1
|
Domingo E, Rathee S, Blake A, Samuel L, Murray G, Sebag-Montefiore D, Gollins S, West N, Begum R, Richman S, Quirke P, Redmond K, Chatzipli A, Barberis A, Hassanieh S, Mahmood U, Youdell M, McDermott U, Koelzer V, Leedham S, Tomlinson I, Dunne P, Buffa FM, Maughan TS. Identification and validation of a machine learning model of complete response to radiation in rectal cancer reveals immune infiltrate and TGFβ as key predictors. EBioMedicine 2024; 106:105228. [PMID: 39013324 DOI: 10.1016/j.ebiom.2024.105228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND It is uncertain which biological features underpin the response of rectal cancer (RC) to radiotherapy. No biomarker is currently in clinical use to select patients for treatment modifications. METHODS We identified two cohorts of patients (total N = 249) with RC treated with neoadjuvant radiotherapy (45Gy/25) plus fluoropyrimidine. This discovery set included 57 cases with pathological complete response (pCR) to chemoradiotherapy (23%). Pre-treatment cancer biopsies were assessed using transcriptome-wide mRNA expression and targeted DNA sequencing for copy number and driver mutations. Biological candidate and machine learning (ML) approaches were used to identify predictors of pCR to radiotherapy independent of tumour stage. Findings were assessed in 107 cases from an independent validation set (GSE87211). FINDINGS Three gene expression sets showed significant independent associations with pCR: Fibroblast-TGFβ Response Signature (F-TBRS) with radioresistance; and cytotoxic lymphocyte (CL) expression signature and consensus molecular subtype CMS1 with radiosensitivity. These associations were replicated in the validation cohort. In parallel, a gradient boosting machine model comprising the expression of 33 genes generated in the discovery cohort showed high performance in GSE87211 with 90% sensitivity, 86% specificity. Biological and ML signatures indicated similar mechanisms underlying radiation response, and showed better AUC and p-values than published transcriptomic signatures of radiation response in RC. INTERPRETATION RCs responding completely to chemoradiotherapy (CRT) have biological characteristics of immune response and absence of immune inhibitory TGFβ signalling. These tumours may be identified with a potential biomarker based on a 33 gene expression signature. This could help select patients likely to respond to treatment with a primary radiotherapy approach as for anal cancer. Conversely, those with predicted radioresistance may be candidates for clinical trials evaluating addition of immune-oncology agents and stromal TGFβ signalling inhibition. FUNDING The Stratification in Colorectal Cancer Consortium (S:CORT) was funded by the Medical Research Council and Cancer Research UK (MR/M016587/1).
Collapse
Affiliation(s)
- Enric Domingo
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Sanjay Rathee
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Andrew Blake
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Leslie Samuel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Graeme Murray
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | | | - Simon Gollins
- North Wales Cancer Treatment Centre, Besti Cadwaladr University Health Board, Bodelwyddan, Denbighshire, LL18 5UJ, UK
| | - Nicholas West
- Leeds Institute of Medical Research, University of Leeds, LS9 7TF, UK
| | - Rubina Begum
- Cancer Research & University College London Clinica Trial Unit, London, United Kingdom
| | - Susan Richman
- Leeds Institute of Medical Research, University of Leeds, LS9 7TF, UK
| | - Phil Quirke
- Leeds Institute of Medical Research, University of Leeds, LS9 7TF, UK
| | - Keara Redmond
- The Patrick G Johnston Centre for Cancer Research, Queens University Belfast, Belfast, BT7 9AE, UK
| | | | - Alessandro Barberis
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Sylvana Hassanieh
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Umair Mahmood
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Michael Youdell
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | | | - Viktor Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Oncology and Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Simon Leedham
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, UK
| | - Ian Tomlinson
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Philip Dunne
- The Patrick G Johnston Centre for Cancer Research, Queens University Belfast, Belfast, BT7 9AE, UK
| | - Francesca M Buffa
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK; Department of Computing Sciences, Bocconi University, Bocconi Institute for Data Science and Analytics (BIDSA), Milano, Italy.
| | - Timothy S Maughan
- Department of Oncology, Medical Sciences Division, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK; Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
2
|
Afshar S, Leili T, Amini P, Dinu I. Introducing novel key genes and transcription factors associated with rectal cancer response to chemoradiation through co-expression network analysis. Heliyon 2023; 9:e18869. [PMID: 37636389 PMCID: PMC10447927 DOI: 10.1016/j.heliyon.2023.e18869] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/16/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Preoperative radiochemotherapy is a promising therapeutic method for locally advanced rectal cancer patients. However, the response of colorectal cancer (CRC) patients to preoperative radiotherapy varies widely. In this study, we aimed to identify novel biomarkers that could predict the response of colorectal tumors to treatment using a systems biology approach. We applied the Weighted Gene Co-Expression Network Analysis to construct co-expression networks and evaluated the correlation of these networks with radiation using the module-trait relationship. We then identified hub genes and related transcription factors in the selected co-expression module. Our analysis of seven constructed modules revealed that one module, which contained 113 nodes and 6066 edges, had the strongest correlation with radiation effects on CRC (correlation = 0.85; p-value = 6e-7). By analyzing the selected module with the CytoHubba plugin, we identified four hub genes, including ZEB2, JAM2, NDN, and PPAP2A. We also identified seven important transcription factors, including KLF4, SUZ12, TCF4, NANOG, POU5F1, SOX2, and SMARCA4, which may play essential roles in regulating the four hub genes. In summary, our findings suggest that ZEB2, JAM2, NDN, and PPAP2A, along with the seven transcription factors related to these hub genes, may be associated with the response of colorectal tumors to chemoradiotherapy.
Collapse
Affiliation(s)
- Saeid Afshar
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Tapak Leili
- Department of Biostatistics, School of Public Health and Modeling of Noncommunicable Diseases Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Payam Amini
- School of Medicine, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Irina Dinu
- School of Public Health, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
3
|
Jeon SH, Chie EK. Characterization of the gene signature correlated with favorable response to chemoradiotherapy in rectal cancer: A hypothesis-generating study. Cancer Med 2023; 12:8981-8990. [PMID: 36621808 PMCID: PMC10134325 DOI: 10.1002/cam4.5586] [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: 09/23/2022] [Revised: 11/17/2022] [Accepted: 12/21/2022] [Indexed: 01/10/2023] Open
Abstract
PURPOSE This study aimed to define the gene signature associated with response to neoadjuvant chemoradiotherapy (nCRT), or chemoradiosensitivity (CRS) signature, in rectal cancer, and investigate the correlation between the CRS signature and characteristics of tumor. MATERIALS AND METHODS Three public microarray datasets of pre-nCRT rectal cancer were used to discover and validate the CRS signature, and the pathway analysis of the CRS signature was performed. Patients in The Cancer Genome Atlas (TCGA) dataset were stratified according to the CRS signature enrichment score, and mutational profile and proportions of infiltrated immune cells were compared. RESULTS In the discovery dataset (GSE53781), 95 genes were upregulated in complete responders compared to non-complete responders and defined as the CRS signature. Pathways regarding DNA replication and repair processes as well as inflammatory response were enriched in the CRS signature. In the validation datasets (GSE35452 and GSE45404), patients with favorable response to nCRT exhibited higher enrichment score of the CRS. In TCGA-READ cohort, patients with high CRS signature harbored KRAS mutation in lower frequency than those with low CRS signature. In addition, proportions of proinflammatory immune cells were higher, but proportion of immunosuppressive M2 macrophages was lower in patients with high CRS signature than those with low CRS signature. CONCLUSIONS The current integrative bioinformatic analysis suggests the CRS signature and showed that the CRS signature is associated with dissimilar mutational profile and increased immune response. The discovered CRS signature and related characteristics may serve as candidate of stratification factor in upcoming studies for rectal cancer.
Collapse
Affiliation(s)
- Seung Hyuck Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Eui Kyu Chie
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Emons G, Auslander N, Jo P, Kitz J, Azizian A, Hu Y, Hess CF, Roedel C, Sax U, Salinas G, Stroebel P, Kramer F, Beissbarth T, Grade M, Ghadimi M, Ruppin E, Ried T, Gaedcke J. Gene-expression profiles of pretreatment biopsies predict complete response of rectal cancer patients to preoperative chemoradiotherapy. Br J Cancer 2022; 127:766-775. [PMID: 35597871 PMCID: PMC9381580 DOI: 10.1038/s41416-022-01842-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/19/2022] [Accepted: 05/04/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Preoperative (neoadjuvant) chemoradiotherapy (CRT) and total mesorectal excision is the standard treatment for rectal cancer patients (UICC stage II/III). Up to one-third of patients treated with CRT achieve a pathological complete response (pCR). These patients could be spared from surgery and its associated morbidity and mortality, and assigned to a "watch and wait" strategy. However, reliably identifying pCR based on clinical or imaging parameters remains challenging. EXPERIMENTAL DESIGN We generated gene-expression profiles of 175 patients with locally advanced rectal cancer enrolled in the CAO/ARO/AIO-94 and -04 trials. One hundred and sixty-one samples were used for building, training and validating a predictor of pCR using a machine learning algorithm. The performance of the classifier was validated in three independent cohorts, comprising 76 patients from (i) the CAO/ARO/AIO-94 and -04 trials (n = 14), (ii) a publicly available dataset (n = 38) and (iii) in 24 prospectively collected samples from the TransValid A trial. RESULTS A 21-transcript signature yielded the best classification of pCR in 161 patients (Sensitivity: 0.31; AUC: 0.81), when not allowing misclassification of non-complete-responders (False-positive rate = 0). The classifier remained robust when applied to three independent datasets (n = 76). CONCLUSION The classifier can identify >1/3 of rectal cancer patients with a pCR while never classifying patients with an incomplete response as having pCR. Importantly, we could validate this finding in three independent datasets, including a prospectively collected cohort. Therefore, this classifier could help select rectal cancer patients for a "watch and wait" strategy. TRANSLATIONAL RELEVANCE Forgoing surgery with its associated side effects could be an option for rectal cancer patients if the prediction of a pathological complete response (pCR) after preoperative chemoradiotherapy would be possible. Based on gene-expression profiles of 161 patients a classifier was developed and validated in three independent datasets (n = 76), identifying over 1/3 of patients with pCR, while never misclassifying a non-complete-responder. Therefore, the classifier can identify patients suited for "watch and wait".
