1
|
Mögele T, Höck M, Sommer F, Friedrich L, Sommer S, Schmutz M, Altenburger A, Messmann H, Anthuber M, Kröncke T, Stüben G, Trepel M, Märkl B, Dintner S, Claus R. Circulating Tumor DNA for Prediction of Complete Pathological Response to Neoadjuvant Radiochemotherapy in Locally Advanced Rectal Cancer (NEORECT Trial). Cancers (Basel) 2024; 16:4173. [PMID: 39766073 PMCID: PMC11674684 DOI: 10.3390/cancers16244173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Locally advanced rectal cancer is treated with neoadjuvant chemoradiotherapy (nCRT) followed by total mesorectal excision (TME). As this approach achieves complete pathologic remissions (pCR) in approximately 30% of patients, it raises the question of whether surgery is always necessary. Non-surgical strategies, such as "watch and wait" (W&W), have shown similarly promising outcomes. However, there is an unmet need for reliable biomarkers predicting pCR. Analysis of circulating tumor DNA (ctDNA) has shown potential for monitoring treatment response and detecting minimal residual disease. We hypothesized that monitoring ctDNA changes during nCRT might facilitate the identification of individuals who achieve pCR. METHODS In the prospective single-center NEORECT trial, the plasma of forty rectal cancer patients was collected before, during, and after nCRT and before TME. Informative somatic mutations were identified in tissue biopsies by NGS and subsequently used for ctDNA quantification by dPCR. RESULTS The results identified three distinct ctDNA patterns: increase, decrease, and absence. Remarkably, undetectable DNA was observed in good responders, while a tenfold ctDNA increase was associated with the emergence of new metastases. Despite these insights, ctDNA alone demonstrated low specificity, with no significant correlation to pCR or long-term prognosis. A multimodal approach incorporating routinely available clinical parameters remains inadequate for accurately predicting pCR prior to TME. CONCLUSIONS In conclusion, the NEORECT trial establishes the feasibility of ctDNA-based personalized monitoring for rectal cancer patients undergoing nCRT. However, the utility of ctDNA in enhancing pCR prediction for a W&W strategy warrants further investigation. Larger studies integrating multi-gene analyses and expanded clinical datasets are essential in the future.
Collapse
Affiliation(s)
- Tatiana Mögele
- Pathology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany; (T.M.); (B.M.)
- Bavarian Cancer Research Center (BZKF), Germany; (M.S.); (M.T.)
| | - Michael Höck
- Radiotherapy, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany (G.S.)
| | - Florian Sommer
- General and Visceral Surgery, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany; (F.S.); (M.A.)
| | - Lena Friedrich
- Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany; (L.F.); (T.K.)
| | - Sebastian Sommer
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
| | - Maximilian Schmutz
- Bavarian Cancer Research Center (BZKF), Germany; (M.S.); (M.T.)
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
| | - Amadeus Altenburger
- Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany; (L.F.); (T.K.)
| | - Helmut Messmann
- Gastroenterology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany;
| | - Matthias Anthuber
- General and Visceral Surgery, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany; (F.S.); (M.A.)
| | - Thomas Kröncke
- Diagnostic and Interventional Neuroradiology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany; (L.F.); (T.K.)
| | - Georg Stüben
- Radiotherapy, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany (G.S.)
| | - Martin Trepel
- Bavarian Cancer Research Center (BZKF), Germany; (M.S.); (M.T.)
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
- Comprehensive Cancer Center Augsburg (CCCA), 86156 Augsburg, Germany
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany; (T.M.); (B.M.)
| | - Sebastian Dintner
- Pathology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany; (T.M.); (B.M.)
| | - Rainer Claus
- Pathology, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany; (T.M.); (B.M.)
- Bavarian Cancer Research Center (BZKF), Germany; (M.S.); (M.T.)
- Comprehensive Cancer Center Augsburg (CCCA), 86156 Augsburg, Germany
| |
Collapse
|
2
|
Li M, Li Z, Wu X, Yubo Pan, Wang L, Xue J, Li T. Exploring the correlation between Tom1L1 and the efficacy of neoadjuvant chemotherapy for locally progressive mid-low rectal cancer. BMC Cancer 2024; 24:1413. [PMID: 39548458 PMCID: PMC11568529 DOI: 10.1186/s12885-024-13154-0] [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: 06/26/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
OBJECTIVE To investigate the specificity of Target of Myb1-Like1 (Tom1L1) expression in colorectal adenocarcinoma tissues and analyze the predictive value of Tom1L1 in the efficacy of neoadjuvant chemotherapy for patients with rectal adenocarcinoma. METHODS The cancerous tissues and paracancerous normal tissues of 102 patients diagnosed with colorectal adenocarcinoma without treatment were selected; quantitative polymerase chain reaction (qPCR), Western blot and immunohistochemistry (IHC) were adopted to validate the expression level of Tom1L1 in the two groups. Furthermore, 34 patients with locally progressive mid-low rectal adenocarcinoma, who were treated with neoadjuvant Xelox chemotherapy prior to the operation, IHC was adopted to detect the expression of Tom1L1 protein in patients before and after neoadjuvant chemotherapy and to analyze the relationship between the expression level of Tom1L1 and the sensitivity of neoadjuvant therapy. RESULTS The results of qPCR, Western blot and IHC showed that the expression of Tom1L1 in colorectal adenocarcinoma tissues was significantly higher than that in paracancerous normal tissues, with a statistically significant difference (P < 0.01); Neoadjuvant chemotherapy was significantly more effective in patients with low expression of Tom1L1 protein than in those with high expression of Tom1L1 protein, with a statistically significant difference (P < 0.05). CONCLUSIONS Tom1L1 is highly expressed in colorectal adenocarcinoma tissues; neoadjuvant Xelox chemotherapy can have an impact on Tom1L1 expression in progressive rectal cancer; patients with locally progressive mid-low rectal adenocarcinoma who have low Tom1L1 expression are more sensitive to neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Meng Li
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, 12 Changqing Rd, Zhangjiakou, 075000, China
- Graduate School, Hebei Medical University, Shijiazhuang, 050011, China
| | - Zeyu Li
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Xueliang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, 12 Changqing Rd, Zhangjiakou, 075000, China.
- Insistute of Oncology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China.
| | - Yubo Pan
- Department of Orthopedics, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China
| | - Likun Wang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China.
| | - Jun Xue
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, 12 Changqing Rd, Zhangjiakou, 075000, China.
- Insistute of Oncology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
3
|
Brzozowa-Zasada M, Piecuch A, Michalski M, Stęplewska K, Matysiak N, Kucharzewski M. Immunohistochemical Expression of Upregulated Gene 4 Protein Expression (URG4/URGCP) and Its Association with 5-Year Survival in Patients with Colon Adenocarcinoma. J Clin Med 2023; 12:5477. [PMID: 37685545 PMCID: PMC10488385 DOI: 10.3390/jcm12175477] [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: 06/14/2023] [Revised: 07/26/2023] [Accepted: 08/08/2023] [Indexed: 09/10/2023] Open
Abstract
(1) Background: Colorectal cancer (CRC) is the third most common cancer in terms of incidence and mortality. Approximately 90% of all colorectal cancer cases are adenocarcinomas, originating from epithelial cells of the colorectal mucosa. Upregulated gene 4 (URG4) is an oncogene involved in cancer development. The aim of the study was to assess the immunohistochemical expression of URG4 protein expression in Polish patients with colon adenocarcinoma who were not treated with any therapy before radical surgery. (2) Methods: The study used colon tissue samples taken from people with a confirmed diagnosis of colorectal adenocarcinoma after a thorough histopathological examination. The associations between the immunohistochemical expression of URG4 and clinical parameters were analyzed by the Chi2 test or Chi2Yatesa test. The study conducted an analysis of the correlation between the expression of URG4 and the five-year survival rate of patients through the application of the Kaplan-Meier analysis and the log-rank statistical test. The intracellular localization of URG4 was identified through the utilization of transmission electron microscopy (TEM) methodology. (3) Results: In univariate Cox regression analyses, immuno-histochemical expression of URG4, grade of histological differentiation, depth of invasion, angioinvasion, PCNA expression, stage of disease and lymph node involvement were found to be significant prognostic factors. Within our patient cohort, it was observed that the degree of tumour differentiation and URG4 expression were found to be distinct prognostic factors in regard to the 5-year survival rates of those with colon adenocarcinoma. (4) Conclusions: High immunohistochemical expression of URG4 correlates with poor prognosis in patients with colon adenocarcinoma.
Collapse
Affiliation(s)
- Marlena Brzozowa-Zasada
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Adam Piecuch
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Marek Michalski
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Katarzyna Stęplewska
- Department of Pathology, Institute of Medical Sciences, University of Opole, 45-052 Opole, Poland
| | - Natalia Matysiak
- Department of Histology and Cell Pathology in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Marek Kucharzewski
- Faculty of Health Sciences, Jan Dlugosz University of Czestochowa, 42-200 Czestochowa, Poland
| |
Collapse
|
4
|
Jácome AA, Johnson B. Minimal Residual Disease in Colorectal Cancer: Are We Finding the Needle in a Haystack? Cells 2023; 12:cells12071068. [PMID: 37048141 PMCID: PMC10092948 DOI: 10.3390/cells12071068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Despite significant advances in the surgical and systemic therapy of colorectal cancer (CRC) in recent decades, recurrence rates remain high. Apart from microsatellite instability status, the decision to offer adjuvant chemotherapy to patients with CRC is solely based on clinicopathologic factors, which offer an inaccurate risk stratification of patients who derive benefit from adjuvant therapy. Owing to the recent improvements of molecular techniques, it has been possible to detect small allelic fractions of circulating tumor DNA (ctDNA), and therefore, to identify patients with minimal residual disease (MRD) after curative-intent therapies. The incorporation of ctDNA identifying MRD in clinical practice may dramatically change the standard of care of CRC, refining the selection of patients who are candidates for escalation and de-escalation of adjuvant chemotherapy, and even for organ-preservation strategies in rectal cancer. In the present review, we describe the current standard of care and the DNA sequencing methodologies and assays, present the data from completed clinical studies and list ongoing potential landmark clinical trials whose results are eagerly awaited, as well as the impact and perspectives for the near future. The discussed data bring optimism for the future of oncologic care through the hope of refined utilization of adjuvant therapies with higher efficacy and safety for patients with both localized and advanced CRC.
Collapse
Affiliation(s)
- Alexandre A. Jácome
- Department of Gastrointestinal Medical Oncology, Oncoclinicas, Belo Horizonte 30360-680, Brazil
| | - Benny Johnson
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
5
|
Evaluation of ctDNA in the Prediction of Response to Neoadjuvant Therapy and Prognosis in Locally Advanced Rectal Cancer Patients: A Prospective Study. Pharmaceuticals (Basel) 2023; 16:ph16030427. [PMID: 36986526 PMCID: PMC10057108 DOI: 10.3390/ph16030427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
“Watch and wait” is becoming a common treatment option for patients with locally advanced rectal cancer (LARC) submitted to neoadjuvant treatment. However, currently, no clinical modality has an acceptable accuracy for predicting pathological complete response (pCR). The aim of this study was to assess the clinical utility of circulating tumor DNA (ctDNA) in predicting the response and prognosis in these patients. We prospectively enrolled a cohort of three Iberian centers between January 2020 and December 2021 and performed an analysis on the association of ctDNA with the main response outcomes and disease-free survival (DFS). The rate of pCR in the total sample was 15.3%. A total of 24 plasma samples from 18 patients were analyzed by next-generation sequencing. At baseline, mutations were detected in 38.9%, with the most common being TP53 and KRAS. Combination of either positive magnetic resonance imaging (MRI) extramural venous invasion (mrEMVI) and ctDNA increased the risk of poor response (p = 0.021). Also, patients with two mutations vs. those with fewer than two mutations had a worse DFS (p = 0.005). Although these results should be read carefully due to sample size, this study suggests that baseline ctDNA combined with mrEMVI could potentially help to predict the response and baseline ctDNA number of mutations might allow the discrimination of groups with different DFS. Further studies are needed to clarify the role of ctDNA as an independent tool in the selection and management of LARC patients.
Collapse
|
6
|
de Abreu AR, Op de Beeck K, Laurent-Puig P, Taly V, Benhaim L. The Position of Circulating Tumor DNA in the Clinical Management of Colorectal Cancer. Cancers (Basel) 2023; 15:1284. [PMID: 36831626 PMCID: PMC9954551 DOI: 10.3390/cancers15041284] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer type worldwide, with over 1.9 million new cases and 935,000 related deaths in 2020. Within the next decade, the incidence of CRC is estimated to increase by 60% and the mortality by 80%. One of the underlying causes of poor prognosis is late detection, with 60 to 70% of the diagnoses occurring at advanced stages. Circulating cell-free DNA (ccfDNA) is probably the most promising tool for screening, diagnosis, prediction of therapeutic response, and prognosis. More specifically, the analysis of the tumor fraction within the ccfDNA (circulating tumor DNA, ctDNA) has great potential to improve the management of CRC. The present review provides an up-to-date and comprehensive overview of the various aspects related to ctDNA detection in CRC.
