air Force Medical University
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, Air Force Medical University, Shaanxi
| | - Jipeng Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Air Force Medical University
| | - Li Sun
- Department of Digestive Surgery, Honghui Hospital, Xi’an Jiaotong University
| |
Collapse
|
2
|
Ma Y, Gan J, Bai Y, Cao D, Jiao Y. Minimal residual disease in solid tumors: an overview. Front Med 2023; 17:649-674. [PMID: 37707677 DOI: 10.1007/s11684-023-1018-6] [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: 02/22/2023] [Accepted: 06/24/2023] [Indexed: 09/15/2023]
Abstract
Minimal residual disease (MRD) is termed as the small numbers of remnant tumor cells in a subset of patients with tumors. Liquid biopsy is increasingly used for the detection of MRD, illustrating the potential of MRD detection to provide more accurate management for cancer patients. As new techniques and algorithms have enhanced the performance of MRD detection, the approach is becoming more widely and routinely used to predict the prognosis and monitor the relapse of cancer patients. In fact, MRD detection has been shown to achieve better performance than imaging methods. On this basis, rigorous investigation of MRD detection as an integral method for guiding clinical treatment has made important advances. This review summarizes the development of MRD biomarkers, techniques, and strategies for the detection of cancer, and emphasizes the application of MRD detection in solid tumors, particularly for the guidance of clinical treatment.
Collapse
Affiliation(s)
- Yarui Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingbo Gan
- Genetron Health (Beijing) Co. Ltd., Beijing, 102206, China
| | - Yinlei Bai
- Genetron Health (Beijing) Co. Ltd., Beijing, 102206, China
| | - Dandan Cao
- Genetron Health (Beijing) Co. Ltd., Beijing, 102206, China
| | - Yuchen Jiao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
3
|
Wang M, Su J, Lu J, Yang Z, Wang J, Zhong Y, Zeng T, Shen K, Lin S. Efficacy and safety of HER2-targeted therapy in patients with colorectal cancer: What should we expect from a meta-analysis? Clin Res Hepatol Gastroenterol 2023; 47:102078. [PMID: 36627051 DOI: 10.1016/j.clinre.2023.102078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/14/2022] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND OBJECTIVE Human epidermal growth factor receptor 2 (HER2) is an effective therapeutic target for breast and stomach cancers. However, the application of HER2-targeted therapy in colorectal cancer (CRC) remains controversial. We sought to assess the efficacy and safety of HER2-targeted therapy in CRC by performing a meta-analysis of relevant studies. METHODS We searched PubMed, Embase, Cochrane Library, Web of Science, and the ClinicalTrials.gov database to retrieve relevant studies. STATA 16 was used for the statistical analysis. The objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and incidence of treatment‑related adverse events (TRAEs) were used as the outcome indicators analyzed by random- or fixed-effects models. RESULTS A total of 267 patients from nine studies were included in this meta-analysis. The overall ORR and DCR were 27.5% (95% CI 16.8% to 39.6%) and 68.9% (95% CI 55.4% to 81.0%), respectively. No significant heterogeneity was found in PFS among these studies and the overall median PFS was 4.35 months (95% CI 3.70 to 4.99). The overall incidence of all-grade and grade 3 or higher adverse events were 93.5% (95% CI 88.4% to 97.4%) and 16.8% (95% CI 4.8% to 33.3%). CONCLUSIONS HER2-targeted therapy was confirmed as a promising treatment for colorectal cancer, warranting further high-quality clinical randomized controlled trials to verify.
Collapse
Affiliation(s)
- Menglei Wang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingyang Su
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinhua Lu
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ze Yang
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jue Wang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yazhen Zhong
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Tianni Zeng
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Kezhan Shen
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Shengyou Lin
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
4
|
Okamura K, Nagayama S, Tate T, Chan HT, Kiyotani K, Nakamura Y. Lymphocytes in tumor-draining lymph nodes co-cultured with autologous tumor cells for adoptive cell therapy. J Transl Med 2022; 20:241. [PMID: 35606862 PMCID: PMC9125345 DOI: 10.1186/s12967-022-03444-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/16/2022] [Indexed: 11/28/2022] Open
Abstract
Background Tumor-draining lymph nodes (TDLNs) are primary sites, where anti-tumor lymphocytes are primed to tumor-specific antigens and play pivotal roles in immune responses against tumors. Although adoptive cell therapy (ACT) using lymphocytes isolated from TDLNs were reported, characterization of immune activity of lymphocytes in TDLNs to tumor cells was not comprehensively performed. Here, we demonstrate TDLNs to have very high potential as cell sources for immunotherapy. Methods Lymphocytes from TDLNs resected during surgical operation were cultured with autologous-tumor cells for 2 weeks and evaluated tumor-reactivity by IFNγ ELISPOT assay. We investigated the commonality of T cell receptor (TCR) clonotypes expanded by the co-culture with tumor cells with those of tumor infiltrating lymphocytes (TILs). Results We found that that TCR clonotypes of PD-1-expressing CD8+ T cells in lymph nodes commonly shared with those of TILs in primary tumors and lymphocytes having tumor-reactivity and TCR clonotypes shared with TILs could be induced from non-metastatic lymph nodes when they were co-cultured with autologous tumor cells. Conclusion Our results imply that tumor-reactive effector T cells were present even in pathologically non-metastatic lymph nodes and could be expanded in vitro in the presence of autologous tumor cells and possibly be applied for ACT. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03444-1.