Collapse
Affiliation(s)
- Georg Emons
- Section of Cancer Genomics, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of General, Visceral and Pediatric Surgery, University Medical Center, Göttingen, Germany
| | - Noam Auslander
- Section of Cancer Genomics, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, PA, USA
| | - Peter Jo
- Department of General, Visceral and Pediatric Surgery, University Medical Center, Göttingen, Germany
| | - Julia Kitz
- Department of Pathology, University Medical Center, Göttingen, Germany
| | - Azadeh Azizian
- Department of General, Visceral and Pediatric Surgery, University Medical Center, Göttingen, Germany
| | - Yue Hu
- Section of Cancer Genomics, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Clemens F Hess
- Department of Radiotherapy and Radio-oncology, University Medical Center, Göttingen, Germany
| | - Claus Roedel
- Department of Radiation Oncology, University Hospital Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Ulrich Sax
- Department of Medical Informatics, University Medical Center, Göttingen, Germany
| | - Gabriela Salinas
- Transcriptome and Genome Analysis Laboratory (TAL), Department of Developmental Biochemistry, University of Göttingen, Göttingen, Germany
| | - Philipp Stroebel
- Department of Pathology, University Medical Center, Göttingen, Germany
| | - Frank Kramer
- Department of Medical Statistics, University Medical Center, Göttingen, Germany
| | - Tim Beissbarth
- Department of Medical Statistics, University Medical Center, Göttingen, Germany
| | - Marian Grade
- Department of General, Visceral and Pediatric Surgery, University Medical Center, Göttingen, Germany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center, Göttingen, Germany
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Ried
- Section of Cancer Genomics, Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jochen Gaedcke
- Department of General, Visceral and Pediatric Surgery, University Medical Center, Göttingen, Germany.
| |
Collapse
|
5
|
Jayasinghe R, Jayarajah U, Seneviratne S. Circulating Biomarkers in Predicting Pathological Response to Neoadjuvant Therapy for Colorectal Cancer. Biomark Med 2022. [DOI: 10.2174/9789815040463122010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Circulating biomarkers show promise in the management of many cancers.
They have become the novel non-invasive approach to complement the current
strategies in colorectal cancer (CRC) management. Their ability in guiding diagnosis,
evaluating response to treatment, screening and prognosis is phenomenal, especially
when it comes to their minimally invasive nature. These “liquid biopsies,” which show
potential for replacing invasive surgical biopsies, provide useful information on the
primary and metastatic disease by providing an insight into cancer biology. Analysis of
blood and body fluids for circulating tumour DNA (ctDNA), carcinoembryonic antigen
(CEA), circulating tumour cells (CTC), or circulating micro RNA (miRNA) shows
potential for improving CRC management. Recognizing a predictive model to assess
response to neoadjuvant chemotherapy would help in better patient selection. This
review was conducted with the aim of outlining the use of circulatory biomarkers in
current practice and their effectiveness in the management of patients having CRC with
a focus on response to neoadjuvant therapy.
Collapse
Affiliation(s)
- Ravindri Jayasinghe
- Department of Surgery, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Umesh Jayarajah
- Department of Surgery, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Sanjeewa Seneviratne
- Department of Surgery, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| |
Collapse
|
6
|
Lee HH, Chen CH, Huang YH, Chiang CH, Huang MY. Biomarkers of Favorable vs. Unfavorable Responses in Locally Advanced Rectal Cancer Patients Receiving Neoadjuvant Concurrent Chemoradiotherapy. Cells 2022; 11:cells11101611. [PMID: 35626648 PMCID: PMC9139800 DOI: 10.3390/cells11101611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/27/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer is the second leading cause of cancer death globally. The gold standard for locally advanced rectal cancer (LARC) nowadays is preoperative concurrent chemoradiation (CCRT). Approximately three quarters of LARC patients do not achieve pathological complete response and hence suffer from relapse, metastases and inevitable death. The exploration of trustworthy and timely biomarkers for CCRT response is urgently called for. This review focused upon a broad spectrum of biomarkers, including circulating tumor cells, DNA, RNA, oncogenes, tumor suppressor genes, epigenetics, impaired DNA mismatch repair, patient-derived xenografts, in vitro tumor organoids, immunity and microbiomes. Utilizing proper biomarkers can assist in categorizing appropriate patients by the most efficient treatment modality with the best outcome and accompanied by minimal side effects. The purpose of this review is to inspect and analyze accessible data in order to fully realize the promise of precision oncology for rectal cancer patients.
Collapse
Affiliation(s)
- Hsin-Hua Lee
- Ph.D. Program in Environmental and Occupational Medicine, National Health Research Institutes, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Hung Chen
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Radiation Oncology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung 801, Taiwan
| | - Yu-Hsiang Huang
- Post-Graduate Year Training, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Cheng-Han Chiang
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
| | - Ming-Yii Huang
- Ph.D. Program in Environmental and Occupational Medicine, National Health Research Institutes, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (C.-H.C.)
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-3121101 (ext. 7158)
| |
Collapse
|
7
|
Kirilovsky A, Sissy CE, Zeitoun G, Marliot F, Haicheur N, Lagorce-Pagès C, Taieb J, Karoui M, Custers P, Dizdarevic E, Iseas S, Hansen TF, Jensen LH, Beets G, Gérard JP, Castillo-Martin M, Figueiredo N, Habr-Gama A, Perez R, Galon J, Pagès F. The "Immunoscore" in rectal cancer: could we search quality beyond quantity of life? Oncotarget 2022; 13:18-31. [PMID: 35018217 PMCID: PMC8734641 DOI: 10.18632/oncotarget.28100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 09/26/2021] [Indexed: 12/24/2022] Open
Abstract
Because of the function and anatomical environment of the rectum, therapeutic strategies for local advanced rectal cancer (LARC) must deal with two challenging stressors that are a high-risk of local and distal recurrences and a high-risk of poor quality of life (QoL). Over the last three decades, advances in screening tests, therapies, and combined-modality treatment options and strategies have improved the prognosis of patients with LARC. However, owing to the heterogeneous nature of LARC and genetic status, the patient may not respond to a specific therapy and may be at increased risk of side-effects without the life-prolonging benefit. Indeed, each therapy can cause its own side-effects, which may worsen by a combination of treatments resulting in long-term poor QoL. In LARC, QoL has become even more essential with the increasing incidence of rectal cancer in young individuals. Herein, we analyzed the value of the Immunoscore-Biopsy (performed on tumor biopsy at diagnosis) in predicting outcomes, alone or in association with clinical and imaging data, for each therapy used in LARC.
Collapse
Affiliation(s)
- Amos Kirilovsky
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France.,These authors contributed equally to this work
| | - Carine El Sissy
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France.,These authors contributed equally to this work
| | - Guy Zeitoun
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France.,These authors contributed equally to this work
| | - Florence Marliot
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| | - Nacilla Haicheur
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| | - Christine Lagorce-Pagès
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Department of Pathology, AP-HP, Georges Pompidou European Hospital, Paris, France
| | - Julien Taieb
- Department of Gastroenterology and Gastrointestinal Oncology, AP-HP, Georges Pompidou European Hospital, Université de Paris, Paris, France
| | - Mehdi Karoui
- Department of Digestive Surgery, AP-HP, Georges Pompidou European Hospital, Université de Paris, Paris, France
| | - Petra Custers
- Department of Surgery, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Edina Dizdarevic
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark.,Danish Colorectal Cancer Center South, Vejle Hospital, Vejle, Denmark
| | - Soledad Iseas
- Oncology Unit, Gastroenterology Hospital, Dr. Carlos Bonorino Udaondo, Ciudad Autónoma de Buenos Aires, Argentina
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark.,Danish Colorectal Cancer Center South, Vejle Hospital, Vejle, Denmark
| | - Lars Henrik Jensen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark.,Danish Colorectal Cancer Center South, Vejle Hospital, Vejle, Denmark
| | - Geerard Beets
- Department of Surgery, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Jean Pierre Gérard
- Department of Radiation Oncology, Centre Antoine Lacassagne, Nice Sophia-Antipolis University, Nice, France
| | - Mireia Castillo-Martin
- Service of Pathology, Champalimaud Foundation Biobank (CFB)/Champalimaud Centre for the Unknown/Champalimaud Foundation, Lisbon, Portugal
| | - Nuno Figueiredo
- Colorectal Surgery, Digestive Department, Champalimaud Foundation, Lisbon, Portugal.,Colorectal Surgery, Lusiadas Hospital Lisboa, Lisbon, Portugal
| | - Angelita Habr-Gama
- Department of Colorectal Surgery, Angelita & Joaquim Gama Institute, São Paulo, Brazil
| | - Rodrigo Perez
- Department of Colorectal Surgery, Angelita & Joaquim Gama Institute, São Paulo, Brazil
| | - Jérôme Galon
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| | - Franck Pagès
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| |
Collapse
|
8
|
Mendis S, To YH, Tie J. Biomarkers in Locally Advanced Rectal Cancer: A Review. Clin Colorectal Cancer 2021; 21:36-44. [PMID: 34961731 DOI: 10.1016/j.clcc.2021.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/03/2022]
Abstract
Locally advanced rectal cancers (LARC) are the subject of a rapidly evolving treatment paradigm. The critical timepoints where management decisions are required during the care of the LARC patient are: prior to the institution of any treatment, post neoadjuvant therapy and post-surgery. This article reviews the clinical, imaging, blood-based, tissue-based, and molecular biomarkers that can assist clinicians at these timepoints in the patient's management, in prognosticating for their LARC patients or in predicting responses to therapy in the multi-modality neoadjuvant treatment era.
Collapse
Affiliation(s)
- Shehara Mendis
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville VIC 3052, Australia; 2. Western Health, Melbourne, VIC, Australia.
| | - Yat Hang To
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville VIC 3052, Australia; Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jeanne Tie
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville VIC 3052, Australia; Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Momma T, Okayama H, Kanke Y, Fukai S, Onozawa H, Fujita S, Sakamoto W, Saito M, Ohki S, Kono K. Validation of Gene Expression-Based Predictive Biomarkers for Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer. Cancers (Basel) 2021; 13:cancers13184642. [PMID: 34572869 PMCID: PMC8467397 DOI: 10.3390/cancers13184642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (nCRT) followed by surgery is widely used for patients with locally advanced rectal cancer. However, response to nCRT varies substantially among patients, highlighting the need for predictive biomarkers that can distinguish non-responsive from responsive patients before nCRT. This study aimed to build novel multi-gene assays for predicting nCRT response, and to validate our signature and previously-reported signatures in multiple independent cohorts. METHODS Three microarray datasets of pre-therapeutic biopsies containing a total of 61 non-responders and 53 responders were used as the discovery cohorts to screen for genes that were consistently associated with nCRT response. The predictive values of signatures were tested in a meta-analysis using six independent datasets as the validation cohorts, consisted of a total of 176 non-responders and 99 responders. RESULTS We identified four genes, including BRCA1, GPR110, TNIK, and WDR4 in the discovery cohorts. Although our 4-gene signature and nine published signatures were evaluated, they were unable to predict nCRT response in the validation cohorts. CONCLUSIONS Although this is one of the largest studies addressing the validity of gene expression-based classifiers using pre-treatment biopsies from patients with rectal cancer, our findings do not support their clinically meaningful values to be predictive of nCRT response.