Collapse
Affiliation(s)
- Ana Regina de Abreu
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
- Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Ken Op de Beeck
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
- Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Pierre Laurent-Puig
- UMR-S1138, CNRS SNC5096, Équipe labélisée Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris, 75006 Paris, France
| | - Valerie Taly
- UMR-S1138, CNRS SNC5096, Équipe labélisée Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris, 75006 Paris, France
| | - Leonor Benhaim
- UMR-S1138, CNRS SNC5096, Équipe labélisée Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris, 75006 Paris, France
- Department of Visceral and Surgical Oncology, Gustave Roussy, Cancer Campus, 114 rue Edouard Vaillant, 94805 Villejuif, France
| |
Collapse
|
7
|
Machado Carvalho JV, Dutoit V, Corrò C, Koessler T. Promises and Challenges of Predictive Blood Biomarkers for Locally Advanced Rectal Cancer Treated with Neoadjuvant Chemoradiotherapy. Cells 2023; 12:413. [PMID: 36766755 PMCID: PMC9913546 DOI: 10.3390/cells12030413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
The treatment of locally advanced rectal cancer (LARC) requires a multimodal approach combining neoadjuvant radiotherapy or chemoradiotherapy (CRT) and surgery. Predicting tumor response to CRT can guide clinical decision making and improve patient care while avoiding unnecessary toxicity and morbidity. Circulating biomarkers offer both the advantage to be easily accessed and followed over time. In recent years, biomarkers such as proteins, blood cells, or nucleic acids have been investigated for their predictive value in oncology. We conducted a comprehensive literature review with the aim to summarize the status of circulating biomarkers predicting response to CRT in LARC. Forty-nine publications, of which forty-seven full-text articles, one review and one systematic review, were retrieved. These studies evaluated circulating markers (CEA and CA 19-9), inflammatory biomarkers (CRP, albumin, and lymphocytes), hematologic markers (hemoglobin and thrombocytes), lipids and circulating nucleic acids (cell-free DNA [cfDNA], circulating tumor DNA [ctDNA], and microRNA [miRNA]). Post-CRT CEA levels had the most consistent association with tumor response, while cfDNA integrity index, MGMT promoter methylation, ERCC-1, miRNAs, and miRNA-related SNPs were identified as potential predictive markers. Although circulating biomarkers hold great promise, inconsistent results, low statistical power, and low specificity and sensibility prevent them from reliably predicting tumor response following CRT. Validation and standardization of methods and technologies are further required to confirm results.
Collapse
Affiliation(s)
- Joao Victor Machado Carvalho
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Swiss Cancer Center Léman, 1005 Lausanne, Switzerland
- Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Valérie Dutoit
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Swiss Cancer Center Léman, 1005 Lausanne, Switzerland
| | - Claudia Corrò
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Swiss Cancer Center Léman, 1005 Lausanne, Switzerland
- Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Thibaud Koessler
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Swiss Cancer Center Léman, 1005 Lausanne, Switzerland
- Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
| |
Collapse
|
8
|
Moran JA, Adams DL, Edelman MJ, Lopez P, He J, Qiao Y, Xu T, Liao Z, Gardner KP, Tang CM, Lin SH. Monitoring PD-L1 Expression on Circulating Tumor-Associated Cells in Recurrent Metastatic Non-Small-Cell Lung Carcinoma Predicts Response to Immunotherapy With Radiation Therapy. JCO Precis Oncol 2022; 6:e2200457. [PMID: 36516370 PMCID: PMC10166406 DOI: 10.1200/po.22.00457] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Current diagnostic methods to determine programmed death 1 (PD-1) receptor and its ligand (PD-L1)/PD-1 immunotherapy (immune checkpoint inhibitor [ICI]) efficacy in recurrent or metastatic non-small-cell lung carcinoma (rmNSCLC) are imprecise. Although previously shown that patients with high tumor PD-L1 (≥ 50%) demonstrate clinical benefit in the form of disease reduction and improved survival, patients with low PD-L1 (< 50%) sometimes benefit from treatment. Since the PD-L1/PD-1 pathway is dynamic, monitoring PD-L1 levels during treatment may be more accurate than a static baseline tumor biopsy; however, rebiopsying the primary or metastatic disease is rarely feasible. Liquid biopsies that measure the upregulation of PD-L1 on tumor-associated cells (TACs), ie, cancer-associated macrophage-like cells and circulating tumor cells, have been performed, but their predictive value for ICI therapy efficacy is unknown. MATERIALS AND METHODS We initiated a single-blind prospective study to evaluate TAC PD-L1 expression changes in rmNSCLC from blood samples before (T0) and after (T1) treatment with ICI (ICI, n = 41) or without ICI (no ICI, n = 41). Anonymized blood was filtered to isolate TACs, which were then quantified for high/low PD-L1 expression. Progression-free survival (PFS) or overall survival (OS) hazard ratios (HRs) were evaluated at 18 and 24 months by censored univariate analysis. RESULTS Increased TAC PD-L1 expression between T0 and T1 in patients who were not treated with ICI had no relationship with PFS or OS. However, increased TAC PD-L1 expression between T0 and T1 in patients treated with ICI had significantly better PFS (HR, 3.49; 95% CI, 1.5 to 8.3; P = .0091) and OS (HR, 3.058; 95% CI, 1.2 to 7.9; P = .0410). CONCLUSION Blood-based monitoring of dynamic changes in PD-L1 in TACs appears to identify patients with rmNSCLC who may benefit from ICI.
Collapse
Affiliation(s)
- Jillian A. Moran
- Rutgers, The State University of New Jersey, New Brunswick, NJ
- Creatv MicroTech, Inc, Monmouth Junction, NJ
| | - Daniel L. Adams
- Rutgers, The State University of New Jersey, New Brunswick, NJ
- Creatv MicroTech, Inc, Monmouth Junction, NJ
| | | | | | | | | | - Ting Xu
- MD Anderson Cancer Center, Houston, TX
| | | | - Kirby P. Gardner
- Creatv MicroTech, Inc, Monmouth Junction, NJ
- Rutgers University, School of Graduate Studies, Piscataway, NJ
| | | | | |
Collapse
|
9
|
Roesel R, Epistolio S, Molinari F, Saletti P, De Dosso S, Valli M, Franzetti-Pellanda A, Deantonio L, Biggiogero M, Spina P, Popeskou SG, Cristaudi A, Mongelli F, Mazzucchelli L, Stefanini FM, Frattini M, Christoforidis D. A Pilot, Prospective, Observational Study to Investigate the Value of NGS in Liquid Biopsies to Predict Tumor Response After Neoadjuvant Chemo-Radiotherapy in Patients With Locally Advanced Rectal Cancer: The LiBReCa Study. Front Oncol 2022; 12:900945. [PMID: 35837093 PMCID: PMC9274270 DOI: 10.3389/fonc.2022.900945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionCirculating tumor DNA (ctDNA) correlates with the response to therapy in different types of cancer. However, in patients with locally advanced rectal cancer (LARC), little is known about how ctDNA levels change with neoadjuvant chemoradiation (Na-ChRT) and how they correlate with treatment response. This work aimed to explore the value of serial liquid biopsies in monitoring response after Na-ChRT with the hypothesis that this could become a reliable biomarker to identify patients with a complete response, candidates for non-operative management.Materials and MethodsTwenty-five consecutive LARC patients undergoing long-term Na-ChRT therapy were included. Applying next-generation sequencing (NGS), we characterized DNA extracted from formalin-fixed paraffin embedded diagnostic biopsy and resection tissue and plasma ctDNA collected at the following time points: the first and last days of radiotherapy (T0, Tend), at 4 (T4), 7 (T7) weeks after radiotherapy, on the day of surgery (Top), and 3–7 days after surgery (Tpost-op). On the day of surgery, a mesenteric vein sample was also collected (TIMV). The relationship between the ctDNA at those time-points and the tumor regression grade (TRG) of the surgical specimen was statistically explored.ResultsWe found no association between the disappearance of ctDNA mutations in plasma samples and pathological complete response (TRG1) as ctDNA was undetectable in the majority of patients from Tend on. However, we observed that the poor (TRG 4) response to Na-ChRT was significantly associated with a positive liquid biopsy at the Top.ConclusionsctDNA evaluation by NGS technology may identify LARC patients with poor response to Na-ChRT. In contrast, this technique does not seem useful for identifying patients prone to developing a complete response.
Collapse
Affiliation(s)
- Raffaello Roesel
- Department of Visceral Surgery, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
- *Correspondence: Raffaello Roesel, ; Samantha Epistolio,
| | - Samantha Epistolio
- Istituto Cantonale di Patologia, Ente Ospedaliero Cantonale, Locarno, Switzerland
- *Correspondence: Raffaello Roesel, ; Samantha Epistolio,
| | - Francesca Molinari
- Istituto Cantonale di Patologia, Ente Ospedaliero Cantonale, Locarno, Switzerland
| | - Piercarlo Saletti
- Clinical Research Unit, Clinica Luganese Moncucco, Lugano, Switzerland
- Medical Oncology Clinic, Clinica Luganese Moncucco, Lugano, Switzerland
- Faculty of Biomedical Sciences, University of Southern Switzerland, Lugano, Switzerland
| | - Sara De Dosso
- Faculty of Biomedical Sciences, University of Southern Switzerland, Lugano, Switzerland
- Istituto Oncologico della Svizzera Italiana, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Mariacarla Valli
- Radiation Oncology Clinic, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | | | - Letizia Deantonio
- Faculty of Biomedical Sciences, University of Southern Switzerland, Lugano, Switzerland
- Radiation Oncology Clinic, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Maira Biggiogero
- Clinical Research Unit, Clinica Luganese Moncucco, Lugano, Switzerland
| | - Paolo Spina
- Istituto Cantonale di Patologia, Ente Ospedaliero Cantonale, Locarno, Switzerland
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Sotirios Georgios Popeskou
- Department of Visceral Surgery, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Alessandra Cristaudi
- Department of Visceral Surgery, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Francesco Mongelli
- Department of Visceral Surgery, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Luca Mazzucchelli
- Istituto Cantonale di Patologia, Ente Ospedaliero Cantonale, Locarno, Switzerland
- Faculty of Biomedical Sciences, University of Southern Switzerland, Lugano, Switzerland
| | | | - Milo Frattini
- Istituto Cantonale di Patologia, Ente Ospedaliero Cantonale, Locarno, Switzerland
| | - Dimitri Christoforidis
- Department of Visceral Surgery, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, University of Southern Switzerland, Lugano, Switzerland
- Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
10
|
Identifying Circulating Tumor DNA Mutations Associated with Neoadjuvant Chemotherapy Efficacy in Local Advanced Breast Cancer. Appl Biochem Biotechnol 2022; 194:3961-3973. [PMID: 35579744 DOI: 10.1007/s12010-022-03946-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Circulating tumor DNA (ctDNA) correlates with tumor burden and provides early detection of treatment response and tumor genetic alterations in breast cancer. Neoadjuvant chemotherapy (NACT) has become standard therapy for local advanced breast cancer (LABC). The aim of our study was to investigate plasma ctDNA as a prognostic marker for outcome in patients with LABC treated with NACT. A total of 56 patients with LABC were involved in this study. ctDNA mutations were investigated by using a 100 gene panel-target capture next-generation sequencing. The patients then received standard NACT therapy: adriamycin and cyclophosphamide and paclitaxel (AC-T) or AC-TH (AC-T+ Trastuzumab) regimen. The efficacy of NACT was evaluated by Miller-Payne grading system. A predictive and weight model was used to screen ctDNA point mutation biomarkers for NACT. The ctDNA mutational profile of LABC patients was identified. For nonsynonymous mutations, the top 5 mutated genes were MTHFR (51/56, 91.1%), XPC (50/56, 89.3%), ABCB1 (48/51, 94.1%), BRCA2 (38/56, 67.9%), and XRCC1 (38/56, 67.9%). In addition, the mutation frequencies of PIK3CA and TP53 were 32.1% (18/56) and 26.8% (15/56), respectively. The predictive model indicated that XRCC1 44055726 (TG>-) mutation (25/56, 44.6%) was significantly associated with Miller-Payne 4-5 and Miller-Payne 3-5 responses. While mTOR 11249132(G>C) mutation (23/56, 41.1%) was associated with Miller-Payne 1-4 or Miller-Payne 1-3 responses. Furthermore, XRCC1 44055726 (TG>-) accompanied by mTOR wild type predicted a good NACT efficacy in all response classification systems. The ROC curves to discriminate good neoadjuvant chemotherapy efficiency (Miller-Payne 4-5) and poor efficiency (Miller-Payne 1-3) were created, and AUC value was 0.77. Our results suggested that ctDNA mutation of XRCC1 44055726 (TG>-) might be a positive biomarker for NACT therapy in LABC, while mTOR 11249132(G>C) mutation was potentially associated with NACT resistance.