Collapse
|
5
|
Amirifar L, Besanjideh M, Nasiri R, Shamloo A, Nasrollahi F, de Barros NR, Davoodi E, Erdem A, Mahmoodi M, Hosseini V, Montazerian H, Jahangiry J, Darabi MA, Haghniaz R, Dokmeci MR, Annabi N, Ahadian S, Khademhosseini A. Droplet-based microfluidics in biomedical applications. Biofabrication 2021; 14. [PMID: 34781274 DOI: 10.1088/1758-5090/ac39a9] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/15/2021] [Indexed: 11/11/2022]
Abstract
Droplet-based microfluidic systems have been employed to manipulate discrete fluid volumes with immiscible phases. Creating the fluid droplets at microscale has led to a paradigm shift in mixing, sorting, encapsulation, sensing, and designing high throughput devices for biomedical applications. Droplet microfluidics has opened many opportunities in microparticle synthesis, molecular detection, diagnostics, drug delivery, and cell biology. In the present review, we first introduce standard methods for droplet generation (i.e., passive and active methods) and discuss the latest examples of emulsification and particle synthesis approaches enabled by microfluidic platforms. Then, the applications of droplet-based microfluidics in different biomedical applications are detailed. Finally, a general overview of the latest trends along with the perspectives and future potentials in the field are provided.
Collapse
Affiliation(s)
- Leyla Amirifar
- Mechanical Engineering, Sharif University of Technology, Tehran, Iran, Tehran, 11365-11155, Iran (the Islamic Republic of)
| | - Mohsen Besanjideh
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Tehran, 11365-11155, Iran (the Islamic Republic of)
| | - Rohollah Nasiri
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Tehran, 11365-11155, Iran (the Islamic Republic of)
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Tehran, 11365-11155, Iran (the Islamic Republic of)
| | | | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, Los Angeles, 90024, UNITED STATES
| | - Elham Davoodi
- Bioengineering, University of California - Los Angeles, Los Angeles, Los Angeles, 90095, UNITED STATES
| | - Ahmet Erdem
- Bioengineering, University of California - Los Angeles, Los Angeles, Los Angeles, 90095, UNITED STATES
| | | | - Vahid Hosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, Los Angeles, 90024, UNITED STATES
| | - Hossein Montazerian
- Bioengineering, University of California - Los Angeles, Los Angeles, Los Angeles, 90095, UNITED STATES
| | - Jamileh Jahangiry
- University of California - Los Angeles, Los Angeles, Los Angeles, 90095, UNITED STATES
| | | | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, Los Angeles, 90024, UNITED STATES
| | - Mehmet R Dokmeci
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, Los Angeles, 90024, UNITED STATES
| | - Nasim Annabi
- Chemical Engineering, UCLA, Los Angeles, Los Angeles, California, 90095, UNITED STATES
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, Los Angeles, 90024, UNITED STATES
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, Los Angeles, 90024, UNITED STATES
| |
Collapse
|
6
|
Procaccio L, Bergamo F, Daniel F, Rasola C, Munari G, Biason P, Crucitta S, Barsotti G, Zanella G, Angerilli V, Magro C, Paccagnella S, Di Antonio V, Loupakis F, Danesi R, Zagonel V, Del Re M, Lonardi S, Fassan M. A Real-World Application of Liquid Biopsy in Metastatic Colorectal Cancer: The Poseidon Study. Cancers (Basel) 2021; 13:5128. [PMID: 34680277 PMCID: PMC8533756 DOI: 10.3390/cancers13205128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND First-line decision making is the key to the successful care of mCRC patients and RAS/BRAF status is crucial to select the best targeted agent. In hub centers, a relevant proportion of patients referred from small volume centers may not have standard tissue-based (STB) molecular results available at the time of the first visit (T0). Liquid biopsy (LB) may help circumvent these hurdles. METHODS A monoinstitutional prospective head-to-head comparison of LB versus (vs.) STB testing was performed in a real-world setting. Selection criteria included: mCRC diagnosis with unknown RAS/BRAF status at T0, tumoral tissue archived in external centers, no previous treatment with anti-EGFR. At T0, patients underwent plasma sampling for LB testing and procedure for tissue recovery. RAS/BRAF genotyping was carried out by droplet digital PCR on circulating-tumoral (ct) DNA. The primary endpoint was the comparison of time to LB (T1) vs. STB (T2) results using the Mann-Whitney U test. Secondary endpoints were the concordance between LB and STB defined as overall percent agreement and the accuracy of LB in terms of specificity, sensitivity, positive and negative predictive value. We also performed an exploratory analysis on urinary (u) ctDNA. RESULTS A total of 33 mCRC patients were included. Mean T1 and T2 was 7 and 22 days (d), respectively (p < 0.00001). T2 included a mean time for archival tissue recovery of 17 d. The overall percent agreement between LB and STB analysis was 83%. Compared to STB testing, LB specificity and sensitivity were 90% and 80%, respectively, with a positive predictive value of 94% and negative one of 69%. In detail, at STB and LB testing, RAS mutation was found in 45% and 42% of patients, respectively; BRAF mutation in 15%. LB results included one false positive and four false negative. False negative cases showed a significantly lower tumor burden at basal CT scan. Concordance between STB and uctDNA testing was 89%. CONCLUSIONS Faster turnaround time, high concordance and accuracy are three key points supporting the adoption of LB in routinary mCRC care, in particular when decision on first-line therapy is urgent and tissue recovery from external centers may require a long time. Results should be interpreted with caution in LB wild-type cases with low tumor burden.