Collapse
Affiliation(s)
- Tomoyuki Momma
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Hirokazu Okayama
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
- Correspondence: ; Tel.: +81-24-547-1259
| | - Yasuyuki Kanke
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Satoshi Fukai
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Hisashi Onozawa
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Shotaro Fujita
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Wataru Sakamoto
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Motonobu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| | - Shinji Ohki
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
- Hospital Director, Shirakawa Kosei General Hospital, 2-1 Kamiyajiro, Shirakawa, Fukushima 961-0005, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan; (T.M.); (Y.K.); (S.F.); (H.O.); (S.F.); (W.S.); (M.S.); (S.O.); (K.K.)
| |
Collapse
|
10
|
Biomarkers and cell-based models to predict the outcome of neoadjuvant therapy for rectal cancer patients. Biomark Res 2021; 9:60. [PMID: 34321074 PMCID: PMC8317379 DOI: 10.1186/s40364-021-00313-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
Rectal cancer constitutes approximately one-third of all colorectal cancers and contributes to considerable mortality globally. In contrast to colon cancer, the standard treatment for localized rectal cancer often involves neoadjuvant chemoradiotherapy. Tumour response rates to treatment show substantial inter-patient heterogeneity, indicating a need for treatment stratification. Consequently researchers have attempted to establish new means for predicting tumour response in order to assist in treatment decisions. In this review we have summarized published findings regarding potential biomarkers to predict neoadjuvant treatment response for rectal cancer tumours. In addition, we describe cell-based models that can be utilized both for treatment prediction and for studying the complex mechanisms involved.
Collapse
|
11
|
Xiao WW, Li M, Guo ZW, Zhang R, Xi SY, Zhang XG, Li Y, Wu DQ, Ren YF, Pang XL, Wan XB, Li K, Zhou CL, Zhai XM, Liang ZK, Wang QX, Zeng ZF, Zhang HZ, Yang XX, Wu YS, Li M, Gao YH. A Genotype Signature for Predicting Pathologic Complete Response in Locally Advanced Rectal Cancer. Int J Radiat Oncol Biol Phys 2021; 110:482-491. [PMID: 33434612 DOI: 10.1016/j.ijrobp.2021.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 02/05/2023]
Abstract
PURPOSE To construct and validate a predicting genotype signature for pathologic complete response (pCR) in locally advanced rectal cancer (PGS-LARC) after neoadjuvant chemoradiation. METHODS AND MATERIALS Whole exome sequencing was performed in 15 LARC tissues. Mutation sites were selected according to the whole exome sequencing data and literature. Target sequencing was performed in a training cohort (n = 202) to build the PGS-LARC model using regression analysis, and internal (n = 76) and external validation cohorts (n = 69) were used for validating the results. Predictive performance of the PGS-LARC model was compared with clinical factors and between subgroups. The PGS-LARC model comprised 15 genes. RESULTS The area under the curve (AUC) of the PGS model in the training, internal, and external validation cohorts was 0.776 (0.697-0.849), 0.760 (0.644-0.867), and 0.812 (0.690-0.915), respectively, and demonstrated higher AUC, accuracy, sensitivity, and specificity than cT stage, cN stage, carcinoembryonic antigen level, and CA19-9 level for pCR prediction. The predictive performance of the model was superior to clinical factors in all subgroups. For patients with clinical complete response (cCR), the positive prediction value was 94.7%. CONCLUSIONS The PGS-LARC is a reliable predictive tool for pCR in patients with LARC and might be helpful to enable nonoperative management strategy in those patients who refuse surgery. It has the potential to guide treatment decisions for patients with different probability of tumor regression after neoadjuvant therapy, especially when combining cCR criteria and PGS-LARC.
Collapse
Affiliation(s)
- Wei-Wei Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Min Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Guangzhou Darui Biotechnology Co, Ltd High-Tech Development Zone, Guangzhou, Guangdong, China; Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhi-Wei Guo
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Rong Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shao-Yan Xi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xiang-Guo Zhang
- Department of Radiation Oncology, Affiliated Yuebei People Hospital of Shantou University Medical College, ShaoGuan, Guangdong, China
| | - Yong Li
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - De-Qing Wu
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yu-Feng Ren
- Department of Radiation Oncology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Lin Pang
- Department of Radiation Oncology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiang-Bo Wan
- Department of Radiation Oncology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kun Li
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chun-Lian Zhou
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiang-Ming Zhai
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhi-Kun Liang
- Guangzhou Darui Biotechnology Co, Ltd High-Tech Development Zone, Guangzhou, Guangdong, China
| | - Qiao-Xuan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhi-Fan Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Hui-Zhong Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xue-Xi Yang
- Guangzhou Darui Biotechnology Co, Ltd High-Tech Development Zone, Guangzhou, Guangdong, China; Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying-Song Wu
- Guangzhou Darui Biotechnology Co, Ltd High-Tech Development Zone, Guangzhou, Guangdong, China; Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Ming Li
- Guangzhou Darui Biotechnology Co, Ltd High-Tech Development Zone, Guangzhou, Guangdong, China
| | - Yuan-Hong Gao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China; Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
Izzotti A, Ceccaroli C, Geretto M, Ruggieri FG, Schenone S, Di Maria E. Predicting Response to Neoadjuvant Therapy in Colorectal Cancer Patients the Role of Messenger-and Micro-RNA Profiling. Cancers (Basel) 2020; 12:cancers12061652. [PMID: 32580435 PMCID: PMC7352797 DOI: 10.3390/cancers12061652] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer patients' responses to neoadjuvant therapy undergo broad inter-individual variations. The aim of this systematic review is to identify a molecular signature that is predictive of colon cancer downstaging and/or downgrading after neoadjuvant therapy. Among the hundreds analysed in the available studies, only 19 messenger-RNAs (mRNAs) and six micro-RNAs (miRNAs) were differentially expressed in responders versus non-responders in two or more independent studies. Therefore, a mRNA/miRNA signature can be designed accordingly, with limitations caused by the retrospective nature of these studies, the heterogeneity in study designs and the downgrading/downstaging assessment criteria. This signature can be proposed to tailor neoadjuvant therapy regimens on an individual basis.
Collapse
Affiliation(s)
- Alberto Izzotti
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Correspondence: ; Tel.: +39-010-353-8522
| | | | - Marta Geretto
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy;
| | | | - Sara Schenone
- Department of Health Sciences, University of Genova, 16132 Genova, Italy; (S.S.); (E.D.M.)
| | - Emilio Di Maria
- Department of Health Sciences, University of Genova, 16132 Genova, Italy; (S.S.); (E.D.M.)
- Unit of Medical Genetics, Galliera Hospital, 16128 Genoa, Italy
| |
Collapse
|
13
|
Kamran SC, Lennerz JK, Margolis CA, Liu D, Reardon B, Wankowicz SA, Van Seventer EE, Tracy A, Wo JY, Carter SL, Willers H, Corcoran RB, Hong TS, Van Allen EM. Integrative Molecular Characterization of Resistance to Neoadjuvant Chemoradiation in Rectal Cancer. Clin Cancer Res 2019; 25:5561-5571. [PMID: 31253631 PMCID: PMC6744983 DOI: 10.1158/1078-0432.ccr-19-0908] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/08/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Molecular properties associated with complete response or acquired resistance to concurrent chemotherapy and radiotherapy (CRT) are incompletely characterized.Experimental Design: We performed integrated whole-exome/transcriptome sequencing and immune infiltrate analysis on rectal adenocarcinoma tumors prior to neoadjuvant CRT (pre-CRT) and at time of resection (post-CRT) in 17 patients [8 complete/partial responders, 9 nonresponders (NR)]. RESULTS CRT was not associated with increased tumor mutational burden or neoantigen load and did not alter the distribution of established somatic tumor mutations in rectal cancer. Concurrent KRAS/TP53 mutations (KP) associated with NR tumors and were enriched for an epithelial-mesenchymal transition transcriptional program. Furthermore, NR was associated with reduced CD4/CD8 T-cell infiltrates and a post-CRT M2 macrophage phenotype. Absence of any local tumor recurrences, KP/NR status predicted worse progression-free survival, suggesting that local immune escape during or after CRT with specific genomic features contributes to distant progression. CONCLUSIONS Overall, while CRT did not impact genomic profiles, CRT impacted the tumor immune microenvironment, particularly in resistant cases.
Collapse
Affiliation(s)
- Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Jochen K Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital, Boston, Massachusetts
| | - Claire A Margolis
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - David Liu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Brendan Reardon
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Stephanie A Wankowicz
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Emily E Van Seventer
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Adam Tracy
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Jennifer Y Wo
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Scott L Carter
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Joint Center for Cancer Precision Medicine, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ryan B Corcoran
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Eliezer M Van Allen
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
14
|
Cho E, Park IJ, Yeom SS, Hong SM, Lee JB, Kim YW, Kim MJ, Lim HM, Lim SB, Yu CS, Kim JC. A Multigene Model for Predicting Tumor Responsiveness After Preoperative Chemoradiotherapy for Rectal Cancer. Int J Radiat Oncol Biol Phys 2019; 105:834-842. [PMID: 31419511 DOI: 10.1016/j.ijrobp.2019.07.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 12/29/2022]
Abstract
PURPOSE Although preoperative chemoradiotherapy (PCRT) is regarded as a standard treatment for locally advanced rectal cancer, there is no reliable biomarker for predicting responsiveness to PCRT. We aimed to develop a biomarker model for predicting response to PCRT. METHODS AND MATERIALS We included 184 patients who received PCRT followed by surgical resection and categorized them as good responders (complete or near-complete regression) or poor responders (all other patients). Candidate gene mRNAs were isolated from formalin-fixed paraffin-embedded tumor specimens and analyzed using the NanoString nCounter gene expression assay. Stepwise logistic regression analysis was used to select genes in discovery and training phases. A quantitative radio-responsiveness prediction model was developed and validated using internal cross-validation groups, and the model's predictive value was assessed based on the area under the receiver operating characteristic curve (AUC). RESULTS By comparing the gene expressions between good and poor responders, we created a multigene mRNA model using FZD9, HRAS, ITGA7, MECOM, MMP3, NKD1, PIK3CD, and PRKCB. This panel showed good ability to predict treatment response (AUC: 0.846 for the whole data set). Internal cross-validation was performed to evaluate the model's predictive stability among 3 cohorts, which provided AUC values of 0.808-0.909. The satisfactory diagnostic performance of the radio-response prediction index persisted regardless of other clinicopathologic features such as clinical T or N stage, interval between radiation and surgery, and pretreatment carcinoembryonic antigen levels (P = .001, 95% CI, 0.686-0.905). CONCLUSIONS We developed a multigene mRNA-based biomarker model that allows prediction of rectal cancer response to PCRT, which may help identify patients who will benefit most from PCRT.