Collapse
|
11
|
|
12
|
Clinical Applications of Minimal Residual Disease Assessments by Tumor-Informed and Tumor-Uninformed Circulating Tumor DNA in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13184547. [PMID: 34572774 PMCID: PMC8471730 DOI: 10.3390/cancers13184547] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Circulating tumor DNA, or ctDNA, are fragments of tumor DNA that can be detected in the blood of patients with colorectal cancer. Measuring ctDNA levels in the blood has shown the potential to provide important information that can be helpful in the clinical care of patients with colorectal cancer. For example, in patients with colon cancer that has been removed by surgery, measuring ctDNA in the blood can predict the likelihood of cancer recurrence, while in those with metastatic colorectal cancer, measuring ctDNA can inform the clinician whether chemotherapy is effective at earlier timepoints than currently available tests. In this review, we discuss the results from ongoing studies describing the utility of ctDNA measurements across all stages of colorectal cancer. We also discuss the various clinical scenarios that ctDNA may have the most immediate impact in colorectal cancer management. Abstract Emerging data suggest that circulating tumor DNA (ctDNA) can detect colorectal cancer (CRC)-specific signals across both non-metastatic and metastatic settings. With the development of multiple platforms, including tumor-informed and tumor-agnostic ctDNA assays and demonstration of their provocative analytic performance to detect minimal residual disease, there are now ongoing, phase III randomized clinical trials to evaluate their role in the management paradigm of CRC. In this review, we highlight landmark studies that have formed the basis for ongoing studies on the clinically applicability of plasma ctDNA assays in resected, stage I–III CRC and metastatic CRC. We discuss clinical settings by which ctDNA may have the most immediate impact in routine clinical practice. These include the potential for ctDNA to (1) guide surveillance and intensification or de-intensification strategies of adjuvant therapy in resected, stage I–III CRC, (2) predict treatment response to neoadjuvant therapy in locally advanced rectal cancer inclusive of total neoadjuvant therapy (TNT), and (3) predict response to systemic and surgical therapies in metastatic disease. We end by considering clinical variables that can influence our ability to reliably interpret ctDNA dynamics in the clinic.
Collapse
|
13
|
Morais M, Pinto DM, Machado JC, Carneiro S. ctDNA on liquid biopsy for predicting response and prognosis in locally advanced rectal cancer: A systematic review. Eur J Surg Oncol 2021; 48:218-227. [PMID: 34511270 DOI: 10.1016/j.ejso.2021.08.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The management of locally advanced rectal cancer (LARC) requires a multidisciplinary approach, with an increasing interest for non-operative strategies. Liquid biopsy for obtaining circulating tumor DNA (ctDNA) can provide information on neoadjuvant chemoradiotherapy (nCRT) pathological response and cancer-specific prognosis, and therefore might be a promising guide for these treatments. METHODS A systematic review of the studies available in literature has been performed to assess the role of ctDNA as a predictive and prognostic biomarker in LARC patients. RESULTS We retrieved 21 publications, of which 17 full-text articles and 4 abstracts. Results have been labelled into two groups: predictive and prognostic. Data about the usefulness of liquid biopsy in this setting is still inconclusive. However, baseline higher levels of longer fragments of cell-free DNA and integrity index, tumor-specific mutations and certain methylated genes could predict non-responders. Also, undetectable baseline ctDNA and decrease of common rectal cancer mutations throughout treatment (dynamic monitoring) were predictive factors of pathological complete response. The continuous detection of ctDNA in different timepoints of treatment (minimal residual disease) was consistently associated with worse prognosis. CONCLUSIONS ctDNA is a promising biomarker that could assist predicting treatment response to nCRT and prognosis in patients with LARC. The ideal methods and timings for the liquid biopsy still have to be defined.
Collapse
Affiliation(s)
- Marina Morais
- Surgery Department, Hospital Pedro Hispano, Matosinhos, Portugal; Surgery Department, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Diogo Melo Pinto
- Surgery Department, Hospital Pedro Hispano, Matosinhos, Portugal
| | - José Carlos Machado
- I3S - Institute for Research and Innovation in Health and IPATIMUP, Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal; Pathology Department, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Silvestre Carneiro
- Surgery Department, Faculty of Medicine, University of Porto, Porto, Portugal; Surgery Department, Centro Hospitalar de São João, Porto, Portugal
| |
Collapse
|
14
|
The relevance of liquid biopsy in surgical oncology: The application of perioperative circulating nucleic acid dynamics in improving patient outcomes. Surgeon 2021; 20:e163-e173. [PMID: 34362650 DOI: 10.1016/j.surge.2021.06.006] [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: 01/06/2021] [Revised: 06/13/2021] [Accepted: 06/23/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Liquid biopsy is gaining increasing clinical utility in the management of cancer patients. The main components of a liquid biopsy are circulating nucleic acids, circulating tumour cells and extracellular vesicles such as exosomes. Circulating nucleic acids including cell free DNA (cfDNA) and circulating tumour DNA (ctDNA) in particular have been the focus of recent attention as they have demonstrated excellent potential in cancer screening, provision of prognostic information and in genomic profiling of a tumour without the need for repeated tissue biopsies. The aim of this review was to explore the current evidence in relation to the use of liquid biopsy in the perioperative setting and identify ways in which liquid biopsy may be applied in the future. METHODS This narrative review is based on a comprehensive literature search up to the 1st of June 2020 for papers relevant to the application of liquid biopsy in surgical oncology, focusing particularly on the perioperative period. RESULTS Recent evidence has demonstrated that perioperative liquid biopsy can accurately stratify patients' risk of recurrence compared to conventional biomarkers. Attention to the perioperative dynamics of liquid biopsy components can potentially provide new understanding of the complex relationship between surgery and cancer outcome. In addition, careful evaluation of liquid biopsy components in the perioperative window may provide important diagnostic and therapeutic information for cancer patients. CONCLUSION The rapidly evolving concept of the liquid biopsy has the potential to become the cornerstone for decision making around surveillance and adjuvant therapies the era of personalised medicine.
Collapse
|
15
|
Abstract
Response evaluation for cancer treatment consists primarily of clinical and radiological assessments. In addition, a limited number of serum biomarkers that assess treatment response are available for a small subset of malignancies. Through recent technological innovations, new methods for measuring tumor burden and treatment response are becoming available. By utilization of highly sensitive techniques, tumor-specific mutations in circulating DNA can be detected and circulating tumor DNA (ctDNA) can be quantified. These so-called liquid biopsies provide both molecular information about the genomic composition of the tumor and opportunities to evaluate tumor response during therapy. Quantification of tumor-specific mutations in plasma correlates well with tumor burden. Moreover, with liquid biopsies, it is also possible to detect mutations causing secondary resistance during treatment. This review focuses on the clinical utility of ctDNA as a response and follow-up marker in patients with non-small cell lung cancer, melanoma, colorectal cancer, and breast cancer. Relevant studies were retrieved from a literature search using PubMed database. An overview of the available literature is provided and the relevance of ctDNA as a response marker in anti-cancer therapy for clinical practice is discussed. We conclude that the use of plasma-derived ctDNA is a promising tool for treatment decision-making based on predictive testing, detection of resistance mechanisms, and monitoring tumor response. Necessary steps for translation to daily practice and future perspectives are discussed.
Collapse
|
16
|
Koessler T, Paradiso V, Piscuoglio S, Nienhold R, Ho L, Christinat Y, Terracciano LM, Cathomas G, Wicki A, McKee TA, Nouspikel T. Reliability of liquid biopsy analysis: an inter-laboratory comparison of circulating tumor DNA extraction and sequencing with different platforms. J Transl Med 2020; 100:1475-1484. [PMID: 32616816 DOI: 10.1038/s41374-020-0459-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 01/11/2023] Open
Abstract
Liquid biopsy, the analysis of circulating tumor DNA (ctDNA), is a promising tool in oncology, especially in personalized medicine. Although its main applications currently focus on selection and adjustment of therapy, ctDNA may also be used to monitor residual disease, establish prognosis, detect relapses, and possibly screen at-risk individuals. CtDNA represents a small and variable proportion of circulating cell-free DNA (ccfDNA) which is itself present at a low concentration in normal individuals and so analyzing ctDNA is technically challenging. Various commercial systems have recently appeared on the market, but it remains difficult for practitioners to compare their performance and to determine whether they yield comparable results. As a first step toward establishing national guidelines for ctDNA analyses, four laboratories in Switzerland joined a comparative exercise to assess ccfDNA extraction and ctDNA analysis by sequencing. Extraction was performed using six distinct methods and yielded ccfDNA of equally high quality, suitable for sequencing. Sequencing of synthetic samples containing predefined amounts of eight mutations was performed on three different systems, with similar results. In all four laboratories, mutations were easily identified down to 1% allele frequency, whereas detection at 0.1% proved challenging. Linearity was excellent in all cases and while molecular yield was superior with one system this did not impact on sensitivity. This study also led to several additional conclusions: First, national guidelines should concentrate on principles of good laboratory practice rather than recommend a particular system. Second, it is essential that laboratories thoroughly validate every aspect of extraction and sequencing, in particular with respect to initial amount of DNA and average sequencing depth. Finally, as software proved critical for mutation detection, laboratories should validate the performance of variant callers and underlying algorithms with respect to various types of mutations.
Collapse
Affiliation(s)
- Thibaud Koessler
- Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
| | - Viola Paradiso
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Salvatore Piscuoglio
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland.,Visceral surgery research laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ronny Nienhold
- Institute of Pathology, Cantonal Hospital Basel-Land, Liestal, Switzerland
| | - Liza Ho
- Clinical Pathology Service, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Yann Christinat
- Clinical Pathology Service, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Luigi M Terracciano
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Gieri Cathomas
- Visceral surgery research laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Andreas Wicki
- Department of Oncology & Hematology, Medical University Clinic, Cantonal Hospital Basel-Land, Liestal, and University of Basel, Basel, Switzerland
| | - Thomas A McKee
- Clinical Pathology Service, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Thierry Nouspikel
- Medical Genetics, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
17
|
De Palma FDE, Luglio G, Tropeano FP, Pagano G, D’Armiento M, Kroemer G, Maiuri MC, De Palma GD. The Role of Micro-RNAs and Circulating Tumor Markers as Predictors of Response to Neoadjuvant Therapy in Locally Advanced Rectal Cancer. Int J Mol Sci 2020; 21:7040. [PMID: 32987896 PMCID: PMC7582560 DOI: 10.3390/ijms21197040] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023] Open
Abstract
The response to neoadjuvant chemoradiation (nCRT) is a critical step in the management of locally advanced rectal cancer (LARC) patients. Only a minority of LARC patients responds completely to neoadjuvant treatments, thus avoiding invasive radical surgical resection. Moreover, toxic side effects can adversely affect patients' survival. The difficulty in separating in advances responder from non-responder patients affected by LARC highlights the need for valid biomarkers that guide clinical decision-making. In this context, microRNAs (miRNAs) seem to be promising candidates for predicting LARC prognosis and/or therapy response, particularly due to their stability, facile detection, and disease-specific expression in human tissues, blood, serum, or urine. Although a considerable number of studies involving potential miRNA predictors to nCRT have been conducted over the years, to date, the identification of the perfect miRNA signatures or single miRNA, as well as their use in the clinical practice, is still representing a challenge for the management of LARC patients. In this review, we will first introduce LARC and its difficult management. Then, we will trace the scientific history and the key obstacles for the identification of specific miRNAs that predict responsiveness to nCRT. There is a high potential to identify non-invasive biomarkers that circulate in the human bloodstream and that might indicate the LARC patients who benefit from the watch-and-wait approach. For this, we will critically evaluate recent advances dealing with cell-free nucleic acids including miRNAs and circulating tumor cells as prognostic or predictive biomarkers.
Collapse
Affiliation(s)
- Fatima Domenica Elisa De Palma
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université of Paris, 75005 Paris, France; (G.K.); (M.C.M.)
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94800 Villejuif, France
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy
| | - Gaetano Luglio
- Department of Public Health, University of Naples “Federico II”, 80138 Naples, Italy; (G.L.); (M.D.)
| | - Francesca Paola Tropeano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80138 Naples, Italy; (F.P.T.); (G.P.)
| | - Gianluca Pagano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80138 Naples, Italy; (F.P.T.); (G.P.)
| | - Maria D’Armiento
- Department of Public Health, University of Naples “Federico II”, 80138 Naples, Italy; (G.L.); (M.D.)
| | - Guido Kroemer
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université of Paris, 75005 Paris, France; (G.K.); (M.C.M.)