Collapse
Affiliation(s)
- Letizia Procaccio
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, 35121 Padova, Italy
| | - Francesca Bergamo
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Francesca Daniel
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Cosimo Rasola
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, 35121 Padova, Italy
| | - Giada Munari
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padova, Italy
| | - Paola Biason
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Stefania Crucitta
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56121 Pisa, Italy; (S.C.); (R.D.); (M.D.R.)
| | - Giulia Barsotti
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, 35121 Padova, Italy
| | - Giulia Zanella
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Valentina Angerilli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
| | - Cristina Magro
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Silvia Paccagnella
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
| | - Veronica Di Antonio
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Fotios Loupakis
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56121 Pisa, Italy; (S.C.); (R.D.); (M.D.R.)
| | - Vittorina Zagonel
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56121 Pisa, Italy; (S.C.); (R.D.); (M.D.R.)
| | - Sara Lonardi
- Oncology Unit 3, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padova, Italy
| |
Collapse
|
7
|
Durán-Vinet B, Araya-Castro K, Calderón J, Vergara L, Weber H, Retamales J, Araya-Castro P, Leal-Rojas P. CRISPR/Cas13-Based Platforms for a Potential Next-Generation Diagnosis of Colorectal Cancer through Exosomes Micro-RNA Detection: A Review. Cancers (Basel) 2021; 13:4640. [PMID: 34572866 PMCID: PMC8466426 DOI: 10.3390/cancers13184640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer with the second highest mortality rate worldwide. CRC is a heterogenous disease with multiple risk factors associated, including obesity, smoking, and use of alcohol. Of total CRC cases, 60% are diagnosed in late stages, where survival can drop to about 10%. CRC screening programs are based primarily on colonoscopy, yet this approach is invasive and has low patient adherence. Therefore, there is a strong incentive for developing molecular-based methods that are minimally invasive and have higher patient adherence. Recent reports have highlighted the importance of extracellular vesicles (EVs), specifically exosomes, as intercellular communication vehicles with a broad cargo, including micro-RNAs (miRNAs). These have been syndicated as robust candidates for diagnosis, primarily for their known activities in cancer cells, including immunoevasion, tumor progression, and angiogenesis, whereas miRNAs are dysregulated by cancer cells and delivered by cancer-derived exosomes (CEx). Quantitative polymerase chain reaction (qPCR) has shown good results detecting specific cancer-derived exosome micro-RNAs (CEx-miRNAs) associated with CRC, but qPCR also has several challenges, including portability and sensitivity/specificity issues regarding experiment design and sample quality. CRISPR/Cas-based platforms have been presented as cost-effective, ultrasensitive, specific, and robust clinical detection tools in the presence of potential inhibitors and capable of delivering quantitative and qualitative real-time data for enhanced decision-making to healthcare teams. Thereby, CRISPR/Cas13-based technologies have become a potential strategy for early CRC diagnosis detecting CEx-miRNAs. Moreover, CRISPR/Cas13-based platforms' ease of use, scalability, and portability also showcase them as a potential point-of-care (POC) technology for CRC early diagnosis. This study presents two potential CRISPR/Cas13-based methodologies with a proposed panel consisting of four CEx-miRNAs, including miR-126, miR-1290, miR-23a, and miR-940, to streamline novel applications which may deliver a potential early diagnosis and prognosis of CRC.
Collapse
Affiliation(s)
- Benjamín Durán-Vinet
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (B.D.-V.); (K.A.-C.); (H.W.)
- Center of Excellence in Translational Medicine (CEMT), Biomedicine and Translational Research Laboratory, Universidad de La Frontera, Temuco 4780000, Chile;
| | - Karla Araya-Castro
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (B.D.-V.); (K.A.-C.); (H.W.)
- Innovation and Entrepreneurship Institute (iDEAUFRO), Universidad de La Frontera, Temuco 4780000, Chile
| | - Juan Calderón
- Center for Genetics and Genomics, School of Medicine, Institute of Science and Innovation in Medicine (ICIM), Clínica Alemana, Universidad del Desarrollo, Santiago 8320000, Chile;
| | - Luis Vergara
- Center of Excellence in Translational Medicine (CEMT), Biomedicine and Translational Research Laboratory, Universidad de La Frontera, Temuco 4780000, Chile;
- Doctoral Program in Cell and Applied Molecular Biology, Universidad de La Frontera, Temuco 4780000, Chile
| | - Helga Weber
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (B.D.-V.); (K.A.-C.); (H.W.)
- Center of Excellence in Translational Medicine (CEMT), Biomedicine and Translational Research Laboratory, Universidad de La Frontera, Temuco 4780000, Chile;
| | - Javier Retamales
- Chilean Cooperative Group for Oncologic Research (GOCCHI), Santiago 8320000, Chile;
| | - Paulina Araya-Castro
- School of Medicine, Clínica Alemana, Universidad del Desarrollo, Santiago 8320000, Chile;
| | - Pamela Leal-Rojas
- Scientific and Technological Bioresource Nucleus (BIOREN-UFRO), Universidad de La Frontera, Temuco 4780000, Chile; (B.D.-V.); (K.A.-C.); (H.W.)