Collapse
Affiliation(s)
- Eunhae Cho
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In Ja Park
- Department of Colon and Rectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Seung-Seop Yeom
- Department of Colon and Rectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jung Bok Lee
- Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yeon Wook Kim
- Asan Institute for Life Science, Asan Medical Center, Seoul, Korea
| | - Mi-Ju Kim
- Asan Institute for Life Science, Asan Medical Center, Seoul, Korea
| | - Hye Min Lim
- Asan Institute for Life Science, Asan Medical Center, Seoul, Korea
| | - Seok-Byung Lim
- Department of Colon and Rectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang Sik Yu
- Department of Colon and Rectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Cheon Kim
- Department of Colon and Rectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
Koyama FC, Lopes Ramos CM, Ledesma F, Alves VAF, Fernandes JM, Vailati BB, São Julião GP, Habr-Gama A, Gama-Rodrigues J, Perez RO, Camargo AA. Effect of Akt activation and experimental pharmacological inhibition on responses to neoadjuvant chemoradiotherapy in rectal cancer. Br J Surg 2018; 105:e192-e203. [PMID: 29341150 DOI: 10.1002/bjs.10695] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (CRT) is one of the preferred initial treatment strategies for locally advanced rectal cancer. Responses are variable, and most patients still require surgery. The aim of this study was to identify molecular mechanisms determining poor response to CRT. METHODS Global gene expression and pathway enrichment were assessed in pretreatment biopsies from patients with non-metastatic cT2-4 N0-2 rectal cancer within 7 cm of the anal verge. Downstream Akt activation was assessed in an independent set of pretreatment biopsies and in colorectal cancer cell lines using immunohistochemistry and western blot respectively. The radiosensitizing effects of the Akt inhibitor MK2206 were assessed using clonogenic assays and xenografts in immunodeficient mice. RESULTS A total of 350 differentially expressed genes were identified, of which 123 were upregulated and 199 downregulated in tumours from poor responders. Mitochondrial oxidative phosphorylation (P < 0·001) and phosphatidylinositol signalling pathways (P < 0·050) were identified as significantly enriched pathways among the set of differentially expressed genes. Deregulation of both pathways is known to result in Akt activation, and high immunoexpression of phosphorylated Akt S473 was observed among patients with a poor histological response (tumour regression grade 0-2) to CRT (75 per cent versus 48 per cent in those with a good or complete response; P = 0·016). Akt activation was also confirmed in the radioresistant cell line SW480, and a 50 per cent improvement in sensitivity to CRT was observed in vitro and in vivo when SW480 cells were exposed to the Akt inhibitor MK2206 in combination with radiation and 5-fluorouracil. CONCLUSION Akt activation is a key event in the response to CRT. Pharmacological inhibition of Akt activation may enhance the effects of CRT. Surgical relevance Organ preservation is an attractive alternative in rectal cancer management following neoadjuvant chemoradiotherapy (CRT) to avoid the morbidity of radical surgery. Molecular steps associated with tumour response to CRT may provide a useful tool for the identification of patients who are candidates for no immediate surgery. In this study, tumours resistant to CRT were more likely to have activation of specific genetic pathways that result in phosphorylated Akt (pAkt) activation. Pretreatment biopsy tissues with high immunoexpression of pAkt were more likely to exhibit a poor histological response to CRT. In addition, the introduction of a pAkt inhibitor to cancer cell lines in vitro and in vivo led to a significant improvement in sensitivity to CRT. Identification of pAkt-activated tumours may thus allow the identification of poor responders to CRT. In addition, the concomitant use of pAkt inhibitors to increase sensitivity to CRT in patients with rectal cancer may constitute an interesting strategy for increasing the chance of a complete response to treatment and organ preservation.
Collapse
Affiliation(s)
- F C Koyama
- Molecular Oncology Centre, Hospital Sírio Libanês, São Paulo, Brazil.,Ludwig Institute for Cancer Research, São Paulo, Brazil
| | - C M Lopes Ramos
- Molecular Oncology Centre, Hospital Sírio Libanês, São Paulo, Brazil
| | - F Ledesma
- Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - V A F Alves
- Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - J M Fernandes
- Molecular Oncology Centre, Hospital Sírio Libanês, São Paulo, Brazil
| | - B B Vailati
- Instituto Angelita and Joaquim Gama, São Paulo, Brazil
| | | | - A Habr-Gama
- Instituto Angelita and Joaquim Gama, São Paulo, Brazil.,Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - J Gama-Rodrigues
- Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - R O Perez
- Ludwig Institute for Cancer Research, São Paulo, Brazil.,Instituto Angelita and Joaquim Gama, São Paulo, Brazil.,Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Digestive Surgical Oncology Division, BP - A Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| | - A A Camargo
- Molecular Oncology Centre, Hospital Sírio Libanês, São Paulo, Brazil.,Ludwig Institute for Cancer Research, São Paulo, Brazil
| |
Collapse
|
16
|
Abstract
The management of locally-advanced rectal cancer involves a combination of chemotherapy, chemoradiation, and surgical resection to provide excellent local tumor control and overall survival. However, aspects of this multimodality approach are associated with significant morbidity and long-term sequelae. In addition, there is growing evidence that patients with a clinical complete response to chemotherapy and chemoradiation treatments may be safely offered initial non-operative management in a rigorous surveillance program. Weighed against the morbidity and significant sequelae of rectal resection, recognizing how to best optimize non-operative strategies without compromising oncologic outcomes is critical to our understanding and treatment of this disease.
Collapse
Affiliation(s)
- Iris H Wei
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering, New York, NY, USA -
| | - Julio Garcia-Aguilar
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering, New York, NY, USA
| |
Collapse
|
17
|
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: 1.2] [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
|
18
|
Kim SC, Shin YK, Kim YA, Jang SG, Ku JL. Identification of genes inducing resistance to ionizing radiation in human rectal cancer cell lines: re-sensitization of radio-resistant rectal cancer cells through down regulating NDRG1. BMC Cancer 2018; 18:594. [PMID: 29801473 PMCID: PMC5970486 DOI: 10.1186/s12885-018-4514-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 05/17/2018] [Indexed: 12/15/2022] Open
Abstract
Background Resistance to preoperative radiotherapy is a major clinical problem in the treatment for locally advanced rectal cancer. The role of NDRG1 in resistance to ionizing radiation in rectal cancer has not been fully elucidated. This study aimed to investigate the effect of the reduced intracellular NDRG1 expression on radio-sensitivity of human rectal cancer cells for exploring novel approaches for treatment of rectal cancer. Methods Three radio-resistant human rectal cancer cell lines (SNU-61R80Gy, SNU-283R80Gy, and SNU-503R80Gy) were established from human rectal cancer cell lines (SNU-61, SNU-283, and SNU-503) using total 80 Gy of fractionated irradiation. Microarray analysis was performed to identify differently expressed genes in newly established radio-resistant human rectal cancer cells compared to parental rectal cancer cells. Results A microarray analysis indicated the RNA expression of five genes (NDRG1, ERRFI1, H19, MPZL3, and UCA1) was highly increased in radio-resistant rectal cancer cell lines. Short hairpin RNA-mediated silencing of NDRG1 sensitized rectal cancer cell lines to clinically relevant doses of radiation by causing more DNA double strand breakages to rectal cancer cells when exposed to radiation. Conclusions Targeting NDRG1 represents a promising strategy to increase response to radiotherapy in human rectal cancer. Electronic supplementary material The online version of this article (10.1186/s12885-018-4514-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Soon-Chan Kim
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Young-Kyoung Shin
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Ye-Ah Kim
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Sang-Geun Jang
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Ja-Lok Ku
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
19
|
Luo J, Liu L, Zhou N, Shen J, Sun Q, Zhu Y, Chen M. miR-519b-3p promotes responsiveness to preoperative chemoradiotherapy in rectal cancer patients by targeting ARID4B. Gene 2018; 655:84-90. [DOI: 10.1016/j.gene.2018.02.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/22/2018] [Indexed: 02/07/2023]
|
20
|
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: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
Karagkounis G, Kalady MF. Molecular Biology: Are We Getting Any Closer to Providing Clinically Useful Information? Clin Colon Rectal Surg 2017; 30:415-422. [PMID: 29184477 DOI: 10.1055/s-0037-1606373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Advances in molecular biology and biomarker research have significantly impacted our understanding and treatment of multiple solid malignancies. In rectal cancer, where neoadjuvant chemoradiation is widely used for locally advanced disease, most efforts have focused on the identification of predictors of response in an attempt to appropriately select patients for multimodality therapy. A variety of biomarkers have been studied, including genetic mutations, chromosomal copy number alterations, and single as well as multigene expression patterns. Also, as transanal resection of rectal tumors requires accurate preoperative detection of lymph node metastasis, the identification of biomarkers of regional nodal involvement has been another important field of active research. While preliminary results have been promising, lack of external validation means has a limited translation to clinical use. This review summarizes recent developments in rectal cancer biomarker research, highlighting the challenges associated with their adoption, and evaluating their potential for clinical use.