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94800 Villejuif, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou 100864, China
- Department of Women’s and Children’s Health, Karolinska Institutet, 171 77 Stockholm, Sweden
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | - Maria Chiara Maiuri
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université of Paris, 75005 Paris, France; (G.K.); (M.C.M.)
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94800 Villejuif, France
| | - Giovanni Domenico De Palma
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80138 Naples, Italy; (F.P.T.); (G.P.)
- Centro Interuniversitario di Studi per l’Innovazione Tecnologica in Chirurgia, University of Naples Federico II, 80138 Naples, Italy
| |
Collapse
|
18
|
Polivka J, Windrichova J, Pesta M, Houfkova K, Rezackova H, Macanova T, Vycital O, Kucera R, Slouka D, Topolcan O. The Level of Preoperative Plasma KRAS Mutations and CEA Predict Survival of Patients Undergoing Surgery for Colorectal Cancer Liver Metastases. Cancers (Basel) 2020; 12:cancers12092434. [PMID: 32867151 PMCID: PMC7565270 DOI: 10.3390/cancers12092434] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 12/30/2022] Open
Abstract
Colorectal cancer (CRC) belongs to the most common cancers. The liver is a predominant site of CRC dissemination. Novel biomarkers for predicting the survival of CRC patients with liver metastases (CLM) undergoing metastasectomy are needed. We examined KRAS mutated circulating cell-free tumor DNA (ctDNA) in CLM patients as a prognostic biomarker, independently or in combination with carcinoembryonic antigen (CEA). Thereby, a total of 71 CLM were retrospectively analyzed. Seven KRAS G12/G13 mutations was analyzed by a ddPCR™ KRAS G12/G13 Screening Kit on QX200 Droplet Digital PCR System (Bio-Rad Laboratories, Hercules, CA, USA) in liver metastasis tissue and preoperative and postoperative plasma samples. CEA were determined by an ACCESS CEA assay with the UniCel DxI 800 Instrument (Beckman Coulter, Brea, CA, USA). Tissue KRAS positive liver metastases was detected in 33 of 69 patients (47.8%). Preoperative plasma samples were available in 30 patients and 11 (36.7%) were KRAS positive. The agreement between plasma- and tissue-based KRAS mutation status was 75.9% (22 in 29; kappa 0.529). Patients with high compared to low levels of preoperative plasma KRAS fractional abundance (cut-off 3.33%) experienced shorter overall survival (OS 647 vs. 1392 days, p = 0.003). The combination of high preoperative KRAS fractional abundance and high CEA (cut-off 3.33% and 4.9 µg/L, resp.) best predicted shorter OS (HR 13.638, 95%CI 1.567–118.725) in multivariate analysis also (OS HR 44.877, 95%CI 1.59–1266.479; covariates: extend of liver resection, biological treatment). KRAS mutations are detectable and quantifiable in preoperative plasma cell-free DNA, incompletely overlapping with tissue biopsy. KRAS mutated ctDNA is a prognostic factor for CLM patients undergoing liver metastasectomy. The best prognostic value can be reached by a combination of ctDNA and tumor marker CEA.
Collapse
Affiliation(s)
- Jiri Polivka
- Department of Histology and Embryology and Biomedical Center, Charles University, Faculty of Medicine in Pilsen, Karlovarska 48, 30166 Pilsen, Czech Republic;
- Laboratory of Immunoanalysis, University Hospital in Pilsen, E. Benese 13, 30599 Pilsen, Czech Republic; (J.W.); (H.R.); (R.K.); (D.S.); (O.T.)
| | - Jindra Windrichova
- Laboratory of Immunoanalysis, University Hospital in Pilsen, E. Benese 13, 30599 Pilsen, Czech Republic; (J.W.); (H.R.); (R.K.); (D.S.); (O.T.)
| | - Martin Pesta
- Laboratory of Immunoanalysis, University Hospital in Pilsen, E. Benese 13, 30599 Pilsen, Czech Republic; (J.W.); (H.R.); (R.K.); (D.S.); (O.T.)
- Department of Biology, Charles University, Faculty of Medicine in Pilsen, alej Svobody 76, 32300 Pilsen, Czech Republic; (K.H.); (T.M.)
- Correspondence: ; Tel.: +420-377-593-261
| | - Katerina Houfkova
- Department of Biology, Charles University, Faculty of Medicine in Pilsen, alej Svobody 76, 32300 Pilsen, Czech Republic; (K.H.); (T.M.)
| | - Hana Rezackova
- Laboratory of Immunoanalysis, University Hospital in Pilsen, E. Benese 13, 30599 Pilsen, Czech Republic; (J.W.); (H.R.); (R.K.); (D.S.); (O.T.)
| | - Tereza Macanova
- Department of Biology, Charles University, Faculty of Medicine in Pilsen, alej Svobody 76, 32300 Pilsen, Czech Republic; (K.H.); (T.M.)
| | - Ondrej Vycital
- Department of Surgery, University Hospital in Pilsen, E. Beneše 13, 30599 Pilsen, Czech Republic;
| | - Radek Kucera
- Laboratory of Immunoanalysis, University Hospital in Pilsen, E. Benese 13, 30599 Pilsen, Czech Republic; (J.W.); (H.R.); (R.K.); (D.S.); (O.T.)
| | - David Slouka
- Laboratory of Immunoanalysis, University Hospital in Pilsen, E. Benese 13, 30599 Pilsen, Czech Republic; (J.W.); (H.R.); (R.K.); (D.S.); (O.T.)
| | - Ondrej Topolcan
- Laboratory of Immunoanalysis, University Hospital in Pilsen, E. Benese 13, 30599 Pilsen, Czech Republic; (J.W.); (H.R.); (R.K.); (D.S.); (O.T.)
| |
Collapse
|
19
|
Pazdirek F, Minarik M, Benesova L, Halkova T, Belsanova B, Macek M, Stepanek L, Hoch J. Monitoring of Early Changes of Circulating Tumor DNA in the Plasma of Rectal Cancer Patients Receiving Neoadjuvant Concomitant Chemoradiotherapy: Evaluation for Prognosis and Prediction of Therapeutic Response. Front Oncol 2020; 10:1028. [PMID: 32793464 PMCID: PMC7394215 DOI: 10.3389/fonc.2020.01028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/22/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction: Patients with locally advanced rectal cancer (LARC) are undergoing neoadjuvant chemoradiotherapy (NCRT) prior to surgery. Although in some patients the NCRT is known to prevent local recurrence, it is also accompanied by side effects. Accordingly, there is an unmet need to identify predictive markers allowing to identify non-responders to avoid its adverse effects. We monitored circulating tumor DNA (ctDNA) as a potential liquid biopsy-based biomarker. We have investigated ctDNA changes plasma during the early days of NCRT and its relationship to the overall therapy outcome. Methods and Patients: The studied cohort included 36 LARC patients (stage II or III) undergoing NCRT with subsequent surgical treatment. We have detected somatic mutations in tissue biopsies taken during endoscopic examination prior to the therapy. CtDNA was extracted from patient plasma samples prior to therapy and at the end of the first week. In order to optimize the analytical costs of liquid-biopsy testing, we have utilized a two-level approach in which first a low-cost detection method of denaturing capillary electrophoresis was used followed by examination of initially negative samples by a high-sensitivity BEAMING assay. The ctDNA was related to clinical parameters including tumor regression grade (TRG) and TNM tumor staging. Results: We have detected a somatic mutation in 33 out of 36 patients (91.7%). Seven patients (7/33, 21.2%) had ctDNA present prior to therapy. The ctDNA positivity before treatment reduced post-operative disease-free survival and overall survival by an average of 1.47 and 1.41 years, respectively (p = 0.015, and p = 0.010). In all patients, ctDNA was strongly reduced or completely eliminated from plasma by the end of the first week of NCRT, with no correlation to any of the parameters analyzed. Conclusions: The baseline ctDNA presence represented a statistically significant negative prognostic biomarker for the overall patient survival. As ctDNA was reduced indiscriminately from circulation of all patients, dynamics during the first week of NCRT is not suited for predicting the outcome of LARC. However, the general effect of rapid ctDNA disappearance apparently occurring during the initial days of NCRT is noteworthy and should further be studied.
Collapse
Affiliation(s)
- Filip Pazdirek
- Department of Surgery, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Marek Minarik
- Department of Surgery, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia.,Elphogene, Prague, Czechia
| | - Lucie Benesova
- Center for Applied Genomics of Solid Tumors (CEGES), Genomac Research Institute, Prague, Czechia
| | - Tereza Halkova
- Center for Applied Genomics of Solid Tumors (CEGES), Genomac Research Institute, Prague, Czechia
| | | | - Milan Macek
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Lubomír Stepanek
- Institute of Biophysics and Informatics, 1st Faculty of Medicine, Charles University, Prague, Czechia
| | - Jiri Hoch
- Department of Surgery, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| |
Collapse
|
20
|
Prognostic Value of Serum NPY Hypermethylation in Neoadjuvant Chemoradiotherapy for Rectal Cancer: Secondary Analysis of a Randomized Trial. Am J Clin Oncol 2020; 43:9-13. [PMID: 31569168 DOI: 10.1097/coc.0000000000000609] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Long-term prevention of metastatic disease remains a challenge in locally advanced rectal cancer, and robust pretreatment prognostic factors for metastatic progression are lacking. We hypothesized that detecting circulating tumor-specific DNA (ctDNA) based on hypermethylation of the neuropeptide Y gene (meth-ctDNA) could be a prognostic marker in the neoadjuvant setting; we examined this in a secondary, explorative analysis of a prospective trial. MATERIALS AND METHODS Serum samples were prospectively collected in a phase III trial for locally advanced rectal cancer. Positivity for and fractional abundance of meth-ctDNA in baseline samples were estimated. Overall survival (OS) and the rate of distant metastases were compared between meth-ctDNA positive and negative patients; other prognostic factors were controlled for in multivariate Cox regression. Importance of quantitative load was examined by considering the fractional abundance of meth-ctDNA relative to total circulating DNA. RESULTS Baseline serum samples were available for 146 patients. In total, 30 patients had presence of meth-ctDNA, with no correlation with cT (P=0.8) or cN (P=0.6) stages. Median follow-up was 10.6 years for OS and 5.1 years for freedom from distant metastases. Patients with meth-ctDNA had significantly worse 5-year OS (47% vs. 69%), even when controlling for other prognostic factors (hazard ratio=2.08; 95% confidence interval, 1.23-1.51). This seemed mainly driven by disparity in the rate of distant metastases (55% vs. 72% at 5 y, P=0.01); hazard ratio=2.20 (95% confidence interval, 1.19-4.07, P=0.01) in multivariate analysis. Increased quantitative load was highly significant for worse outcomes. CONCLUSIONS Meth-ctDNA could be a potential prognostic marker in the neoadjuvant setting and may, if validated, identify patients at increased risk of distant metastases.
Collapse
|
21
|
Khakoo S, Carter PD, Brown G, Valeri N, Picchia S, Bali MA, Shaikh R, Jones T, Begum R, Rana I, Wotherspoon A, Terlizzo M, von Loga K, Kalaitzaki E, Saffery C, Watkins D, Tait D, Chau I, Starling N, Hubank M, Cunningham D. MRI Tumor Regression Grade and Circulating Tumor DNA as Complementary Tools to Assess Response and Guide Therapy Adaptation in Rectal Cancer. Clin Cancer Res 2020; 26:183-192. [PMID: 31852830 DOI: 10.1158/1078-0432.ccr-19-1996] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/30/2019] [Accepted: 10/21/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Response to preoperative chemo-radiotherapy (CRT) varies. We assessed whether circulating tumor DNA (ctDNA) might be an early indicator of tumor response or progression to guide therapy adaptation in rectal cancer. EXPERIMENTAL DESIGN A total of 243 serial plasma samples were analyzed from 47 patients with localized rectal cancer undergoing CRT. Up to three somatic variants were tracked in plasma using droplet digital PCR. RECIST and MRI tumor regression grade (mrTRG) evaluated response. Survival analyses applied Kaplan-Meier method and Cox regression. RESULTS ctDNA detection rates were: 74% (n = 35/47) pretreatment, 21% (n = 10/47) mid CRT, 21% (n = 10/47) after completing CRT, and 13% (n = 3/23) after surgery. ctDNA status after CRT was associated with primary tumor response by mrTRG (P = 0.03). With a median follow-up of 26.4 months, metastases-free survival was shorter in patients with detectable ctDNA after completing CRT [HR 7.1; 95% confidence interval (CI), 2.4-21.5; P < 0.001], persistently detectable ctDNA pre and mid CRT (HR 3.8; 95% CI, 1.2-11.7; P = 0.02), and pre, mid, and after CRT (HR 11.5; 95% CI, 3.3-40.4; P < 0.001) compared with patients with undetectable or nonpersistent ctDNA. In patients with detectable ctDNA, a fractional abundance threshold of ≥0.07% mid CRT or ≥0.13% after completing CRT predicted for metastases with 100% sensitivity and 83.3% specificity for mid CRT and 66.7% for CRT completion. All 3 patients with detectable ctDNA post-surgery relapsed compared with none of the 20 patients with undetectable ctDNA (P = 0.001). CONCLUSIONS ctDNA identified patients at risk of developing metastases during the neoadjuvant period and post-surgery, and could be used to tailor treatment.