- Center of Excellence in Translational Medicine (CEMT), Biomedicine and Translational Research Laboratory, Universidad de La Frontera, Temuco 4780000, Chile;
- Department of Agricultural Sciences and Natural Resources, Faculty of Agricultural and Forestry Science, Universidad de La Frontera, Temuco 4780000, Chile
| |
Collapse
|
8
|
Loupakis F, Sharma S, Derouazi M, Murgioni S, Biason P, Rizzato MD, Rasola C, Renner D, Shchegrova S, Koyen Malashevich A, Malhotra M, Sethi H, Zimmermann BG, Aleshin A, Moshkevich S, Billings PR, Sedgwick JD, Schirripa M, Munari G, Cillo U, Pilati P, Dei Tos AP, Zagonel V, Lonardi S, Fassan M. Detection of Molecular Residual Disease Using Personalized Circulating Tumor DNA Assay in Patients With Colorectal Cancer Undergoing Resection of Metastases. JCO Precis Oncol 2021; 5:PO.21.00101. [PMID: 34327297 PMCID: PMC8315303 DOI: 10.1200/po.21.00101] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/16/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
PURPOSE More than 50% of patients with stage IV colorectal cancer (metastatic colorectal cancer [mCRC]) relapse postresection. The efficacy of postoperative systemic treatment is limited in this setting. Thus, these patients would greatly benefit from the use of a reliable prognostic biomarker, such as circulating tumor DNA (ctDNA) to identify minimal or molecular residual disease (MRD). PATIENTS AND METHODS We analyzed a cohort of 112 patients with mCRC who had undergone metastatic resection with curative intent as part of the PREDATOR clinical trial. The study evaluated the prognostic value of ctDNA, correlating MRD status postsurgery with clinical outcomes by using a personalized and tumor-informed ctDNA assay (bespoke multiple PCR, next-generation sequencing assay). Postresection, systemic therapy was given to 39.2% of the patients at the discretion of the treating physician. RESULTS Postsurgical, MRD positivity was observed in 54.4% (61 of 112) of patients, of which 96.7% (59 of 61) progressed at the time of data cutoff (hazard ratio [HR]: 5.8; 95% CI, 3.5 to 9.7; P < .001). MRD-positive status was also associated with an inferior overall survival: HR: 16.0; 95% CI, 3.9 to 68.0; P < .001. At the time of analyses, 96% (49 of 51) of patients were alive in the MRD-negative arm compared with 52.4% (32 of 61) in the MRD-positive arm. Patients who did not receive systemic therapy and were MRD-negative in the combined ctDNA analysis at two time points had an overall survival of 100%. In the multivariate analysis, ctDNA-based MRD status was the most significant prognostic factor associated with disease-free survival (HR: 5.78; 95% CI, 3.34 to 10.0; P < .001). CONCLUSION This study confirms that in mCRC undergoing resection of metastases, postoperative MRD analysis is a strong prognostic biomarker. It holds promises for being implemented in clinical decision making, informing clinical trial design, and further translational research.
Collapse
Affiliation(s)
- Fotios Loupakis
- Oncology Unit 1, Department Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
| | | | - Madiha Derouazi
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT
- AMAL Therapeutics, Genève, Switzerland
| | - Sabina Murgioni
- Oncology Unit 1, Department Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
| | - Paola Biason
- Oncology Unit 1, Department Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
| | - Mario Domenico Rizzato
- Oncology Unit 1, Department Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
| | - Cosimo Rasola
- Oncology Unit 1, Department Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | | | | | | | | | | | | | | | | | | | - Jonathon D. Sedgwick
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT
| | - Marta Schirripa
- Oncology Unit 1, Department Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
| | - Giada Munari
- Oncology Unit 1, Department Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
| | - Umberto Cillo
- Hepatobiliary Surgery and Liver Transplant Unit, Department of Surgery, Oncology and Gastroenterology, University of Padua, Italy
| | - Pierluigi Pilati
- Unit of Surgical Oncology of the Digestive Tract, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy
| | - Angelo Paolo Dei Tos
- Unit of Surgical Pathology, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Vittorina Zagonel
- Oncology Unit 1, Department Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
| | - Sara Lonardi
- Oncology Unit 3, Department of Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Castelfranco Veneto, Veneto, Italy
- Early Phase Clinical Trial Unit, Department of Oncology, Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
| | - Matteo Fassan
- Unit of Surgical Pathology, Department of Medicine (DIMED), University of Padua, Padua, Italy
- Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy
| |
Collapse
|
9
|
Cazacu IM, Semaan A, Stephens B, Swartzlander DB, Guerrero PA, Singh BS, Lungulescu CV, Danciulescu MM, Cherciu Harbiyeli IF, Streata I, Popescu C, Saftoiu A, Roy-Chowdhuri S, Maitra A, Bhutani MS. Diagnostic value of digital droplet polymerase chain reaction and digital multiplexed detection of single-nucleotide variants in pancreatic cytology specimens collected by EUS-guided FNA. Gastrointest Endosc 2021; 93:1142-1151.e2. [PMID: 33058885 DOI: 10.1016/j.gie.2020.09.051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS EUS-guided FNA is recommended as a first-line procedure for the histopathologic diagnosis of pancreatic cancer. Molecular analysis of EUS-FNA samples might be used as an auxiliary tool to strengthen the diagnosis. The current study aimed to evaluate the diagnostic performances of K-ras testing using droplet digital polymerase chain reaction (ddPCR) and a novel single-nucleotide variant (SNV) assay performed on pancreatic EUS-FNA samples. METHODS EUS-FNA specimens from 31 patients with pancreatic masses (22 pancreatic ductal adenocarcinomas, 7 chronic pancreatitis, and 2 pancreatic neuroendocrine tumors) were included in the study. K-ras testing was initially performed by ddPCR. In addition, mutational status was evaluated using an SNV assay by NanoString technology, using digital enumeration of unique barcoded probes to detect 97 SNVs from 24 genes of clinical significance. RESULTS The overall specificity and sensitivity of cytologic examination were 100% and 63%, respectively. K-ras mutation testing was performed using ddPCR, and the sensitivity increased to 87% with specificity 90%. The SNV assay detected at least 1 variant in 90% of pancreatic ductal adenocarcinoma samples; the test was able to detect 2 K-ras codon 61 mutations in 2 cases of pancreatic ductal adenocarcinoma, which were missed by ddPCR. The overall diagnostic accuracy of the cytologic examination alone was 74%, and it increased to 91% when the results of both molecular tests were considered for the cases with negative and inconclusive results. CONCLUSIONS The current study illustrated that integration of K-ras analysis with cytologic evaluation, especially in inconclusive cases, can enhance the diagnostic accuracy of EUS-FNA for pancreatic lesions.