Collapse
Affiliation(s)
- Georgios Karagkounis
- Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Matthew F Kalady
- Department of Colorectal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
22
|
Biomarkers that Predict Response to Neoadjuvant Chemoradiation in Locally Advanced Rectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2017. [DOI: 10.1007/s11888-017-0376-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
23
|
Quantitative imaging outperforms molecular markers when predicting response to chemoradiotherapy for rectal cancer. Radiother Oncol 2017. [PMID: 28647399 DOI: 10.1016/j.radonc.2017.06.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE To explore the integration of imaging and molecular data for response prediction to chemoradiotherapy (CRT) for rectal cancer. MATERIAL AND METHODS Eighty-five rectal cancer patients underwent preoperative CRT. 18F-FDG PET/CT and diffusion-weighted imaging (DWI) were acquired before (TP1) and during CRT (TP2) and prior to surgery (TP3). Inflammatory cytokines and gene expression were analysed. Tumour response was defined as ypT0-1N0. Multivariate models were built combining the obtained parameters. Final models were calculated on the data combination with the highest AUC. RESULTS Twenty-two patients (26%) achieved ypT0-1N0 response. 18F-FDG PET/CT had worse predictive performance than DWI and T2-volumetry (AUC 0.61±0.04, 0.72±0.03, and 0.72±0.02, respectively). Combining all imaging parameters increased the AUC to 0.81±0.03. Adding cytokines or gene expression did not improve the AUC (AUC of 0.72±0.06 and 0.79±0.04 respectively). Final models combining 18F-FDG PET/CT, DWI, and T2-weighted volumetry at all TPs and using only TP1 and TP3, allowed ypT0-1N0 prediction with a 75% sensitivity, 94% specificity and PPV of 80%. CONCLUSIONS Combining 18F-FDG PET/CT, DWI, and T2-weighted MRI volumetry obtained before CRT and prior to surgery may help physicians in selecting rectal cancer patients for organ-preservation.
Collapse
|
24
|
Intratumoral Genetic Heterogeneity in Rectal Cancer: Are Single Biopsies representative of the entirety of the tumor? Ann Surg 2017; 265:e4-e6. [PMID: 27479130 DOI: 10.1097/sla.0000000000001937] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Demonstrate intratumoral genetic heterogeneity in rectal cancer. BACKGROUND Several clinical management decisions in rectal cancer may be influenced by pretreatment biopsy information. However, in the setting of significant intratumoral heterogeneity, biopsies may not be representative of the entirety of the tumor and limit the reliability of the information provided from them for clinical decision management. METHODS Three fragments from a single rectal adenocarcinoma were chosen for whole-exome sequencing followed by mutation detection analysis. About 25 Gb of unambiguously mapped sequences were generated for each sample resulting in a median fold-coverage of 35x. Captured sequences mapped to the reference human genome were then used for the detection of somatic point mutations. RESULTS Overall, 193 unique somatic point mutations were identified. Only 53 (27%) of these were shared by all three fragments, including known genes involved in early phases of the adenoma-carcinoma sequence (such as, APC). Approximately, 115 (59%) mutations were exclusively present in only one of the fragments, including mutations in "driver" genes (DNAH12). Jaccard distances showed a median distance of 0.603 for pair-wise comparison of fragments indicating significant heterogeneity between them. CONCLUSIONS Considerable intratumoral heterogeneity is present among naive rectal cancers. The majority of point mutations detected in different fragments from rectal cancers are frequently unique to a single fragment. These findings support that gene mutations found on single pretreatment biopsies will not necessarily be representative of mutations present in the entirety of the tumor and therefore may limit the utility of the biological information provided by single biopsy fragments for clinical management decisions.
Collapse
|
25
|
Discriminating cancer-related and cancer-unrelated chemoradiation-response genes for locally advanced rectal cancers. Sci Rep 2016; 6:36935. [PMID: 27845363 PMCID: PMC5109405 DOI: 10.1038/srep36935] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/24/2016] [Indexed: 02/07/2023] Open
Abstract
For patients with locally advanced rectal cancer (LARC) treated with preoperation chemoradiation (pCRT), identifying differentially expressed (DE) genes between non-responders and responders is a common approach for investigating mechanisms of chemoradiation resistance. However, some of such DE genes might be irrelevant to cancer itself but simply reflect the pharmacokinetic differences of the normal tissues. In this study, we adopted the RankComp algorithm to identify DE genes for each of LARC sample compared with its own normal state. Then, we identified genes with significantly different deregulation frequencies between the non-responders and responders, defined as cancer-related pCRT-response genes. Pathway enrichment and protein-protein interaction analyses showed that these genes specifically and intensively interacted with currently known effective genes of pCRT, involving in DNA replication, cell cycle and DNA repair. In contrast, after excluding the cancer-related pCRT-response genes, the other DE genes between non-responders and responders were enriched in many pathways of drug and protein metabolisms and transports, and interacted with both the known effective genes and pharmacokinetic genes. Hence, these two types of DE genes should be distinguished for investigating mechanisms of pCRT response in LARCs.
Collapse
|
26
|
Millino C, Maretto I, Pacchioni B, Digito M, De Paoli A, Canzonieri V, D'Angelo E, Agostini M, Rizzolio F, Giordano A, Barina A, Rajendran S, Esposito G, Lanfranchi G, Nitti D, Pucciarelli S. Gene and MicroRNA Expression Are Predictive of Tumor Response in Rectal Adenocarcinoma Patients Treated With Preoperative Chemoradiotherapy. J Cell Physiol 2016; 232:426-435. [PMID: 27225591 DOI: 10.1002/jcp.25441] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 05/24/2016] [Indexed: 01/05/2023]
Abstract
Preoperative chemoradiotherapy (pCRT) followed by surgery is the standard treatment for locally advanced rectal cancer (LARC). However, tumor response to pCRT is not uniform, and there are no effective predictive methods. This study investigated whether specific gene and miRNA expression are associated with tumor response to pCRT. Tissue biopsies were obtained from patients before pCRT and resection. Gene and miRNA expression were analyzed using a one-color microarray technique that compares signatures between responders (R) and non-responders (NR), as measured based on tumor regression grade. Two groups composed of 38 "exploration cohort" and 21 "validation cohort" LARC patients were considered for a total of 32 NR and 27 R patients. In the first cohort, using SAM Two Class analysis, 256 genes and 29 miRNAs that were differentially expressed between the NR and R patients were identified. The anti-correlation analysis showed that the same 8 miRNA interacted with different networks of transcripts. The miR-630 appeared only with the NR patients and was anti-correlated with a single transcript: RAB5B. After PAM, the following eight transcripts were strong predictors of tumor response: TMEM188, ITGA2, NRG, TRAM1, BCL2L13, MYO1B, KLF7, and GTSE1. Using this gene set, an unsupervised cluster analysis was applied to the validation cohort and correctly assigned the patients to the NR or R group with 85.7% accuracy, 90% sensitivity, and 82% specificity. All three parameters reached 100% when both cohorts were considered together. In conclusion, gene and miRNA expression profiles may be helpful for predicting response to pCRT in LARC patients. J. Cell. Physiol. 232: 426-435, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Caterina Millino
- Microarray Service, Department of Biology, CRIBI Biotechnology Centre, University of Padua, Padua, Italy
| | - Isacco Maretto
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Beniamina Pacchioni
- Microarray Service, Department of Biology, CRIBI Biotechnology Centre, University of Padua, Padua, Italy
| | - Maura Digito
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Antonino De Paoli
- Department of Radiation Oncology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Vincenzo Canzonieri
- Department of Pathology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Edoardo D'Angelo
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy.,Nanoinspired Biomedicine Lab., Institute of Pediatric Research, Fondazione Città della Speranza, Padova, Italy
| | - Marco Agostini
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy.,Nanoinspired Biomedicine Lab., Institute of Pediatric Research, Fondazione Città della Speranza, Padova, Italy.,Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, Texas
| | - Flavio Rizzolio
- Department of Translational Research, National Cancer Institute, CRO-IRCSS, Aviano, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Andrea Barina
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Senthilkumar Rajendran
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Giovanni Esposito
- Sperimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Gerolamo Lanfranchi
- Microarray Service, Department of Biology, CRIBI Biotechnology Centre, University of Padua, Padua, Italy
| | - Donato Nitti
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Salvatore Pucciarelli
- 1st Surgical Clinic, Department of Surgical, Oncological, and Gastroenterological Sciences, University of Padua, Padua, Italy.
| |
Collapse
|
27
|
Huang SH, Chi P, Lin HM, Lu XR, Huang YW, Xu ZB, Sun YW, Ye DX, Wang XJ, Wang X. Selecting stage ypT0-1N0 for locally advanced rectal cancer following preoperative chemoradiotherapy: implications for potential candidates of organ-sparing management. Colorectal Dis 2016; 18:989-996. [PMID: 26880193 DOI: 10.1111/codi.13297] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 12/01/2015] [Indexed: 12/11/2022]
Abstract
AIM Local excision or a wait-and-see policy may offer the possibility of organ preservation for locally advanced rectal cancer (LARC) after preoperative chemoradiotherapy (CRT). Identifying associated factors of good responders (GR) with stage ypT0-1N0 would probably influence the selection of potential candidates who were theoretically eligible for organ-sparing management. This study was to establish a scoring system to select stage ypT0-1N0 for LARC following preoperative CRT. METHOD Between 2009 and 2014, 262 patients with middle and low LARC were treated with CRT and radical surgery. Clinicopathological data which were found to be significantly associated with GR were incorporated into a scoring system. RESULTS Fifty-seven (21.8%) patients were GR with stage ypT0-1N0 in the operative specimen. Multivariate analyses indicated that a low level of pretreatment carcinoembryonic antigen (CEA) and post-treatment CEA <2.55 ng/ml (P = 0.008 and P = 0.009 respectively) and long-axis diameter of residual tumours (P = 0.006) were independently associated with stage ypT0-1N0. The three factors were incorporated into a scoring system. Using receiver operating characteristic curve analysis, we determined a cutoff value of -0.3 for scores, at which the system's sensitivity was 71.9% and specificity 73.1%. When applied to testing samples, the sensitivity was 74.1% and specificity 76.2%. CONCLUSION We demonstrated that low levels of pretreatment and post-treatment CEA and the long-axis diameter of residual tumours were associated with stage ypT0-1N0 for LARC after CRT. Therefore, the three-factor scoring system may be used to select potential candidates for organ-sparing management.