Collapse
Affiliation(s)
- Shelize Khakoo
- Department of Medicine, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Paul David Carter
- Clinical Genomics, The Royal Marsden Hospital NHS Foundation Trust, Sutton, United Kingdom
| | - Gina Brown
- Department of Radiology, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Nicola Valeri
- Molecular Pathology, The Institute of Cancer Research/The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Simona Picchia
- Department of Radiology, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Maria Antonietta Bali
- Department of Radiology, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Ridwan Shaikh
- Clinical Genomics, The Royal Marsden Hospital NHS Foundation Trust, Sutton, United Kingdom
| | - Thomas Jones
- Clinical Genomics, The Royal Marsden Hospital NHS Foundation Trust, Sutton, United Kingdom
| | - Ruwaida Begum
- Department of Medicine, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Isma Rana
- Department of Medicine, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Andrew Wotherspoon
- Department of Histopathology, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Monica Terlizzo
- Department of Histopathology, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Katharina von Loga
- Department of Histopathology, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Eleftheria Kalaitzaki
- Clinical Research and Development, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Claire Saffery
- Department of Medicine, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - David Watkins
- Department of Medicine, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Diana Tait
- Department of Medicine, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Ian Chau
- Department of Medicine, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Naureen Starling
- Department of Medicine, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | - Michael Hubank
- Clinical Genomics, The Royal Marsden Hospital NHS Foundation Trust, Sutton, United Kingdom
| | - David Cunningham
- Department of Medicine, The Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom.
| |
Collapse
|
22
|
Chen Z, Miao H, Zeng Q, Xu S, Chen Z, Liu K. Circulating cell-free DNA as a diagnostic and prognostic biomarker for non-small-cell lung cancer: a systematic review and meta-analysis. Biomark Med 2019; 14:587-597. [PMID: 31845833 DOI: 10.2217/bmm-2018-0093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim: A meta-analysis was conducted to assess the application of circulating cell-free DNA (cfDNA) in non-small-cell lung carcinoma (NSCLC) screening, EGFR and KRAS mutation detection. Materials & methods: A comprehensive literature search was conducted. The summary sensitivity and specificity for cfDNA in NSCLC diagnosis, EGFR and KRAS mutation detection were calculated. Results: The sensitivity and specificity for NSCLC diagnosis, EGFR and KRAS mutation detection were 0.80 (95% CI: 0.72-0.87) and 0.81 (95% CI: 0.68-0.91), 0.780 (95% CI: 0.711-0.853) and 0.962 (95% CI: 0.942-0.984), 0.628 (95% CI: 0.244-0.919) and 0.959 (95% CI: 0.932-0.998), respectively. Conclusion: cfDNA was a minimally invasive approach for NSCLC diagnosis, but its clinical utility warranted more future investigations because of the suboptimal sensitivity.
Collapse
Affiliation(s)
- Zhoumiao Chen
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qinchun Road, Hangzhou, Zhejiang 310016, China
| | - Huiwen Miao
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qinchun Road, Hangzhou, Zhejiang 310016, China
| | - Qingxin Zeng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qinchun Road, Hangzhou, Zhejiang 310016, China
| | - Shaohua Xu
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qinchun Road, Hangzhou, Zhejiang 310016, China
| | - Zhao Chen
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qinchun Road, Hangzhou, Zhejiang 310016, China
| | - Kai Liu
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qinchun Road, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
23
|
Massihnia D, Pizzutilo EG, Amatu A, Tosi F, Ghezzi S, Bencardino K, Di Masi P, Righetti E, Patelli G, Scaglione F, Vanzulli A, Siena S, Sartore-Bianchi A. Liquid biopsy for rectal cancer: A systematic review. Cancer Treat Rev 2019; 79:101893. [PMID: 31499407 DOI: 10.1016/j.ctrv.2019.101893] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND The management of locally advanced rectal cancer (RC) is an evolving clinical field where the multidisciplinary approach can reach its best, and liquid biopsy for obtaining tumor-derived component such as circulating tumor DNA (ctDNA) might provide complementary informations. METHODS A systematic review of studies available in literature of liquid biopsy in non-metastatic RC has been performed according to PRISMA criteria to assess the role of ctDNA as a diagnostic, predictive and prognostic biomarker in this setting. RESULTS Twenty-five publications have been retrieved, of which 8 full-text articles, 7 abstracts and 10 clinical trials. Results have been categorized into three groups: diagnostic, predictive and prognostic. Few but promising data are available about the use of liquid biopsy for early diagnosis of RC, with the main limitation of sensitivity due to low concentrations of ctDNA in this setting. In terms of prediction of response to chemoradiation, still inconclusive data are available about the utility of a pre-treatment liquid biopsy, whereas some studies report a positive correlation with a dynamic (pre/post-treatment) monitoring. The presence of minimal residual disease by ctDNA was consistently associated with worse prognosis across studies. CONCLUSIONS The use of liquid biopsy for monitoring response to chemoradiation and assess the risk of disease recurrence are the most advanced potential applications for liquid biopsy in RC, with implications also in the context of non-operative management strategies.
Collapse
Affiliation(s)
- Daniela Massihnia
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Elio Gregory Pizzutilo
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Tosi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Silvia Ghezzi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Katia Bencardino
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Pietro Di Masi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Elena Righetti
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Giorgio Patelli
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Francesco Scaglione
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Angelo Vanzulli
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Università degli Studi di Milano, Department of Oncology and Hemato-Oncology, Milan, Italy.
| |
Collapse
|
24
|
Gately L, Wong HL, Tie J, Wong R, Lee M, Lee B, Jalali A, Gibbs P. Emerging strategies in the initial management of locally advanced rectal cancer. Future Oncol 2019; 15:2955-2965. [PMID: 31424262 DOI: 10.2217/fon-2018-0941] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The initial management of locally advanced rectal cancer continues to evolve and formulating the ideal treatment plan remains challenging, with a multitude of emerging treatment strategies and either limited or inconsistent data to support these. The main objective of neoadjuvant treatment is to maximize disease control and minimize toxicity and impact on quality of life. Ultimately, the optimal approach needs to be personalized to the individual. In this Review, we discuss the various strategies currently used and being further investigated in the initial treatment of patients presenting with locally advanced rectal cancer. We describe the evidence behind the current standard of care recommendations and emerging new options, as well as potential biomarkers that may assist with further refining treatment selection.
Collapse
Affiliation(s)
- Lucy Gately
- Personalised Oncology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Hui-Li Wong
- Personalised Oncology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jeanne Tie
- Personalised Oncology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia.,Department of Medical Oncology, Western Health Medical School, University of Melbourne, Footscray, Victoria, Australia
| | - Rachel Wong
- Personalised Oncology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Oncology, Eastern Health, Box Hill, Victoria, Australia.,Eastern Health Clinical School, Monash University, Box Hill, Victoria, Australia
| | - Margaret Lee
- Personalised Oncology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Oncology, Western Health Medical School, University of Melbourne, Footscray, Victoria, Australia.,Department of Medical Oncology, Eastern Health, Box Hill, Victoria, Australia
| | - Belinda Lee
- Personalised Oncology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia.,Department of Medical Oncology, Northern Health, Epping, Victoria, Australia
| | - Azim Jalali
- Personalised Oncology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Gibbs
- Personalised Oncology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Oncology, Western Health Medical School, University of Melbourne, Footscray, Victoria, Australia
| |
Collapse
|
25
|
Christensen E, Birkenkamp-Demtröder K, Sethi H, Shchegrova S, Salari R, Nordentoft I, Wu HT, Knudsen M, Lamy P, Lindskrog SV, Taber A, Balcioglu M, Vang S, Assaf Z, Sharma S, Tin AS, Srinivasan R, Hafez D, Reinert T, Navarro S, Olson A, Ram R, Dashner S, Rabinowitz M, Billings P, Sigurjonsson S, Andersen CL, Swenerton R, Aleshin A, Zimmermann B, Agerbæk M, Lin CHJ, Jensen JB, Dyrskjøt L. Early Detection of Metastatic Relapse and Monitoring of Therapeutic Efficacy by Ultra-Deep Sequencing of Plasma Cell-Free DNA in Patients With Urothelial Bladder Carcinoma. J Clin Oncol 2019; 37:1547-1557. [PMID: 31059311 DOI: 10.1200/jco.18.02052] [Citation(s) in RCA: 342] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Novel sensitive methods for early detection of relapse and for monitoring therapeutic efficacy may have a huge impact on risk stratification, treatment, and ultimately outcome for patients with bladder cancer. We addressed the prognostic and predictive impact of ultra-deep sequencing of cell-free DNA in patients before and after cystectomy and during chemotherapy. PATIENTS AND METHODS We included 68 patients with localized advanced bladder cancer. Patient-specific somatic mutations, identified by whole-exome sequencing, were used to assess circulating tumor DNA (ctDNA) by ultra-deep sequencing (median, 105,000×) of plasma DNA. Plasma samples (n = 656) were procured at diagnosis, during chemotherapy, before cystectomy, and during surveillance. Expression profiling was performed for tumor subtype and immune signature analyses. RESULTS Presence of ctDNA was highly prognostic at diagnosis before chemotherapy (hazard ratio, 29.1; P = .001). After cystectomy, ctDNA analysis correctly identified all patients with metastatic relapse during disease monitoring (100% sensitivity, 98% specificity). A median lead time over radiographic imaging of 96 days was observed. In addition, for high-risk patients (ctDNA positive before or during treatment), the dynamics of ctDNA during chemotherapy was associated with disease recurrence (P = .023), whereas pathologic downstaging was not. Analysis of tumor-centric biomarkers showed that mutational processes (signature 5) were associated with pathologic downstaging (P = .024); however, no significant correlation for tumor subtypes, DNA damage response mutations, and other biomarkers was observed. Our results suggest that ctDNA analysis is better associated with treatment efficacy compared with other available methods. CONCLUSION ctDNA assessment for early risk stratification, therapy monitoring, and early relapse detection in bladder cancer is feasible and provides a basis for clinical studies that evaluate early therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Ann Taber
- 1 Aarhus University Hospital, Aarhus, Denmark
| | | | - Søren Vang
- 1 Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lars Dyrskjøt
- 1 Aarhus University Hospital, Aarhus, Denmark.,3 Aarhus University, Aarhus, Denmark
| |
Collapse
|
26
|
Schou JV, Larsen FO, Sørensen BS, Abrantes R, Boysen AK, Johansen JS, Jensen BV, Nielsen DL, Spindler KL. Circulating cell-free DNA as predictor of treatment failure after neoadjuvant chemo-radiotherapy before surgery in patients with locally advanced rectal cancer. Ann Oncol 2019; 29:610-615. [PMID: 29253083 DOI: 10.1093/annonc/mdx778] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background Treatment of patients with locally advanced rectal cancer (LARC) is based on a combination of chemo-radiotherapy (CRT) and surgery. The rate of distant recurrences remains over 25%. Circulating cell-free DNA (cfDNA) in plasma is a mixture of normal and cancer-specific DNA segments and is a promising biomarker in patients with colorectal cancer. The aim of our study was to investigate plasma cfDNA as a prognostic marker for outcome in patients with LARC treated with neoadjuvant CRT and surgery. Patients and methods In total, 123 patients with LARC were included in 2 biomarker studies. Patients were treated with neoadjuvant CRT before TME surgery. Fifty-two (42%) of the patients received induction chemotherapy with capecitabine + oxaliplatin. Total cfDNA was measured by direct fluorescent assay in EDTA plasma samples obtained at baseline, after induction chemotherapy, and after CRT. Serial samples 5 years after surgery were collected in 51 patients (41%). Results Median follow-up was 55 months. Distant or local recurrence was seen in 30.9% of the patients. Patients with baseline cfDNA levels above the 75th quartile had a higher risk of local or distant recurrence and shorter time to recurrence compared with patients with plasma cfDNA below the 75th percentile (HR = 2.48, 95% CI: 1.3-4.8, P = 0.007). The same applied to disease-free survival (DFS) (HR = 2.43, 95% CI: 1.27-4.7, P = 0.015). In multivariate analysis, a high cfDNA level was significantly associated with time to progression and DFS. During follow-up, the association remained significant regardless of time point for sample analysis. Conclusion We have demonstrated an association between a high baseline plasma level of cfDNA and increased risk of recurrence, shorter time to recurrence, and shorter DFS in patients with LARC. Consequently, cfDNA could potentially improve pre- and post-treatment risk assessment and facilitate individualized therapy for patients with LARC.