Collapse
Affiliation(s)
- Irina M Cazacu
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA; Research Center of Gastroenterology and Hepatology, Craiova, Romania
| | - Alexander Semaan
- Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Bret Stephens
- Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel B Swartzlander
- Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Paola A Guerrero
- Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Ben S Singh
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | - Ioana Streata
- Research Center of Gastroenterology and Hepatology, Craiova, Romania
| | - Carmen Popescu
- Research Center of Gastroenterology and Hepatology, Craiova, Romania
| | - Adrian Saftoiu
- Research Center of Gastroenterology and Hepatology, Craiova, Romania
| | - Sinchita Roy-Chowdhuri
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anirban Maitra
- Sheikh Ahmed Pancreatic Cancer Research Center, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Manoop S Bhutani
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
10
|
Cusenza VY, Bisagni A, Rinaldini M, Cattani C, Frazzi R. Copy Number Variation and Rearrangements Assessment in Cancer: Comparison of Droplet Digital PCR with the Current Approaches. Int J Mol Sci 2021; 22:ijms22094732. [PMID: 33946969 PMCID: PMC8124143 DOI: 10.3390/ijms22094732] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
The cytogenetic and molecular assessment of deletions, amplifications and rearrangements are key aspects in the diagnosis and therapy of cancer. Not only the initial evaluation and classification of the disease, but also the follow-up of the tumor rely on these laboratory approaches. The therapeutic choice can be guided by the results of the laboratory testing. Genetic deletions and/or amplifications directly affect the susceptibility or the resistance to specific therapies. In an era of personalized medicine, the correct and reliable molecular characterization of the disease, also during the therapeutic path, acquires a pivotal role. Molecular assays like multiplex ligation-dependent probe amplification and droplet digital PCR represent exceptional tools for a sensitive and reliable detection of genetic alterations and deserve a role in molecular oncology. In this manuscript we provide a technical comparison of these two approaches with the golden standard represented by fluorescence in situ hybridization. We also describe some relevant targets currently evaluated with these techniques in solid and hematologic tumors.
Collapse
Affiliation(s)
- Vincenza Ylenia Cusenza
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
| | - Alessandra Bisagni
- Pathology Unit, Azienda Unità Sanitaria Locale—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
| | - Monia Rinaldini
- Medical Genetics Unit, Azienda Unità Sanitaria Locale—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (M.R.); (C.C.)
| | - Chiara Cattani
- Medical Genetics Unit, Azienda Unità Sanitaria Locale—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (M.R.); (C.C.)
| | - Raffaele Frazzi
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
- Correspondence:
| |
Collapse
|
11
|
Strati A, Zavridou M, Economopoulou P, Gkolfinopoulos S, Psyrri A, Lianidou E. Development and Analytical Validation of a Reverse Transcription Droplet Digital PCR (RT-ddPCR) Assay for PD-L1 Transcripts in Circulating Tumor Cells. Clin Chem 2021; 67:642-652. [PMID: 33421061 DOI: 10.1093/clinchem/hvaa321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/09/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND PD-L1, an immune checkpoint protein, is an important biomarker for monitoring cancer patients during the administration of cancer immunotherapy. Droplet digital PCR (ddPCR), is a highly sensitive and accurate tool for the quantification of cancer biomarkers in liquid biopsy. We report the development and analytical validation of a novel duplex RT-ddPCR assay for the simultaneous quantification of PD-L1 and hypoxanthine phosphoribosyltransferase (HPRT) (used as reference gene) transcripts in circulating tumor cells (CTCs). METHODS RT-ddPCR experimental conditions were first optimized and the assay was analytically validated using synthetic standards and the BB49 and SCC47 cancer cell lines. The developed assay was further applied in 71 peripheral blood (PB) samples from head and neck squamous cell carcinoma (HNSCC) patients and 20 PB samples from healthy donors. PD-L1 and HPRT transcripts were quantified in cDNAs derived from CTCs isolated by a size-dependent microfluidic device. The developed RT-ddPCR assay was directly compared to RT-qPCR using 71 identical patient cDNA samples. RESULTS Analytical sensitivity was 0.64 copies/μL, while estimation of intra- and interassay variation revealed a high reproducibility (within-run CV%:4.7-23%; between-run CV%:13%). Using the developed RT-ddPCR assay 33/71(46.5%) HNSCC patients' samples were found positive for PD-L1 expression in CTCs, while by using RT-qPCR fewer samples (23/71, 32.4%) were positive (concordance: 55/71, 77.5%). CONCLUSIONS The developed RT-ddPCR assay for PD-L1 in CTCs is highly sensitive, specific, and reproducible; additionally, it offers improved diagnostic sensitivity over RT-qPCR. The clinical utility of the assay should be prospectively evaluated for the real-time monitoring of CTCs of cancer patients under immunotherapy.
Collapse
Affiliation(s)
- Areti Strati
- Department of Chemistry, Analysis of Circulating Tumor Cells Lab, National and Kapodistrian University of Athens, Athens, Greece
| | - Martha Zavridou
- Department of Chemistry, Analysis of Circulating Tumor Cells Lab, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota Economopoulou
- Oncology Unit, 2nd Department of Internal Medicine-Propaedeutic, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stavros Gkolfinopoulos
- Oncology Unit, 2nd Department of Internal Medicine-Propaedeutic, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Amanda Psyrri
- Oncology Unit, 2nd Department of Internal Medicine-Propaedeutic, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evi Lianidou
- Department of Chemistry, Analysis of Circulating Tumor Cells Lab, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Wu B, Chen S, Zhuang L, Zeng J. The expression level of COX7C associates with venous thromboembolism in colon cancer patients. Clin Exp Med 2020; 20:527-533. [PMID: 32653968 DOI: 10.1007/s10238-020-00644-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
Venous thromboembolism (VTE) is a common complication of colon cancer. In the present study, we aimed to explore the association of the oncogene COX7C to VTE in colon cancer patients. Samples from 580 patients were examined histologically for VTE and pathological characteristic of cancer. Gene mutation and expression analysis were performed using polymerase chain reaction-based assays to evaluate genes related to VTE, including COX7C. Univariate analysis between clinical pathological factors and VTE was conducted. Logistic regression analysis was performed for the prediction of VTE by pathological factors and gene expressions. Among patients investigated, a total of 56 patients had VTE. COX7C had a significant correlation with VTE (p < 0.001). Despite a correlation between tumor size, invasion depth of tumor, lymph node metastasis, lymph node metastasis, distant metastasis, lymphovascular invasion, histologic type and pathology type, Ki-67, and some other genes, to VTE (p > 0.05), only COX7C expression demonstrated significance in its ability to predict VTE. Here, we show that COX7C upregulation strongly correlates with VTE in colon cancer, which implicates its role as a biomarker and therapeutic target of VTE in colon cancer.