Collapse
Affiliation(s)
- S H Huang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - P Chi
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - H M Lin
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - X R Lu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Y W Huang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Z B Xu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Y W Sun
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - D X Ye
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - X J Wang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - X Wang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
28
|
Should We Give Up The Search for a Clinically Useful Gene Signature for the Prediction of Response of Rectal Cancer to Neoadjuvant Chemoradiation? Dis Colon Rectum 2016; 59:895-7. [PMID: 27505119 DOI: 10.1097/dcr.0000000000000620] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
29
|
Lessons Learned From the Quest for Gene Signatures That Predict Treatment Response in Rectal Cancer. Dis Colon Rectum 2016; 59:898-900. [PMID: 27505120 DOI: 10.1097/dcr.0000000000000621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
30
|
An integrative approach for the identification of prognostic and predictive biomarkers in rectal cancer. Oncotarget 2016; 6:32561-74. [PMID: 26359356 PMCID: PMC4741712 DOI: 10.18632/oncotarget.4935] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/20/2015] [Indexed: 12/22/2022] Open
Abstract
Introduction Colorectal cancer is the third most common cancer in the world, a small fraction of which is represented by locally advanced rectal cancer (LARC). If not medically contraindicated, preoperative chemoradiotherapy, represent the standard of care for LARC patients. Unfortunately, patients shows a wide range of response rates in which approximately 20% has a complete pathological response, whereas in 20 to 40% the response is poor or absent. Results The following specific gene signature, able to discriminate responders' patients from non-responders, were founded: AKR1C3, CXCL11, CXCL10, IDO1, CXCL9, MMP12 and HLA-DRA. These genes are mainly involved in immune system pathways and interact with drugs traditionally used in the adjuvant treatment of rectal cancer. Discussion The present study suggests that new ideas for therapy could be found not only limited to studying genes differentially expressed between the two groups of patients but deepening the mechanisms, associated to response, in which they are involved. Methods Gene expression studies performed by: Agostini et al., Rimkus et al. and Kim et al. have been merged through a meta-analysis of the raw data. Gene expression data-sets have been processed using A-MADMAN. Common differentially expressed gene (DEG) were identified through SAM analysis. To further characterize the identified DEG we deeply investigated its biological role using an integrative computational biology approach.
Collapse
|
31
|
Gim J, Cho YB, Hong HK, Kim HC, Yun SH, Wu HG, Jeong SY, Joung JG, Park T, Park WY, Lee WY. Predicting multi-class responses to preoperative chemoradiotherapy in rectal cancer patients. Radiat Oncol 2016; 11:50. [PMID: 27005571 PMCID: PMC4804643 DOI: 10.1186/s13014-016-0623-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/15/2016] [Indexed: 02/06/2023] Open
Abstract
Background Preoperative chemoradiotherapy (CRT) has become a widely used treatment for improving local control of disease and increasing survival rates of rectal cancer patients. We aimed to identify a set of genes that can be used to predict responses to CRT in patients with rectal cancer. Methods Gene expression profiles of pre-therapeutic biopsy specimens obtained from 77 rectal cancer patients were analyzed using DNA microarrays. The response to CRT was determined using the Dworak tumor regression grade: grade 1 (minimal, MI), grade 2 (moderate, MO), grade 3 (near total, NT), or grade 4 (total, TO). Results Top ranked genes for three different feature scores such as a p-value (pval), a rank product (rank), and a normalized product (norm) were selected to distinguish pre-defined groups such as complete responders (TO) from the MI, MO, and NT groups. Among five different classification algorithms, supporting vector machine (SVM) with the top 65 norm features performed at the highest accuracy for predicting MI using a 5-fold cross validation strategy. On the other hand, 98 pval features were selected for predicting TO by elastic net (EN). Finally we combined TO- and MI-finder models to build a three-class classification model and validated it using an independent dataset of rectal cancer mRNA expression. Conclusions We identified MI- and TO-finders for predicting preoperative CRT responses, and validated these data using an independent public dataset. This stepwise prediction model requires further evaluation in clinical studies in order to develop personalized preoperative CRT in patients with rectal cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13014-016-0623-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jungsoo Gim
- Institute of Health and Environment, Seoul National University, Seoul, 151-742, Korea
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Hye Kyung Hong
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Seong Hyeon Yun
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea
| | - Hong-Gyun Wu
- Department of Radiation Oncology, College of Medicine, Seoul National University, Seoul, 110-799, Korea
| | - Seung-Yong Jeong
- Department of Surgery, College of Medicine, Seoul National University, Seoul, 110-799, Korea
| | - Je-Gun Joung
- Samsung Medical Center, Samsung Genome Institute, Seoul, 135-710, Korea
| | - Taesung Park
- Institute of Health and Environment, Seoul National University, Seoul, 151-742, Korea. .,Department of Statistics, College of Natural Science, Seoul National University, Seoul, 151-742, Korea.
| | - Woong-Yang Park
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea. .,Samsung Medical Center, Samsung Genome Institute, Seoul, 135-710, Korea.
| | - Woo Yong Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 135-710, Korea.
| |
Collapse
|
32
|
Ryan JE, Warrier SK, Lynch AC, Ramsay RG, Phillips WA, Heriot AG. Predicting pathological complete response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer: a systematic review. Colorectal Dis 2016; 18:234-46. [PMID: 26531759 DOI: 10.1111/codi.13207] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 09/17/2015] [Indexed: 02/08/2023]
Abstract
AIM Approximately 20% of patients treated with neoadjuvant chemoradiotherapy (nCRT) for locally advanced rectal cancer achieve a pathological complete response (pCR) while the remainder derive the benefit of improved local control and downstaging and a small proportion show a minimal response. The ability to predict which patients will benefit would allow for improved patient stratification directing therapy to those who are likely to achieve a good response, thereby avoiding ineffective treatment in those unlikely to benefit. METHOD A systematic review of the English language literature was conducted to identify pathological factors, imaging modalities and molecular factors that predict pCR following chemoradiotherapy. PubMed, MEDLINE and Cochrane Database searches were conducted with the following keywords and MeSH search terms: 'rectal neoplasm', 'response', 'neoadjuvant', 'preoperative chemoradiation', 'tumor response'. After review of title and abstracts, 85 articles addressing the prediction of pCR were selected. RESULTS Clear methods to predict pCR before chemoradiotherapy have not been defined. Clinical and radiological features of the primary cancer have limited ability to predict response. Molecular profiling holds the greatest potential to predict pCR but adoption of this technology will require greater concordance between cohorts for the biomarkers currently under investigation. CONCLUSION At present no robust markers of the prediction of pCR have been identified and the topic remains an area for future research. This review critically evaluates existing literature providing an overview of the methods currently available to predict pCR to nCRT for locally advanced rectal cancer. The review also provides a comprehensive comparison of the accuracy of each modality.
Collapse
Affiliation(s)
- J E Ryan
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Epworth Healthcare, Melbourne, Victoria, Australia.,Austin Academic Centre, University of Melbourne, Parkville, Victoria, Australia
| | - S K Warrier
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - A C Lynch
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - R G Ramsay
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - W A Phillips
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Cancer Biology and Surgical Oncology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - A G Heriot
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
33
|
NPTX2 is associated with neoadjuvant therapy response in rectal cancer. J Surg Res 2015; 202:112-7. [PMID: 27083956 DOI: 10.1016/j.jss.2015.12.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/08/2015] [Accepted: 12/23/2015] [Indexed: 11/22/2022]
Abstract
BACKGROUND Neoadjuvant chemoradiation (CRT) is recommended for locally advanced rectal cancer. Tumor response varies from pathologic complete response (pCR) to no tumor regression. The mechanisms behind CRT resistance remain undefined. In our previously generated complementary DNA microarrays of pretreatment biopsies from rectal cancer patients, neuronal pentraxin 2 (NPTX2) expression discriminated patients with pCR from those with residual tumor. As tumor response is prognostic for survival, we sought to evaluate the clinical relevance of NPTX2 in rectal cancer. MATERIALS AND METHODS Real-time quantitative polymerase chain reaction was used to evaluate NPTX2 messenger RNA expression in individual rectal cancers before CRT. Tumors with NPTX2 expression <50% of normal rectum were defined as NPTX2-low and those with >50% were defined as NPTX2-high. NPTX2 levels were compared to response to therapy and oncologic outcomes using Mann-Whitney, Kruskal-Wallis, chi-square, and Mantel-Cox (log-rank) tests, as appropriate. RESULTS Rectal cancers from 40 patients were included. The mean patient age was 56.8 years, and 30% were female. pCR was achieved in eight of 40 patients (20%). In these patients, messenger RNA NPTX2 levels were significantly decreased compared to those with residual cancer (fold change 30.4, P = 0.017). Patients with NPTX2-low tumors (n = 13) achieved improved response to treatment (P = 0.012 versus NPXT2-high tumors), with 38.5% and 46.1% of patients achieving complete or moderate response, respectively. Of patients with NPTX2-high tumors (n = 27), 11.1% and 18.5% achieved complete or moderate response, respectively. No recurrence or death was recorded in patients with NPTX2-low tumors, reflecting more favorable disease-free survival (P = 0.045). CONCLUSIONS Decreased NPTX2 expression in rectal adenocarcinomas is associated with improved response to CRT and improved prognosis. Further studies to validate these results and elucidate the biological role of NPTX2 in rectal cancer are needed.
Collapse
|
34
|
Kim JY, Park IJ, Hong SM, Lee JL, Yoon YS, Kim CW, Lim SB, Lee JB, Yu CS, Kim JC. Is Pathologic Near-Total Regression an Appropriate Indicator of a Good Response to Preoperative Chemoradiotherapy Based on Oncologic Outcome of Disease? Medicine (Baltimore) 2015; 94:e2257. [PMID: 26683945 PMCID: PMC5058917 DOI: 10.1097/md.0000000000002257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
We evaluated the oncologic outcomes of patients with rectal cancer who demonstrated pathologic near-total regression (NTR) after preoperative chemoradiotherapy (PCRT) and compared with total regression (TR). Pathologic NTR in rectal cancer by tumor regression grade (TRG) is usually considered to indicate a good response, when evaluating tumor response to PCRT. We retrospectively analyzed the outcomes in 263 patients who received PCRT for advanced T3/4 or N+ rectal cancer followed by radical resection. Patients were diagnosed with TR (n = 132) or NTR (n = 131) according to the TRG. Recurrence-free survival (RFS) was evaluated and compared between groups. For evaluating the consistency between the result and previously published data, meta-analysis for summing up survival curve was performed using generalized linear mixed model. ypT status was heterogeneous in the NTR group as follows; 3 Tis (2.3%), 21 T1 (16%), 72 T2 (55%), and 35 T3 (26.7%). Metastatic lymph nodes were more frequently found in the NTR group (6.8% in TR vs 24.4% in NTR patients; P = 0.003). The cumulative recurrence rate was higher in the NTR group (19.8% vs 6.1%; P = 0.003). The 5-year RFS was lower in the NTR group (94% vs 77.8%; P = 0.001). Significant differences in the RFS rate were found in comparison with the published literature. Based on differences in the oncologic outcomes between the TR and NTR groups, it might not be suitable to use NTR as an indicator of good response to PCRT together with TR.