Collapse
Affiliation(s)
- J V Schou
- Department of Oncology, Herlev & Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
| | - F O Larsen
- Department of Oncology, Herlev & Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - B S Sørensen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - R Abrantes
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - A K Boysen
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - J S Johansen
- Department of Oncology, Herlev & Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - B V Jensen
- Department of Oncology, Herlev & Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - D L Nielsen
- Department of Oncology, Herlev & Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - K L Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
27
|
Carchman E, Chu DI, Kennedy GD, Morris M, Dakermandji M, Monson JRT, Fernandez LM, Perez RO, Fichera A, Allaix ME, Liska D. SSAT State-of-the-Art Conference: Advances in the Management of Rectal Cancer. J Gastrointest Surg 2019; 23:433-442. [PMID: 30215203 DOI: 10.1007/s11605-018-3965-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/03/2018] [Indexed: 01/31/2023]
Affiliation(s)
- Evie Carchman
- Department of Surgery, Section of Colorectal Surgery, University of Wisconsin- Madison, Madison, WI, USA.
| | - Daniel I Chu
- Department of Surgery, Division of Gastrointestinal Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory D Kennedy
- Department of Surgery, Division of Gastrointestinal Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Melanie Morris
- Department of Surgery, Division of Gastrointestinal Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marc Dakermandji
- Center for Colon & Rectal Surgery, Florida Hospital, Orlando, FL, USA
| | - John R T Monson
- Center for Colon & Rectal Surgery, Florida Hospital, Orlando, FL, USA
| | | | - Rodrigo Oliva Perez
- Angelita & Joaquim Gama Institute, São Paulo, Brazil.,Colorectal Surgery Division, University of São Paulo School of Medicine, São Paulo, Brazil.,Ludwig Institute for Cancer Research, São Paulo Branch, São Paulo, Brazil
| | - Alessandro Fichera
- Department of Surgery, University of North Carolina, Chapel Hill, NC, USA
| | - Marco E Allaix
- Department of Surgical Sciences, University of Torino, Torino, Italy
| | - David Liska
- Department of Colorectal Surgery, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
28
|
Koessler T, Addeo A, Nouspikel T. Implementing circulating tumor DNA analysis in a clinical laboratory: A user manual. Adv Clin Chem 2019; 89:131-188. [PMID: 30797468 DOI: 10.1016/bs.acc.2018.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Liquid biopsy, the analysis of cell-free circulating tumor DNA (ctDNA), is becoming one of the most promising tools in oncology. It has already shown its usefulness in selecting and modulating therapy via remote analysis of the tumor genome and holds important promises in cancer therapy and management, such as assessing the success of key therapeutic steps, monitoring residual disease, early detection of relapses, and establishing prognosis. Yet, ctDNA analysis is technically challenging and its implementation in the laboratory raises multiple strategic and practical issues. As for oncology clinics, integration of this novel test in well-established therapeutic protocols can also pose numerous questions. The current review is intended as a field guide for (1) diagnostic laboratories wishing to implement, validate and possibly accredit ctDNA testing and (2) clinical oncologists interested in integrating the various applications of liquid biopsies in their daily practice. We provide advice and practical recommendations based on our own experience with the technical validations of these methods and on a review of the current literature, with a focus toward gastro-intestinal, lung and breast cancers.
Collapse
Affiliation(s)
- Thibaud Koessler
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Alfredo Addeo
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Thierry Nouspikel
- Service of Medical Genetics, Diagnostics Department, Geneva University Hospital, Geneva, Switzerland.
| |
Collapse
|
29
|
Nardone V, Reginelli A, Scala F, Carbone SF, Mazzei MA, Sebaste L, Carfagno T, Battaglia G, Pastina P, Correale P, Tini P, Pellino G, Cappabianca S, Pirtoli L. Magnetic-Resonance-Imaging Texture Analysis Predicts Early Progression in Rectal Cancer Patients Undergoing Neoadjuvant Chemoradiation. Gastroenterol Res Pract 2019; 2019:8505798. [PMID: 30847005 PMCID: PMC6360039 DOI: 10.1155/2019/8505798] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/05/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND We hypothesized that texture analysis (TA) from the preoperative MRI can predict early disease progression (ePD), defined as the percentage of patients who relapsed or showed distant metastasis within three months from the radical surgery, in patients with locally advanced rectal cancer (LARC, stage II and III, AJCC) undergoing neoadjuvant chemoradiotherapy (C-RT). METHODS This retrospective monoinstitutional cohort study included 49 consecutive patients in total with a newly diagnosed rectal cancer. All the patients underwent baseline abdominal MRI and CT scan of the chest and abdomen to exclude distant metastasis before C-RT. Texture parameters were extracted from MRI performed before C-RT (T1, DWI, and ADC sequences) using LifeX Software, a dedicated software for extracting texture parameters from radiological imaging. We divided the cohort in a training set of 34 patients and a validation set of 15 patients, and we tested the data sets for homogeneity, considering the clinical variables. Then we performed univariate and multivariate analysis, and a ROC curve was also generated. RESULTS Thirteen patients (26.5%) showed an ePD, three of whom with lung metastases and ten with liver relapse. The model was validated based on the prediction accuracy calculated in a previously unseen set of 15 patients. The prediction accuracy of the generated model was 82% (AUC = 0.853) in the training and 80% (AUC = 0.833) in the validation cohort. The only significant features at multivariate analysis was DWI GLCM Correlation (OR: 0.239, p < 0.001). CONCLUSION Our results suggest that TA could be useful to identify patients that may develop early progression.
Collapse
Affiliation(s)
- Valerio Nardone
- Istituto Toscano Tumori, Florence, Italy
- Unit of Radiation Oncology, University Hospital of Siena, Siena, Italy
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Fernando Scala
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | | | | | - Lucio Sebaste
- Istituto Toscano Tumori, Florence, Italy
- Unit of Radiation Oncology, University Hospital of Siena, Siena, Italy
| | - Tommaso Carfagno
- Istituto Toscano Tumori, Florence, Italy
- Unit of Radiation Oncology, University Hospital of Siena, Siena, Italy
| | - Giuseppe Battaglia
- Istituto Toscano Tumori, Florence, Italy
- Unit of Radiation Oncology, University Hospital of Siena, Siena, Italy
| | - Pierpaolo Pastina
- Istituto Toscano Tumori, Florence, Italy
- Unit of Radiation Oncology, University Hospital of Siena, Siena, Italy
| | - Pierpaolo Correale
- Unit of Medical Oncology, Grand Metropolitan Hospital “Bianchi Melacrino Morelli”, Reggio Calabria, Italy
| | - Paolo Tini
- Istituto Toscano Tumori, Florence, Italy
- Unit of Radiation Oncology, University Hospital of Siena, Siena, Italy
- Sbarro Health Research Organization, Temple University, Philadelphia, PA, USA
| | - Gianluca Pellino
- Unit of Colon-Rectal Surgery, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Luigi Pirtoli
- Istituto Toscano Tumori, Florence, Italy
- Unit of Radiation Oncology, University Hospital of Siena, Siena, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| |
Collapse
|
30
|
Circulating tumor DNA detection: A potential tool for colorectal cancer management. Oncol Lett 2018; 17:1409-1416. [PMID: 30675194 PMCID: PMC6341840 DOI: 10.3892/ol.2018.9794] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 08/31/2018] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is frequently diagnosed at an advanced stage of the disease, the pathogenesis of which is influenced by genetic and epigenetic events. Circulating tumor DNA (ctDNA) is extracellular DNA that is present in a number of bodily fluids, including blood, synovial fluid and cerebrospinal fluid. Compared with performing a tissue biopsy, ctDNA examination presents the advantages of minimal invasion and greater convenience. ctDNA is commonly used to identify actionable genomic alterations, monitor treatment responses, unravel therapeutic resistance and potentially detect disease progression prior to clinical and radiological confirmation. The technique can potentially serve as a non-invasive diagnostic tool in personalized medicine, as it demonstrates prognostic value in the management of patients with CRC. ctDNA detection continues to demonstrate inherent advantages compared with other methods, thus serving an increasingly important role in tumor monitoring and oncotherapy. The aim of the current review was to explore the clinical applications of ctDNA in patients with CRC, including early detection and screening, medication guidance, resistance prediction, and residual lesion and recurrence monitoring. Furthermore, several technical methods for ctDNA detection and analysis are explored, as well as other potential biomarkers.
Collapse
|
31
|
Boysen AK, Schou JV, Spindler KLG. Cell-free DNA and preoperative chemoradiotherapy for rectal cancer: a systematic review. Clin Transl Oncol 2018; 21:874-880. [PMID: 30506526 DOI: 10.1007/s12094-018-1997-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/17/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Preoperative chemoradiotherapy is the standard of care for patients with locally advanced rectal cancer, yet valid circulating biomarkers are lacking. We aimed at systematically reviewing the literature of cell-free DNA and locally advanced rectal cancer. METHODS A systematic literature search was performed. We retrieved papers reporting a correlation between a clinical outcome and cell-free DNA for patients receiving chemoradiotherapy for locally advanced rectal cancer. RESULTS We included nine studies of a total of 615 patients. Only single-arm studies were identified, analyzing either the total level of cell-free DNA or tumor-specific DNA. Despite differences in the methodology and outcomes, eight of the nine studies showed a correlation between cell-free DNA and a clinical outcome. CONCLUSIONS Cell-free DNA might hold prognostic and predictive information for patients with locally advanced rectal cancer receiving preoperative chemoradiotherapy; although, firm conclusions are limited by the heterogeneity in this field.
Collapse
Affiliation(s)
- Anders Kindberg Boysen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Nørrebrogade 44, 8000, Aarhus, Denmark. .,Department of Oncology, Aarhus University Hospital, Nørrebrogade 44, 8000, Aarhus, Denmark.
| | - Jakob Vasehus Schou
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| | - Karen-Lise Garm Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Nørrebrogade 44, 8000, Aarhus, Denmark.,Department of Oncology, Aarhus University Hospital, Nørrebrogade 44, 8000, Aarhus, Denmark
| |
Collapse
|
32
|
Vymetalkova V, Cervena K, Bartu L, Vodicka P. Circulating Cell-Free DNA and Colorectal Cancer: A Systematic Review. Int J Mol Sci 2018; 19:ijms19113356. [PMID: 30373199 PMCID: PMC6274807 DOI: 10.3390/ijms19113356] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/22/2018] [Accepted: 10/24/2018] [Indexed: 02/06/2023] Open
Abstract
There is a strong demand for the identification of new biomarkers in colorectal cancer (CRC) diagnosis. Among all liquid biopsy analysts, cell-free circulating DNA (cfDNA) is probably the most promising tool with respect to the identification of minimal residual diseases, assessment of treatment response and prognosis, and identification of resistance mechanisms. Circulating cell-free tumor DNA (ctDNA) maintains the same genomic signatures that are present in the matching tumor tissue allowing for the quantitative and qualitative evaluation of mutation burdens in body fluids. Thus, ctDNA-based research represents a non-invasive method for cancer detection. Among the numerous possible applications, the diagnostic, predictive, and/or prognostic utility of ctDNA in CRC has attracted intense research during the last few years. In the present review, we will describe the different aspects related to cfDNA research and evidence from studies supporting its potential use in CRC diagnoses and the improvement of therapy efficacy. We believe that ctDNA-based research should be considered as key towards the introduction of personalized medicine and patient benefits.
Collapse
Affiliation(s)
- Veronika Vymetalkova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic.
- Institute of Biology and Medical Genetics, 1st Medical Faculty, Charles University, Albertov 4, 128 00 Prague, Czech Republic.
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University in Prague, 323 00 Pilsen, Czech Republic.
| | - Klara Cervena
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic.
- Institute of Biology and Medical Genetics, 1st Medical Faculty, Charles University, Albertov 4, 128 00 Prague, Czech Republic.
| | - Linda Bartu
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic.
- Institute of Biology and Medical Genetics, 1st Medical Faculty, Charles University, Albertov 4, 128 00 Prague, Czech Republic.
| | - Pavel Vodicka
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic.
- Institute of Biology and Medical Genetics, 1st Medical Faculty, Charles University, Albertov 4, 128 00 Prague, Czech Republic.