Collapse
Affiliation(s)
- Biyu Wu
- Department of Nursing, Quanzhou First Hospital Affiliated To Fujian Medical University, No.250 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Shurong Chen
- Department of Geriatric, Quanzhou First Hospital Affiliated To Fujian Medical University, No.250 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Lihong Zhuang
- Second Department of Gastrointestinal Surgery, Quanzhou First Hospital Affiliated To Fujian Medical University, No.250 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Jingyang Zeng
- Department of Anesthesiology, Quanzhou First Hospital Affiliated To Fujian Medical University, No.250 East Street, Licheng District, Quanzhou, 362000, Fujian, China.
| |
Collapse
|
13
|
Lee KH, Lee TH, Choi MK, Kwon IS, Bae GE, Yeo MK. Identification of a Clinical Cutoff Value for Multiplex KRAS G12/G13 Mutation Detection in Colorectal Adenocarcinoma Patients Using Digital Droplet PCR, and Comparison with Sanger Sequencing and PNA Clamping Assay. J Clin Med 2020; 9:jcm9072283. [PMID: 32708359 PMCID: PMC7409004 DOI: 10.3390/jcm9072283] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/08/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
KRAS (Kirsten rat sarcoma 2 viral oncogene homolog) is a major predictive marker for anti-epidermal growth factor receptor treatment, and determination of KRAS mutational status is crucial for successful management of colorectal adenocarcinoma. More standardized and accurate methods for testing KRAS mutation, which is vital for therapeutic decision-making, are required. Digital droplet polymerase chain reaction (ddPCR) is an advanced digital PCR technology developed to provide absolute quantitation of target DNA. In this study, we validated the clinical performance of ddPCR in determination of KRAS mutational status, and compared ddPCR results with those obtained by Sanger sequencing and peptide nucleic acid-clamping. Of 81 colorectal adenocarcinoma tissue samples, three repeated sets of KRASG12/G13 mutation were measured by ddPCR, yielding high consistency (ICC = 0.956). Receiver operating characteristic (ROC) curves were constructed to determine KRASG12/G13 mutational status based on mutant allele frequency generated by ddPCR. Using the best threshold cutoff (mutant allele frequency of 7.9%), ddPCR had superior diagnostic sensitivity (100%) and specificity (100%) relative to the two other techniques. Thus, ddPCR is effective for detecting the KRASG12/G13 mutation in colorectal adenocarcinoma tissue samples. By allowing definition of the optimal cutoff, ddPCR represents a potentially useful diagnostic tool that could improve diagnostic sensitivity and specificity.
Collapse
Affiliation(s)
- Kyung Ha Lee
- Department of Surgery, Chungnam National University Hospital, Daejeon 282, Korea;
| | - Tae Hee Lee
- The Biobank of Chungnam National University Hospital, Daejeon 282, Korea;
| | - Min Kyung Choi
- Department of Pathology, Chungnam National University School of Medicine, Daejeon 266, Korea; (M.K.C.); (G.E.B.)
| | - In Sun Kwon
- Clinical Trials Center of Chungnam National University Hospital, Daejeon 282, Korea;
| | - Go Eun Bae
- Department of Pathology, Chungnam National University School of Medicine, Daejeon 266, Korea; (M.K.C.); (G.E.B.)
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, Daejeon 266, Korea; (M.K.C.); (G.E.B.)
- Correspondence: ; Tel.: +82-42-280-7196; Fax: +82-42-580-8231
| |
Collapse
|
14
|
Xie J, Lin Y. Patient-derived xenograft models for personalized medicine in colorectal cancer. Clin Exp Med 2020; 20:167-172. [PMID: 32100151 DOI: 10.1007/s10238-020-00609-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022]
Abstract
Establishing superior preclinical models is critical for translational cancer research owing to the high failure rates of novel therapeutics in clinical studies. Even though cell line-derived xenograft models are easy to create, they have numerous limitations since these models do not represent the distinctive features of each cancer patient adequately. To circumvent the discrepancies between xenograft models and tumors, patient-derived xenograft (PDX) models have been developed. These models are established through the engraftment of tissue from a patient's tumor into an immune-deficient mouse, which preserves cell-cell interactions and tumor microenvironment. Since PDXs precisely replicate intratumoral heterogeneity, a range of chemotherapeutic agents can be tested on individual tumors. Colorectal cancer represents a unique case to demonstrate clinical perspectives revealed by PDX models since they surmount limitations of conventional ex vivo models. Even though PDX models have been associated with drawbacks with respect to prediction of clinical outcomes, they are currently the model of choice for preclinical investigations in colorectal cancer. In the current review, we provide an overview of the methodology and applications of PDX for colorectal cancer and discuss critical issues for the advancement of these models for preclinical research.