Collapse
Affiliation(s)
- Jee Yeon Kim
- From the Department of Colon and Rectal Surgery (JYK, IJP, JLL, YSY, CWK, S-BL, CSY, JCK), Department of Pathology (SMH), and Department of Clinical Epidemiology and Biostatistics, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea (JBL)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Clinical Complete Response After Neoadjuvant Therapy in Rectal Cancer: Is Surgery Needed? CURRENT COLORECTAL CANCER REPORTS 2015. [DOI: 10.1007/s11888-015-0299-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
36
|
Patel PM, Harris K, Huerta S. Clinical and molecular diagnosis of pathologic complete response in rectal cancer. Expert Rev Mol Diagn 2015; 15:1505-16. [DOI: 10.1586/14737159.2015.1091728] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
37
|
Nakao T, Iwata T, Hotchi M, Yoshikawa K, Higashijima J, Nishi M, Takasu C, Eto S, Teraoku H, Shimada M. Prediction of response to preoperative chemoradiotherapy and establishment of individualized therapy in advanced rectal cancer. Oncol Rep 2015; 34:1961-7. [PMID: 26260776 DOI: 10.3892/or.2015.4196] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/09/2015] [Indexed: 11/06/2022] Open
Abstract
Preoperative chemoradiotherapy (CRT) has become the standard treatment for patients with locally advanced rectal cancer. However, no specific biomarker has been identified to predict a response to preoperative CRT. The aim of the present study was to assess the gene expression patterns of patients with advanced rectal cancer to predict their responses to preoperative CRT. Fifty-nine rectal cancer patients were subjected to preoperative CRT. Patients were randomly assigned to receive CRT with tegafur/gimeracil/oteracil (S-1 group, n=30) or tegafur-uracil (UFT group, n=29). Gene expression changes were studied with cDNA and miRNA microarray. The association between gene expression and response to CRT was evaluated. cDNA microarray showed that 184 genes were significantly differentially expressed between the responders and the non‑responders in the S-1 group. Comparatively, 193 genes were significantly differentially expressed in the responders in the UFT group. TBX18 upregulation was common to both groups whereas BTNL8, LOC375010, ADH1B, HRASLS2, LOC284232, GCNT3 and ALDH1A2 were significantly differentially lower in both groups when compared with the non-responders. Using miRNA microarray, we found that 7 and 16 genes were significantly differentially expressed between the responders and non-responders in the S-1 and UFT groups, respectively. miR-223 was significantly higher in the responders in the S-1 group and tended to be higher in the responders in the UFT group. The present study identified several genes likely to be useful for establishing individualized therapies for patients with rectal cancer.
Collapse
Affiliation(s)
- Toshihiro Nakao
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Takashi Iwata
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Masanori Hotchi
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Kozo Yoshikawa
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Jun Higashijima
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Masaaki Nishi
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Chie Takasu
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Shohei Eto
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Hiroki Teraoku
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Mitsuo Shimada
- Department of Surgery, Tokushima University, Tokushima 770-8503, Japan
| |
Collapse
|
38
|
Putte DV, Nieuwenhove YV, Willaert W, Pattyn P, Ceelen W. Organ preservation in rectal cancer: current status and future perspectives. COLORECTAL CANCER 2015. [DOI: 10.2217/crc.15.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
With the introduction of population screening initiatives, more patients may be amenable to local, transanal excision (LE) of early-stage rectal cancer. The most important drawback of LE is the risk of understaging node-positive disease. The most powerful predictors of node-positive disease are lymphatic invasion, submucosal invasion depth and width, tumor budding and poor differentiation. Therefore, LE should be reserved for low-risk T1 tumors in those reluctant or unable to undergo major surgery. Neoadjuvant chemoradiation followed by LE for T2 tumors allows adequate local control, and is currently being compared with anterior resection alone in randomized trials. A mere watchful waiting approach has been proposed in clinical complete responders to chemoradiation. However, given the very poor accuracy of current imaging modalities to predict a true pathological complete response, this strategy should not be offered outside of well-controlled trials.
Collapse
Affiliation(s)
- Dirk Vande Putte
- Department of Gastrointestinal Surgery, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | - Yves Van Nieuwenhove
- Department of Gastrointestinal Surgery, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | - Wouter Willaert
- Department of Gastrointestinal Surgery, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | - Piet Pattyn
- Department of Gastrointestinal Surgery, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | - Wim Ceelen
- Department of Gastrointestinal Surgery, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| |
Collapse
|
39
|
Caramés C, Cristóbal I, Moreno V, del Puerto L, Moreno I, Rodriguez M, Marín JP, Correa AV, Hernández R, Zenzola V, Hernández T, León A, Martín JI, Sánchez-Fayos P, García-Olmo D, Rojo F, Goel A, Fernandez-Aceñero MJ, García-Foncillas J. MicroRNA-21 predicts response to preoperative chemoradiotherapy in locally advanced rectal cancer. Int J Colorectal Dis 2015; 30:899-906. [PMID: 25953218 DOI: 10.1007/s00384-015-2231-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/28/2015] [Indexed: 02/04/2023]
Abstract
PURPOSE The treatment of choice for locally advanced rectal cancer is preoperative chemoradiotherapy. Despite half of patients do not respond and suffer unnecessary toxicities and surgery delays, there are no biomarkers to guide preoperative CRT outcome. MicroRNA-21 has been related to acquisition of 5-fluorouracil resistance; however, its potential predictive value of response to preoperative chemoradiotherapy in locally advanced rectal cancer remains unknown. METHODS Nighty-two patients diagnosed with locally advanced rectal cancer who were preoperatively treated with chemoradiotherapy were selected for this study. Moreover, microRNA-21 expression was quantified in formalin-fixed paraffin-embedded biopsies from this cohort, and the results obtained were correlated with clinical and molecular characteristics, pathological response, and outcome. RESULTS MicroRNA-21 was found overexpressed in 77.6% cases, and significantly correlated with tumor grade after preoperative chemoradiotherapy (P = 0.013) and with pathological response (P = 0.013). The odds ratio of having miR-21 overexpression and not getting a respond to chemoradiotherapy resulted in 9.75 CI 2.24 to 42. Sensitivity, specificity, negative predictive values, and positive predictive value were 86.6, 60, 42.8, and 92%, respectively. Multivariate analysis confirmed the clinical significance of miR-21 determining preoperative chemoradiotherapy response. CONCLUSIONS MicroRNA-21 expression efficiently predicts preoperative chemoradiotherapy pathological response in locally advanced rectal cancer.
Collapse
Affiliation(s)
- Cristina Caramés
- Medical Oncology Department, University Hospital "Fundación Jimenez Diaz", Avda. Reyes Católicos-2, 28040, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Molinari C, Matteucci F, Caroli P, Passardi A. Biomarkers and Molecular Imaging as Predictors of Response to Neoadjuvant Chemoradiotherapy in Patients With Locally Advanced Rectal Cancer. Clin Colorectal Cancer 2015; 14:227-38. [PMID: 26170142 DOI: 10.1016/j.clcc.2015.05.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/29/2015] [Indexed: 12/11/2022]
Abstract
Standard treatment of patients with locally advanced rectal cancer (LARC) includes neoadjuvant chemoradiotherapy (NCRT) followed by surgery. Tumor regression after NCRT varies substantially among individuals and pathological complete response is a known prognostic factor for LARC. The identification of a predictive model for response to chemoradiotherapy would help clinicians to identify patients who would probably benefit from multimodal treatment and to perform an early assessment of individual prognosis. Carcinoembryonic antigen has proven to be a good predictor of response in several clinical trials. Other widely studied predictive models in LARC include molecular biomarkers, analyzed at various levels and by different techniques, and molecular imaging, in particular magnetic resonance imaging and positron emission tomography/computed tomography. Although none of the studied markers have been approved in clinical practice, their evaluation in larger, prospective trials and in combined predictive models could be of use to define tailored therapeutic strategies.
Collapse
Affiliation(s)
- Chiara Molinari
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Federica Matteucci
- Diagnostic Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Caroli
- Diagnostic Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| |
Collapse
|
41
|
Di Paolo A, Polillo M, Lastella M, Bocci G, Del Re M, Danesi R. Methods: for studying pharmacogenetic profiles of combination chemotherapeutic drugs. Expert Opin Drug Metab Toxicol 2015; 11:1253-67. [PMID: 26037261 DOI: 10.1517/17425255.2015.1053460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Molecular and genetic analysis of tumors and individuals has led to patient-centered therapies, through the discovery and identification of genetic markers predictive of drug efficacy and tolerability. Present therapies often include a combination of synergic drugs, each of them directed against different targets. Therefore, the pharmacogenetic profiling of tumor masses and patients is becoming a challenge, and several questions may arise when planning a translational study. AREAS COVERED The review presents the different techniques used to stratify oncology patients and to tailor antineoplastic treatments according to individual pharmacogenetic profiling. The advantages of these methodologies are discussed as well as current limits. EXPERT OPINION Facing the rapid technological evolution for genetic analyses, the most pressing issues are the choice of appropriate strategies (i.e., from gene candidate up to next-generation sequencing) and the possibility to replicate study results for their final validation. It is likely that the latter will be the major obstacle in the future. However, the present landscape is opening up new possibilities, overcoming those hurdles that have limited result translation into clinical settings for years.
Collapse
Affiliation(s)
- Antonello Di Paolo
- University of Pisa, Department of Clinical and Experimental Medicine, Via Roma 55, 56126 Pisa , Italy +39 050 2218755 ; +39 050 2218758 ;
| | | | | | | | | | | |
Collapse
|
42
|
Agostini M, Zangrando A, Pastrello C, D'Angelo E, Romano G, Giovannoni R, Giordan M, Maretto I, Bedin C, Zanon C, Digito M, Esposito G, Mescoli C, Lavitrano M, Rizzolio F, Jurisica I, Giordano A, Pucciarelli S, Nitti D. A functional biological network centered on XRCC3: a new possible marker of chemoradiotherapy resistance in rectal cancer patients. Cancer Biol Ther 2015; 16:1160-71. [PMID: 26023803 DOI: 10.1080/15384047.2015.1046652] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Preoperative chemoradiotherapy is widely used to improve local control of disease, sphincter preservation and to improve survival in patients with locally advanced rectal cancer. Patients enrolled in the present study underwent preoperative chemoradiotherapy, followed by surgical excision. Response to chemoradiotherapy was evaluated according to Mandard's Tumor Regression Grade (TRG). TRG 3, 4 and 5 were considered as partial or no response while TRG 1 and 2 as complete response. From pretherapeutic biopsies of 84 locally advanced rectal carcinomas available for the analysis, only 42 of them showed 70% cancer cellularity at least. By determining gene expression profiles, responders and non-responders showed significantly different expression levels for 19 genes (P < 0.001). We fitted a logistic model selected with a stepwise procedure optimizing the Akaike Information Criterion (AIC) and then validated by means of leave one out cross validation (LOOCV, accuracy = 95%). Four genes were retained in the achieved model: ZNF160, XRCC3, HFM1 and ASXL2. Real time PCR confirmed that XRCC3 is overexpressed in responders group and HFM1 and ASXL2 showed a positive trend. In vitro test on colon cancer resistant/susceptible to chemoradioterapy cells, finally prove that XRCC3 deregulation is extensively involved in the chemoresistance mechanisms. Protein-protein interactions (PPI) analysis involving the predictive classifier revealed a network of 45 interacting nodes (proteins) with TRAF6 gene playing a keystone role in the network. The present study confirmed the possibility that gene expression profiling combined with integrative computational biology is useful to predict complete responses to preoperative chemoradiotherapy in patients with advanced rectal cancer.