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University in Prague, 323 00 Pilsen, Czech Republic.
| |
Collapse
|
33
|
Timmerman C, Taveras LR, Huerta S. Clinical and molecular diagnosis of pathologic complete response in rectal cancer: an update. Expert Rev Mol Diagn 2018; 18:887-896. [PMID: 30124091 DOI: 10.1080/14737159.2018.1514258] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The standard of care for locally advanced rectal cancer includes neoadjuvant chemoradiation with subsequent total mesorectal excision. This approach has shown various degrees of response to neoadjuvant chemoradiation (ranging from complete response to further tumor growth), which have substantial prognostic and therapeutic implications. A total regression of the tumor is a predictor of superior oncologic outcomes compared with partial responders and non-responders. Further, this concept has opened the possibility of nonoperative strategies for complete responders and explains the widespread research interest in finding clinical, radiographic, pathologic, and biochemical parameters that allow for identification of these patients. Areas covered: The present review evaluates the most recent efforts in the literature to identify predictors of patients likely to achieve a complete response following neoadjuvant treatment for the management of rectal cancer. This includes clinical predictors of pathologic complete response such as tumor location, size, and stage, molecular predictors such as tumor biology and microRNA, serum biomarkers such as carcinoembryogenic antigen and nomograms. Expert commentary: There has been significant progress in our ability to predict pathological complete response. However, more high-quality research is still needed to use this concept to confidently dictate clinical management.
Collapse
Affiliation(s)
- Corey Timmerman
- a University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Luis R Taveras
- a University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Sergio Huerta
- a University of Texas Southwestern Medical Center , Dallas , TX , USA.,b VA North Texas Healthcare System , Dallas , TX , USA
| |
Collapse
|
34
|
Yang YC, Wang D, Jin L, Yao HW, Zhang JH, Wang J, Zhao XM, Shen CY, Chen W, Wang XL, Shi R, Chen SY, Zhang ZT. Circulating tumor DNA detectable in early- and late-stage colorectal cancer patients. Biosci Rep 2018; 38:BSR20180322. [PMID: 29914973 PMCID: PMC6066652 DOI: 10.1042/bsr20180322] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/13/2018] [Accepted: 06/18/2018] [Indexed: 12/15/2022] Open
Abstract
Characterization, diagnosis, and treatment of colorectal cancers (CRC) is difficult due to limited biopsy information, impracticality of repeated biopsies, and cancer biomarker fallibility. Circulating tumor DNA (ctDNA) has recently been investigated as a non-invasive way to gain representative gene mutations in tumors, in addition to monitoring disease progression and response to treatment. We analyzed ctDNA mutations and concentrations in 47 early- and late-stage CRC patients using a targetted sequencing approach using a panel that covers 50 cancer-related genes. ctDNA mutations in 37 genes were identified in 93.6% of the patients (n=47). The results showed that TP53, PIK3CA, APC, and EGFR were the most frequently mutated genes. Stage IV patients had significantly higher ctDNA concentration than Stage I patients, and increased ctDNA concentration correlated with increased tumor size. Additionally, ctDNA detection was found to be a greater predictor of disease when compared with five known commonly used tumor biomarkers. The present study supports the use of ctDNA as a liquid biopsy to gain clinical tumor information that may facilitate early diagnosis and treatment and improve CRC patient prognosis.
Collapse
Affiliation(s)
- Ying-Chi Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Dong Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Lan Jin
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Hong-Wei Yao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Jing-Hui Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Jin Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Xiao-Mu Zhao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| | - Chun-Ying Shen
- San Valley Biotechnology Incorporated, Beijing 100094, China
| | - Wei Chen
- San Valley Biotechnology Incorporated, Beijing 100094, China
| | - Xue-Liang Wang
- San Valley Biotechnology Incorporated, Beijing 100094, China
| | - Rong Shi
- San Valley Biotechnology Incorporated, Beijing 100094, China
| | - Si-Yi Chen
- Departments of Molecular Microbiology and Immunology and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, U.S.A
| | - Zhong-Tao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, China
| |
Collapse
|
35
|
Perez RO, Habr-Gama A. Putting down the scalpel in rectal cancer management - a historical perspective. Colorectal Dis 2018; 20 Suppl 1:12-15. [PMID: 29878677 DOI: 10.1111/codi.14070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The surgical management of rectal cancer has evolved from a disease without any possibility of cure in the early 1700s where surgical management consisted of the palliative drainage of disease related abscesses to the present day where surgical cure is not only possible but also possible with sphincter or even organ preservation. Prof Habr-Gama's lecture describes the evolution of the surgical management of rectal cancer and the current focus on organ preservation.
Collapse
Affiliation(s)
- R O Perez
- Angelita and Joaquim Gama Institute, São Paulo, Brazil
- Colorectal Surgery Division, School of Medicine, University of São Paulo, São Paulo, Brazil
- Ludwig Institute for Cancer Research, São Paulo, Brazil
- Gastrointestinal Surgical Oncology Division, Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| | - A Habr-Gama
- Angelita and Joaquim Gama Institute, São Paulo, Brazil
- Colorectal Surgery Division, School of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
The role of circulating tumour cells and nucleic acids in blood for the detection of bladder cancer: A systematic review. Cancer Treat Rev 2018; 66:56-63. [PMID: 29684744 DOI: 10.1016/j.ctrv.2018.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Blood-based biomarkers are a neglected resource in bladder cancer, where the mainstay of focus has been on urinary biomarkers. However, blood-based biomarkers are gaining popularity in other solid cancers, particularly circulating tumour cells (CTCs) and circulating nucleic acids. In this systematic review, we identify and discuss the diagnostic value of CTC, cell-free DNA and RNA based biomarkers in bladder cancer. METHODS A MEDLINE/Pubmed systematic search was performed using the following keywords: (bladder cancer) AND (blood OR plasma OR serum) AND biomarker AND (DNA OR RNA OR cfDNA OR cell-free DNA OR RNA OR CTC). All studies including blood-based biomarkers based on DNA, RNA and CTCs were reviewed. Of the included studies, studies reporting sensitivity, specificity and/or AUC/ROC values were further described. RESULTS Systematic searched yielded 47 studies that were eligible, of which 21, 19 and 3 studies reported DNA, RNA and CTC biomarkers respectively. 15 of these studies included sensitivity, specificity and/or AUC/ROC values. Biomarkers sensitivity and specificity ranged widely at 2.4-97.6% and 43.3-100% respectively. Median number of patients recruited in the studies was 56 (IQR 41-90). Only 3 studies included an independent validation cohort. The highest sensitivity and specificity pairing achieved in the validation cohort was 80.0% and 89.1% respectively. CONCLUSIONS This systematic review provides a comprehensive overview of the blood-based CTC and nucleic acid biomarkers that have been investigated. An overlap in interest of targets between studies suggests that these could be promising biomarkers, but few biomarkers achieve high sensitivity and specificity, and fewer still have been validated independently.
Collapse
|
37
|
Taveras LR, Cunningham HB, Imran JB. Can We Reliably Predict a Clinical Complete Response in Rectal Cancer? Current Trends and Future Strategies. CURRENT COLORECTAL CANCER REPORTS 2018. [DOI: 10.1007/s11888-018-0401-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
38
|
Birkman EM, Elzagheid A, Jokilehto T, Avoranta T, Korkeila E, Kulmala J, Syrjänen K, Westermarck J, Sundström J. Protein phosphatase 2A (PP2A) inhibitor CIP2A indicates resistance to radiotherapy in rectal cancer. Cancer Med 2018; 7:698-706. [PMID: 29441695 PMCID: PMC5852361 DOI: 10.1002/cam4.1361] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/30/2017] [Indexed: 12/17/2022] Open
Abstract
Preoperative (chemo)radiotherapy, (C)RT, is an essential part of the treatment of rectal cancer patients, but tumor response to this therapy among patients is variable. Thus far, there are no clinical biomarkers that could be used to predict response to (C)RT or to stratify patients into different preoperative treatment groups according to their prognosis. Overexpression of cancerous inhibitor of protein phosphatase 2A (CIP2A) has been demonstrated in several cancers and is frequently associated with reduced survival. Recently, high CIP2A expression has also been indicated to contribute to radioresistance in head and neck squamous cell carcinoma, but few studies have examined the connection between CIP2A and radiation response regarding other malignancies. We have evaluated CIP2A protein expression levels in relation to tumor regression after preoperative (C)RT and survival of rectal adenocarcinoma patients. The effects of CIP2A knockdown by siRNA on cell survival were further investigated in colorectal cancer cells exposed to radiation. Patients with low‐CIP2A‐expressing tumors had more frequently moderate or excellent response to long‐course (C)RT than patients with high‐CIP2A‐expressing tumors. They also had higher 36‐month disease‐specific survival (DSS) rate in categorical analysis. In the multivariate analysis, low CIP2A expression level remained as an independent predictive factor for increased DSS. Suppression of CIP2A transcription by siRNA was found to sensitize colorectal cancer cells to irradiation and decrease their survival in vitro. In conclusion, these results suggest that by contributing to radiosensitivity of cancer cells, low CIP2A protein expression level associates with a favorable response to long‐course (C)RT in rectal cancer patients.
Collapse
Affiliation(s)
- Eva-Maria Birkman
- Department of Pathology, University of Turku, Turku, Finland.,Department of Pathology, Turku University Hospital, Turku, Finland
| | - Adam Elzagheid
- Department of Pathology, Faculty of Medicine, Benghazi University, Benghazi, Libya.,Department of Genetic Engineering, Biotechnology Research Center, Tripoli, Libya
| | - Terhi Jokilehto
- Department of Pathology, University of Turku, Turku, Finland.,Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Tuulia Avoranta
- Department of Pathology, University of Turku, Turku, Finland.,Department of Oncology, University of Turku and Turku University Hospital, Turku, Finland
| | - Eija Korkeila
- Department of Oncology, University of Turku and Turku University Hospital, Turku, Finland
| | - Jarmo Kulmala
- Department of Oncology, University of Turku and Turku University Hospital, Turku, Finland
| | - Kari Syrjänen
- Department of Clinical Research, Biohit Oyj, Helsinki, Finland.,Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Jukka Westermarck
- Department of Pathology, University of Turku, Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jari Sundström
- Department of Pathology, University of Turku, Turku, Finland.,Department of Pathology, Turku University Hospital, Turku, Finland
| |
Collapse
|
39
|
Khakoo S, Georgiou A, Gerlinger M, Cunningham D, Starling N. Circulating tumour DNA, a promising biomarker for the management of colorectal cancer. Crit Rev Oncol Hematol 2018; 122:72-82. [PMID: 29458792 DOI: 10.1016/j.critrevonc.2017.12.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/18/2017] [Accepted: 12/06/2017] [Indexed: 02/08/2023] Open
Abstract
Circulating cell free tumour DNA (ctDNA) maintains the same genomic alterations that are present in the corresponding tumour, thereby allowing for quantitative and qualitative real-time evaluation in body fluids as an alternative to onerous repeat biopsies. Improvements in the sensitivity of techniques used to identify ctDNA has led to a surge of research investigating its role in the detection of: early disease, relapse, response to therapy and emerging drug resistance mechanisms. Following curative surgery, ctDNA detection is a promising marker of minimal residual disease and could better select patients for adjuvant chemotherapy. Longitudinal monitoring could help identify early relapse. In metastatic disease, ctDNA can predict response to chemotherapy prior to evidence of disease progression on imaging and investigate novel primary and acquired resistance mechanisms to targeted therapies. More experience in detecting, analysing and interpreting ctDNA within prospective trials, will better define its role for implementation into routine clinical practice.
Collapse
Affiliation(s)
- Shelize Khakoo
- Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Alexandros Georgiou
- Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom; Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Marco Gerlinger
- Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom; Centre of Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - David Cunningham
- Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom
| | - Naureen Starling
- Department of Medicine, Royal Marsden Hospital, London and Surrey, United Kingdom.
| |
Collapse
|
40
|
Sclafani F, Chau I, Cunningham D, Hahne JC, Vlachogiannis G, Eltahir Z, Lampis A, Braconi C, Kalaitzaki E, De Castro DG, Wotherspoon A, Capdevila J, Glimelius B, Tarazona N, Begum R, Lote H, Hulkki Wilson S, Mentrasti G, Brown G, Tait D, Oates J, Valeri N. KRAS and BRAF mutations in circulating tumour DNA from locally advanced rectal cancer. Sci Rep 2018; 8:1445. [PMID: 29362371 PMCID: PMC5780472 DOI: 10.1038/s41598-018-19212-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022] Open
Abstract
There are limited data on circulating, cell-free, tumour (ct)DNA analysis in locally advanced rectal cancer (LARC). Digital droplet (dd)PCR was used to investigate KRAS/BRAF mutations in ctDNA from baseline blood samples of 97 LARC patients who were treated with CAPOX followed by chemoradiotherapy, surgery and adjuvant CAPOX ± cetuximab in a randomised phase II trial. KRAS mutation in G12D, G12V or G13D was detected in the ctDNA of 43% and 35% of patients with tumours that were mutant and wild-type for these hotspot mutations, respectively, according to standard PCR-based analyses on tissue. The detection rate in the ctDNA of 10 patients with less common mutations was 50%. In 26 cases ctDNA analysis revealed KRAS mutations that were not previously found in tissue. Twenty-two of these (84.6%) were detected following repeat tissue testing by ddPCR. Overall, the ctDNA detection rate in the KRAS mutant population was 66%. Detection of KRAS mutation in ctDNA failed to predict prognosis or refine patient selection for cetuximab. While this study confirms the feasibility of ctDNA analysis in LARC and the high sensitivity of ddPCR, larger series are needed to better address the role of ctDNA as a prognostic or predictive tool in this setting.