Collapse
Affiliation(s)
- Jun Xie
- General Surgery, Affiliated Hospital of Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Yan Lin
- Department of Gastroenterology, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
15
|
Li W, Xiao D, Wu H, Xu L. The epithelial cadherin -160C/A polymorphism is associated with decreased risk of colorectal cancer: a case-control study. Clin Exp Med 2020; 20:73-78. [PMID: 31625007 DOI: 10.1007/s10238-019-00586-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/11/2019] [Indexed: 01/03/2023]
Abstract
The epithelial cadherin (CDH1) is an important determinant of tumor progression. Previous studies have indicated that the CDH1 -160C/A polymorphism was associated with the risk of colorectal cancer (CRC). However, they yielded conflicting results. Thus, we conducted this case-control study to evaluate the association between the CDH1 -160C/A polymorphism and susceptibility to CRC in a Chinese population. We recruited 351 cases and 411 controls in this case-control study. The genotype of the CDH1 -160C/A polymorphism was performed by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOFMS). This study found that the CDH1 -160C/A polymorphism was associated with decreased risk of CRC in this Chinese Han population. Subgroup analyses showed that the CDH1 -160C/A polymorphism decreased the risk of CRC among the males and non-drinkers. In addition, a significantly decreased risk was observed in CRC patients with tumor size ≤ 5 cm, and AA genotype showed a protective role in the CRC patients with no lymph node metastasis. In conclusion, this study shows that CDH1 -160C/A polymorphism is associated with decreased risk of CRC in a Chinese Han population.
Collapse
Affiliation(s)
- Wenhuan Li
- Department of Gastrointestinal Surgery, The Affliated Wenling Hospital of Wenzhou Medical University, The First People's Hospital of Wenling, No. 333 Chuan'an South Road, Chengxi Street, Wenling, Taizhou, China
| | - Deshuang Xiao
- Department of Liver and Gallbladder Surgery, The Affliated Wenling Hospital of Wenzhou Medical University, The First People's Hospital of Wenling, No. 333 Chuan'an South Road, Chengxi Street, Wenling, Taizhou, China
| | - Huawen Wu
- Department of Gastrointestinal Surgery, The Affliated Wenling Hospital of Wenzhou Medical University, The First People's Hospital of Wenling, No. 333 Chuan'an South Road, Chengxi Street, Wenling, Taizhou, China
| | - Lewei Xu
- Department of Surgery, The Affliated Wenling Hospital of Wenzhou Medical University, The First People's Hospital of Wenling, No. 333 Chuan'an South Road, Chengxi Street, Wenling, Taizhou, China.
| |
Collapse
|
16
|
Li C, Zhan Y, Ma X, Fang H, Gai X. B7-H4 facilitates proliferation and metastasis of colorectal carcinoma cell through PI3K/Akt/mTOR signaling pathway. Clin Exp Med 2020; 20:79-86. [PMID: 31664539 DOI: 10.1007/s10238-019-00590-7] [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] [Received: 07/03/2019] [Accepted: 10/22/2019] [Indexed: 12/16/2022]
Abstract
B7-H4 is over-expressed in various tumors and may affect many aspects of cancer biology. Our previous studies have reported that the over-expressed B7-H4 in serum or tumor tissue of colorectal carcinoma (CRC) patients was closely related to CRC progression. However, B7-H4 in cell biological characteristics of CRC is not well studied. Here, we investigate the effect of the B7-H4 on cell proliferation, migration and its expression regulated by PI3K/Akt/mTOR signaling pathway in CRC. Firstly, pSilencer 4.1-B7-H4-shRNA vector was constructed and stable transfection was performed on HT-29 cells. Secondly, cell proliferation, cell cycle, cell apoptosis and cell migration were evaluated after B7-H4 silencing, and the expression of Bcl-2, caspase-3, MMP-2 and MMP-9 was also measured. Finally, the regulation of B7-H4 by PI3K/Akt/mTOR signaling pathway was measured followed by treatment with or without PI3K/Akt and mTOR inhibitor. The results showed that the viability of HT-29 cells was significantly decreased after B7-H4 silencing (P < 0.05). B7-H4 silencing significantly increased the apoptosis rate and caspase-3 protein expression while decreased Bcl-2 protein expression (P all < 0.05). B7-H4 silencing also significantly reduced the migration of HT-29 cells (P < 0.01) and the secretion of MMP-2 or MMP-9 (P all < 0.05). Following treatment with PI3K/Akt and mTOR inhibitor in HT-29 cells, the expression of B7-H4 was significantly downregulated compared with untreated group (P all < 0.05). Our results strongly suggest that B7-H4 may be involved in cell proliferation and migration by PI3K/Akt/mTOR signaling pathway. Therefore, blocking B7-H4 signaling might be a novel treatment strategy for CRC.
Collapse
Affiliation(s)
- Chun Li
- Department of Pathology, Beihua University Faculty of Medicine, No. 3999 Binjiang East Road, Jilin, 132013, Jilin, People's Republic of China
| | - Yudong Zhan
- Department of Pathology, Beihua University Faculty of Medicine, No. 3999 Binjiang East Road, Jilin, 132013, Jilin, People's Republic of China
- Department of Pathology, Jingmen No. 1 People's Hospital, Jingmen, 448000, Hubei, People's Republic of China
| | - Xuzhe Ma
- Department of Pathology, Beihua University Faculty of Medicine, No. 3999 Binjiang East Road, Jilin, 132013, Jilin, People's Republic of China
| | - Hui Fang
- Department of Pathology, Beihua University Faculty of Medicine, No. 3999 Binjiang East Road, Jilin, 132013, Jilin, People's Republic of China
| | - Xiaodong Gai
- Department of Pathology, Beihua University Faculty of Medicine, No. 3999 Binjiang East Road, Jilin, 132013, Jilin, People's Republic of China.