Collapse
Affiliation(s)
- Francesco Sclafani
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Ian Chau
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - David Cunningham
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Jens C Hahne
- The Institute of Cancer Research, London and Surrey, United Kingdom
| | | | - Zakaria Eltahir
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Andrea Lampis
- The Institute of Cancer Research, London and Surrey, United Kingdom
| | - Chiara Braconi
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
- The Institute of Cancer Research, London and Surrey, United Kingdom
| | | | | | - Andrew Wotherspoon
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Jaume Capdevila
- Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Noelia Tarazona
- Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - Ruwaida Begum
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Hazel Lote
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
- The Institute of Cancer Research, London and Surrey, United Kingdom
| | | | - Giulia Mentrasti
- The Institute of Cancer Research, London and Surrey, United Kingdom
| | - Gina Brown
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Diana Tait
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Jacqueline Oates
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Nicola Valeri
- The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom.
- The Institute of Cancer Research, London and Surrey, United Kingdom.
| |
Collapse
|
41
|
Ai B, Liu H, Huang Y, Peng P. Circulating cell-free DNA as a prognostic and predictive biomarker in non-small cell lung cancer. Oncotarget 2018; 7:44583-44595. [PMID: 27323821 PMCID: PMC5190120 DOI: 10.18632/oncotarget.10069] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 05/29/2016] [Indexed: 01/15/2023] Open
Abstract
Circulating cell-free DNA (cfDNA), which can be obtained from plasma or serum by non-invasive procedures, has showed great potential to predict treatment response and survival for cancer patients. Several studies have assessed the prognostic and predictive value of cfDNA in non-small cell lung cancer (NSCLC). However, these studies were often small and reported varying results. To address this issue, a meta-analysis was carried out. A total of 22 studies involving 2518 patients were subjected to the final analysis. Our results indicated that NSCLC patients with higher cfDNA concentration had shorter median progression-free survival (PFS) and overall survival (OS) time. In addition, high levels of cfDNA were significantly associated with poor PFS (hazard ratio or HR, 1.32; 95% CI, 1.02-1.71) and OS (HR, 1.64; 95% CI, 1.26-2.15). With respect to tumor specific mutations, we failed to reveal significant differences for PFS (HR, 1.30; 95% CI, 0.66-2.56) and OS (HR, 1.05; 95% CI, 0.49-2.25) when NSCLC patients were grouped according to KRAS genotype detected in cfDNA. However, NSCLC patients which harbored EGFR activating mutation in cfDNA had a greater chance of response to EGFR-TKIs (odds ratio or OR, 1.96; 95% CI, 1.59-2.42). No significant publication bias was detected in this study. In conclusion, cfDNA could act as a prognostic and predictive biomarker for patients with NSCLC.
Collapse
Affiliation(s)
- Bo Ai
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan 430030, People's Republic of China
| | - Huiquan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan 430030, People's Republic of China
| | - Yu Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan 430030, People's Republic of China
| | - Ping Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan 430030, People's Republic of China
| |
Collapse
|
42
|
Mamoshina P, Ojomoko L, Yanovich Y, Ostrovski A, Botezatu A, Prikhodko P, Izumchenko E, Aliper A, Romantsov K, Zhebrak A, Ogu IO, Zhavoronkov A. Converging blockchain and next-generation artificial intelligence technologies to decentralize and accelerate biomedical research and healthcare. Oncotarget 2017; 9:5665-5690. [PMID: 29464026 PMCID: PMC5814166 DOI: 10.18632/oncotarget.22345] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/02/2017] [Indexed: 12/19/2022] Open
Abstract
The increased availability of data and recent advancements in artificial intelligence present the unprecedented opportunities in healthcare and major challenges for the patients, developers, providers and regulators. The novel deep learning and transfer learning techniques are turning any data about the person into medical data transforming simple facial pictures and videos into powerful sources of data for predictive analytics. Presently, the patients do not have control over the access privileges to their medical records and remain unaware of the true value of the data they have. In this paper, we provide an overview of the next-generation artificial intelligence and blockchain technologies and present innovative solutions that may be used to accelerate the biomedical research and enable patients with new tools to control and profit from their personal data as well with the incentives to undergo constant health monitoring. We introduce new concepts to appraise and evaluate personal records, including the combination-, time- and relationship-value of the data. We also present a roadmap for a blockchain-enabled decentralized personal health data ecosystem to enable novel approaches for drug discovery, biomarker development, and preventative healthcare. A secure and transparent distributed personal data marketplace utilizing blockchain and deep learning technologies may be able to resolve the challenges faced by the regulators and return the control over personal data including medical records back to the individuals.
Collapse
Affiliation(s)
- Polina Mamoshina
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA.,Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Lucy Ojomoko
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | | | | | | | | | - Eugene Izumchenko
- Department of Otolaryngology-Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander Aliper
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | - Konstantin Romantsov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | - Alexander Zhebrak
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA
| | - Iraneus Obioma Ogu
- Africa Blockchain Artificial Intelligence for Healthcare Initiative, Insilico Medicine, Inc, Abuja, Nigeria
| | - Alex Zhavoronkov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, Maryland, USA.,The Biogerontology Research Foundation, London, United Kingdom
| |
Collapse
|
43
|
Chang Y, Tolani B, Nie X, Zhi X, Hu M, He B. Review of the clinical applications and technological advances of circulating tumor DNA in cancer monitoring. Ther Clin Risk Manag 2017; 13:1363-1374. [PMID: 29066904 PMCID: PMC5644666 DOI: 10.2147/tcrm.s141991] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Circulating cell-free DNA (cfDNA) released by tumor cells, termed ctDNA, closely reflects the heterogeneity of primary cancers and their metastases. As a noninvasive, real-time monitoring biomarker, ctDNA is a promising tool for detecting driver gene mutations, assessing tumor burden and acquired resistance, and early diagnosis. However, isolation and enrichment of cfDNA is a big challenge due to the high degree of DNA fragmentation and its relatively low abundance in the bloodstream. This review aims to provide insights into the recent technological advances in acquisition of optimal quality cfDNA, the use of preservatives, isolation methods, processing timelines, and detection techniques. It also describes clinical applications of ctDNA in cancer patient management.
Collapse
Affiliation(s)
- Yi Chang
- Department of Respiratory Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Bhairavi Tolani
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Xiuhong Nie
- Department of Respiratory Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiuyi Zhi
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Mu Hu
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Biao He
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
44
|
Pereira AAL, Morelli MP, Overman M, Kee B, Fogelman D, Vilar E, Shureiqi I, Raghav K, Eng C, Manuel S, Crosby S, Wolff RA, Banks K, Lanman R, Talasaz A, Kopetz S, Morris V. Clinical utility of circulating cell-free DNA in advanced colorectal cancer. PLoS One 2017; 12:e0183949. [PMID: 28850629 PMCID: PMC5574560 DOI: 10.1371/journal.pone.0183949] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/15/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Circulating cell-free DNA (cfDNA) isolated from the plasma of cancer patients (pts) has been shown to reflect the genomic mutation profile of the tumor. However, physician and patient assessment of clinical utility of these assays in patients with metastatic colorectal cancer (mCRC) has not been previously described. METHODS Patients were prospectively consented to a prospective genomic matching protocol (Assessment of Targeted Therapies Against Colorectal Cancer [ATTACC]), with collection of blood for cfDNA extraction and sequencing of a 54-gene panel in a CLIA-certified lab. Formalin-fixed, paraffin-embedded (FFPE) tissue from prior resections or biopsies underwent 50-gene sequencing. Results from both assays were returned to the treating physicians for patient care and clinical trial selection. Follow-up surveys of treating physicians and chart reviews assessed clinical utility. RESULTS 128 mCRC pts were enrolled between 6/2014 and 1/2015. Results were returned in median of 13 and 26 days for cfDNA and FFPE sequencing, respectively. With cfDNA sequencing, 78% (100/128) of samples had a detectable somatic genomic alteration. 50% of cfDNA cases had potentially actionable alterations, and 60% of these could be genomically matched to at least one clinical trial in our institution. 50% (15/30) of these pts enrolled onto an identified matched trial. Physicians reported that the cfDNA testing improved the quality of care they could provide in 73% of the cases, and that 89% of pts reported greater satisfaction with the efforts to personalize experimental therapeutic agents. CONCLUSIONS cfDNA sequencing can provide timely information on potentially actionable mutations and amplifications, thereby facilitating clinical trial enrollment and improving the perceived quality of care.
Collapse
Affiliation(s)
- Allan A. Lima Pereira
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Maria Pia Morelli
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Michael Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - David Fogelman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Imad Shureiqi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Cathy Eng
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Shanequa Manuel
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Shadarra Crosby
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Kimberly Banks
- Guardant Health, Redwood City, California, United States of America
| | - Richard Lanman
- Guardant Health, Redwood City, California, United States of America
| | - AmirAli Talasaz
- Guardant Health, Redwood City, California, United States of America
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Van Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
45
|
Organ preservation in rectal cancer - Challenges and future strategies. Clin Transl Radiat Oncol 2017; 3:9-15. [PMID: 29658007 PMCID: PMC5893528 DOI: 10.1016/j.ctro.2017.02.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 02/16/2017] [Indexed: 12/14/2022] Open
Abstract
Neoadjuvant radiochemotherapy with subsequent total mesorectal excision is the standard of care for locally advanced rectal cancer. While this multimodal strategy has decreased local recurrences rates below 5%, long-term morbidities are considerable in terms of urinary, sexual or bowel functioning. At the same time approximately 10–20% of patients have no evidence of residual tumour in their surgical specimen. Pioneering studies from Brazil have suggested that surgery can safely be omitted in carefully selected patients with a clinical complete response after radiochemotherapy. Although confirmatory studies showed similar results, challenges in terms of optimizing radiochemotherapy for organ-preservation, appropriate selection of patients for non-operative management and the safety of this approach remain. The present review will summarize the current data on organ-preservation in rectal cancer and discuss the challenges that need to be addressed in future trials.
Collapse
|
46
|
Rapisuwon S, Vietsch EE, Wellstein A. Circulating biomarkers to monitor cancer progression and treatment. Comput Struct Biotechnol J 2016; 14:211-22. [PMID: 27358717 PMCID: PMC4913179 DOI: 10.1016/j.csbj.2016.05.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/11/2022] Open
Abstract
Tumor heterogeneity is a major challenge and the root cause of resistance to treatment. Still, the standard diagnostic approach relies on the analysis of a single tumor sample from a local or metastatic site that is obtained at a given time point. Due to intratumoral heterogeneity and selection of subpopulations in diverse lesions this will provide only a limited characterization of the makeup of the disease. On the other hand, recent developments of nucleic acid sequence analysis allows to use minimally invasive serial blood samples to assess the mutational status and altered gene expression patterns for real time monitoring in individual patients. Here, we focus on cell-free circulating tumor-specific mutant DNA and RNA (including mRNA and non-coding RNA), as well as current limitations and challenges associated with circulating nucleic acids biomarkers.
Collapse
Affiliation(s)
| | | | - Anton Wellstein
- Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd, NW, Washington, DC 20007, USA
| |
Collapse
|
47
|
Eke I, Makinde AY, Aryankalayil MJ, Ahmed MM, Coleman CN. Comprehensive molecular tumor profiling in radiation oncology: How it could be used for precision medicine. Cancer Lett 2016; 382:118-126. [PMID: 26828133 DOI: 10.1016/j.canlet.2016.01.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 12/16/2022]
Abstract
New technologies enabling the analysis of various molecules, including DNA, RNA, proteins and small metabolites, can aid in understanding the complex molecular processes in cancer cells. In particular, for the use of novel targeted therapeutics, elucidation of the mechanisms leading to cell death or survival is crucial to eliminate tumor resistance and optimize therapeutic efficacy. While some techniques, such as genomic analysis for identifying specific gene mutations or epigenetic testing of promoter methylation, are already in clinical use, other "omics-based" assays are still evolving. Here, we provide an overview of the current status of molecular profiling methods, including promising research strategies, as well as possible challenges, and their emerging role in radiation oncology.
Collapse
Affiliation(s)
- Iris Eke
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Adeola Y Makinde
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mansoor M Ahmed
- Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| |
Collapse
|