| |
Collapse
|
17
|
Snahnicanova Z, Kasubova I, Kalman M, Grendar M, Mikolajcik P, Gabonova E, Laca L, Caprnda M, Rodrigo L, Ciccocioppo R, Kruzliak P, Plank L, Lasabova Z. Genetic and epigenetic analysis of the beta-2-microglobulin gene in microsatellite instable colorectal cancer. Clin Exp Med 2020; 20:87-95. [PMID: 31853669 DOI: 10.1007/s10238-019-00601-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
One of the most common mechanisms of immune evasion in MSI colorectal cancers (CRCs) is loss of HLA class I expression due to mutations in B2M gene which can become a negative predictor for checkpoint blockade therapy. The aim of this study was the determination of prevalence of B2M somatic mutations in MSI CRC patients and relationship between B2M mutations and lymphocytes infiltration and other clinicopathological features as well as detection of methylation changes in B2M promoter region which can be another mechanism of immune escape. In our study, 37 MSI-H and 5 MSI-L patients were selected for screening of B2M mutational and methylation status. The characterization of patients was based on standard histopathological diagnosis and TNM classification; BRAF, KRAS mutations, tumor-infiltrating lymphocytes and peritumoral lymphoid reaction were also determined. MSI analysis was performed using fragment analysis. B2M mutations were identified by Sanger sequencing, and methylation of CpG islands in promoter region was detected by methylation-specific PCR. Heterozygous mutations in the B2M gene were detected in five MSI-H patients (13.5%), while the mutation c.45_48delTTCT was determined in four patients and mutation c.276delC was found in two patients. One of these five patients was compound heterozygote harboring both mutations. Methylation of the promoter region of the B2M gene was observed in one patient with MSI-H colorectal cancer. Detection of genetic and epigenetic changes in B2M gene could be important in personalized therapy for CRC patients as these changes may be one of the mechanisms of secondary resistance of MSI positive tumors to immunotherapy.
Collapse
Affiliation(s)
- Zuzana Snahnicanova
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Ivana Kasubova
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Michal Kalman
- Department of Pathological Anatomy, Jessenius Faculty of Medicine and University Hospital in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Marian Grendar
- Department of Bioinformatics, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Peter Mikolajcik
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Eva Gabonova
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Ludovit Laca
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria, Integrata Policlinico GB Rossi, University of Verona, Verona, Italy
| | - Peter Kruzliak
- Department of Internal Medicine, Brothers of Mercy Hospital, Polní 553/3, 63900, Brno, Czech Republic.
- 2nd Department of Surgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic.
| | - Lukas Plank
- Department of Pathological Anatomy, Jessenius Faculty of Medicine and University Hospital in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Zora Lasabova
- Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
- Department of Molecular Biology and Genomics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia.
| |
Collapse
|
18
|
Brunsell TH, Sveen A, Bjørnbeth BA, Røsok BI, Danielsen SA, Brudvik KW, Berg KCG, Johannessen B, Cengija V, Abildgaard A, Guren MG, Nesbakken A, Lothe RA. High Concordance and Negative Prognostic Impact of RAS/BRAF/PIK3CA Mutations in Multiple Resected Colorectal Liver Metastases. Clin Colorectal Cancer 2019; 19:e26-e47. [PMID: 31982351 DOI: 10.1016/j.clcc.2019.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 07/11/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prevalence and clinical implications of genetic heterogeneity in patients with multiple colorectal liver metastases remain largely unknown. In a prospective series of patients undergoing resection of colorectal liver metastases, the aim was to investigate the inter-metastatic and primary-to-metastatic heterogeneity of mutations in KRAS, NRAS, BRAF, and PIK3CA and their prognostic impact. PATIENTS AND METHODS We analyzed the mutation status among 372 liver metastases and 78 primary tumors from 106 patients by methods used in clinical routine testing, by Sanger sequencing, by next-generation sequencing (NGS), and/or by droplet digital polymerase chain reaction. The 3-year cancer-specific survival (CSS) was analyzed using the Kaplan-Meier method. RESULTS Although Sanger sequencing indicated inter-metastatic mutation heterogeneity in 14 of 97 patients (14%), almost all cases were refuted by high-sensitive NGS. Also, heterogeneity among metastatic deposits was concluded only for PIK3CA in 2 patients. Similarly, primary-to-metastatic heterogeneity was indicated in 8 of 78 patients (10%) using Sanger sequencing but for only 2 patients after NGS, showing the emergence of 1 KRAS and 1 PIK3CA mutation in the metastatic lesions. KRAS mutations were present in 53 of 106 patients (50%) and were associated with poorer 3-year CSS after liver resection (37% vs. 61% for KRAS wild-type; P = .004). Poor prognostic associations were found also for the combination of KRAS/NRAS/BRAF mutations compared with triple wild-type (P = .002). CONCLUSION Intra-patient mutation heterogeneity was virtually undetected, both between the primary tumor and the liver metastases and among the metastatic deposits. KRAS mutations separately, and KRAS/NRAS/BRAF mutations combined, were associated with poor patient survival after partial liver resection.
Collapse
Affiliation(s)
- Tuva Høst Brunsell
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anita Sveen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjørn Atle Bjørnbeth
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Bård I Røsok
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Stine Aske Danielsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | - Kristoffer Watten Brudvik
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Kaja C G Berg
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjarne Johannessen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vanja Cengija
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Radiology and Nuclear Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Andreas Abildgaard
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Radiology and Nuclear Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Marianne Grønlie Guren
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Oncology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Arild Nesbakken
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway; Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
19
|
Chen CL, Chen CK, Ho CL, Chi WM, Yeh CH, Hu SP, Friebe P, Palmer S, Huang CS. Clinical Evaluation of IntelliPlex™ KRAS G12/13 Mutation Kit for Detection of KRAS Mutations in Codon 12 and 13: A Novel Multiplex Approach. Mol Diagn Ther 2019; 23:645-656. [DOI: 10.1007/s40291-019-00418-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|