Collapse
Key Words
- CEA, carcinoembryonic antigen
- CRT, Chemoradiotherapy
- DSB, Double-strand breaks
- Gy, Gray
- HT, High throughput
- PPI, Protein-protein interaction
- RC, Rectal cancer
- RIN, RNA integrity number
- SNP, Single nucleotide polymorphism
- SSB, Single-strand breaks
- XRCC3
- biological network
- integrated approach
- mRNA, mRNA
- microarray
- pCRT, Preoperative chemoradiotherapy
- preoperative chemoradiotherapy
- rectal cancer
- siRNA, Small interfering RNA
- treatment response
Collapse
Affiliation(s)
- Marco Agostini
- a Department of Surgical , Oncological and Gastroenterological Sciences ; Section of Surgery ; University of Padova ; Padua , Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Predictive and prognostic biomarkers for neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Crit Rev Oncol Hematol 2015; 96:67-80. [PMID: 26032919 DOI: 10.1016/j.critrevonc.2015.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/03/2015] [Accepted: 05/05/2015] [Indexed: 02/08/2023] Open
Abstract
Locally advanced rectal cancer is regularly treated with trimodality therapy consisting of neoadjuvant chemoradiation, surgery and adjuvant chemotherapy. There is a need for biomarkers to assess treatment response, and aid in stratification of patient risk to adapt and personalise components of the therapy. Currently, pathological stage and tumour regression grade are used to assess response. Experimental markers include proteins involved in cell proliferation, apoptosis, angiogenesis, the epithelial to mesenchymal transition and microsatellite instability. As yet, no single marker is sufficiently robust to have clinical utility. Microarrays that screen a tumour for multiple promising candidate markers, gene expression and microRNA profiling will likely have higher yield and it is expected that a combination or panel of markers would prove most useful. Moving forward, utilising serial samples of circulating tumour cells or circulating nucleic acids can potentially allow us to demonstrate tumour heterogeneity, document mutational changes and subsequently measure treatment response.
Collapse
|
44
|
Senetta R, Duregon E, Sonetto C, Spadi R, Mistrangelo M, Racca P, Chiusa L, Munoz FH, Ricardi U, Arezzo A, Cassenti A, Castellano I, Papotti M, Morino M, Risio M, Cassoni P. YKL-40/c-Met expression in rectal cancer biopsies predicts tumor regression following neoadjuvant chemoradiotherapy: a multi-institutional study. PLoS One 2015; 10:e0123759. [PMID: 25875173 PMCID: PMC4398550 DOI: 10.1371/journal.pone.0123759] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/21/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Neoadjuvant chemo-radiotherapy (CRT) followed by surgical resection is the standard treatment for locally advanced rectal cancer, although complete tumor pathological regression is achieved in only up to 30% of cases. A clinicopathological and molecular predictive stratification of patients with advanced rectal cancer is still lacking. Here, c-Met and YKL-40 have been studied as putative predictors of CRT response in rectal cancer, due to their reported involvement in chemoradioresistance in various solid tumors. MATERIAL AND METHODS A multicentric study was designed to assess the role of c-Met and YKL-40 expression in predicting chemoradioresistance and to correlate clinical and pathological features with CRT response. Immunohistochemistry and fluorescent in situ hybridization for c-Met were performed on 81 rectal cancer biopsies from patients with locally advanced rectal adenocarcinoma. All patients underwent standard (50.4 gy in 28 fractions + concurrent capecitabine 825 mg/m2) neoadjuvant CRT or the XELOXART protocol. CRT response was documented on surgical resection specimens and recorded as tumor regression grade (TRG) according to the Mandard criteria. RESULTS A significant correlation between c-Met and YKL-40 expression was observed (R = 0.43). The expressions of c-Met and YKL-40 were both significantly associated with a lack of complete response (86% and 87% of c-Met and YKL-40 positive cases, p< 0.01 and p = 0.006, respectively). Thirty of the 32 biopsies co-expressing both markers had partial or absent tumor response (TRG 2-5), strengthening their positive predictive value (94%). The exclusive predictive role of YKL-40 and c-Met was confirmed using a multivariate analysis (p = 0.004 and p = 0.007 for YKL-40 and c-Met, respectively). TRG was the sole morphological parameter associated with poor outcome. CONCLUSION c-Met and YKL-40 expression is a reliable predictor of partial/absent response to neoadjuvant CRT in rectal cancer. Targeted therapy protocols could take advantage of prior evaluations of c-MET and YKL-40 expression levels to increase therapeutic efficacy.
Collapse
Affiliation(s)
- Rebecca Senetta
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Cristina Sonetto
- SSCVD Colorectal Cancer Unit, City of Health and Science Hospital of Turin, Turin, Italy
| | - Rossella Spadi
- SSCVD Colorectal Cancer Unit, City of Health and Science Hospital of Turin, Turin, Italy
| | - Massimiliano Mistrangelo
- Digestive and Colorectal Surgery, Centre of Minimal Invasive Surgery, University of Turin, Turin, Italy
| | - Patrizia Racca
- SSCVD Colorectal Cancer Unit, City of Health and Science Hospital of Turin, Turin, Italy
| | - Luigi Chiusa
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | | | - Alberto Arezzo
- Digestive and Colorectal Surgery, Centre of Minimal Invasive Surgery, University of Turin, Turin, Italy
| | - Adele Cassenti
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Mauro Papotti
- Department of Oncology, University of Turin, Turin, Italy
| | - Mario Morino
- Digestive and Colorectal Surgery, Centre of Minimal Invasive Surgery, University of Turin, Turin, Italy
| | - Mauro Risio
- Candiolo Cancer Institute—FPO (Fondazione del Piemonte per l'0ncologia), IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), Candiolo, Turin, Italy
| | - Paola Cassoni
- Department of Medical Sciences, University of Turin, Turin, Italy
- * E-mail:
| |
Collapse
|
45
|
Saab AM, Guerrini A, Zeino M, Wiench B, Rossi D, Gambari R, Sacchetti G, Greten HJ, Efferth T. Laurus nobilisL. Seed Extract Reveals Collateral Sensitivity in Multidrug-Resistant P-Glycoprotein-Expressing Tumor Cells. Nutr Cancer 2015; 67:664-75. [DOI: 10.1080/01635581.2015.1019632] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
46
|
Comprehensive evaluation of the effectiveness of gene expression signatures to predict complete response to neoadjuvant chemoradiotherapy and guide surgical intervention in rectal cancer. Cancer Genet 2015; 208:319-26. [PMID: 25963525 DOI: 10.1016/j.cancergen.2015.03.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/23/2015] [Accepted: 03/11/2015] [Indexed: 12/24/2022]
Abstract
Neoadjuvant chemoradiotherapy (nCRT) may lead to complete tumor regression in rectal cancer patients. Prediction of complete response to nCRT may allow a personalized management of rectal cancer and spare patients from unnecessary radical total mesorectal excision with or without sphincter preservation. To identify a gene expression signature capable of predicting complete pathological response (pCR) to nCRT, we performed a gene expression analysis in 25 pretreatment biopsies from patients who underwent 5FU-based nCRT using RNA-Seq. A supervised learning algorithm was used to identify expression signatures capable of predicting pCR, and the predictive value of these signatures was validated using independent samples. We also evaluated the utility of previously published signatures in predicting complete response in our cohort. We identified 27 differentially expressed genes between patients with pCR and patients with incomplete responses to nCRT. Predictive gene signatures using subsets of these 27 differentially expressed genes peaked at 81.8% accuracy. However, signatures with the highest sensitivity showed poor specificity, and vice-versa, when applied in an independent set of patients. Testing previously published signatures on our cohort also showed poor predictive value. Our results indicate that currently available predictive signatures are highly dependent on the sample set from which they are derived, and their accuracy is not superior to current imaging and clinical parameters used to assess response to nCRT and guide surgical intervention.
Collapse
|
47
|
Lopes-Ramos CM, Habr-Gama A, Quevedo BDS, Felício NM, Bettoni F, Koyama FC, Asprino PF, Galante PA, Gama-Rodrigues J, Camargo AA, Perez RO, Parmigiani RB. Overexpression of miR-21-5p as a predictive marker for complete tumor regression to neoadjuvant chemoradiotherapy in rectal cancer patients. BMC Med Genomics 2014; 7:68. [PMID: 25496125 PMCID: PMC4279677 DOI: 10.1186/s12920-014-0068-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 11/28/2014] [Indexed: 12/16/2022] Open
Abstract
Background Neoadjuvant chemoradiotherapy (nCRT) followed by radical surgery is the preferred treatment strategy for locally advanced rectal cancer. However, complete tumor regression is observed in a significant proportion of patients after nCRT, making them ideal candidates for alternative treatment strategies to this considerably morbid procedure. Identification of such patients based on clinical findings (complete clinical response - cCR) is difficult mainly because it relies on subjective clinical and imaging studies. Our goal was to identify biomarkers capable of predicting complete response to nCRT. Methods We analyzed miRNA expression profile using deep sequencing in rectal tumor biopsies prior to nCRT. Differential expression was investigated by EdgeR for a training (n = 27) and a validation (n = 16) set of patients to identify miRNAs associated with treatment response (complete vs. incomplete). In vitro experiments with two cancer cell lines were also performed in order to evaluate the possible role of miRNAs on response to nCRT. Results We found 4 miRNAs differentially expressed between complete and incomplete responders to nCRT. In addition, validation was performed using an independent group of patients and miR-21-5p was confirmed as being overexpressed in complete responders. Overall sensitivity and specificity of miR-21-5p expression in predicting complete response to nCRT was 78% and 86% respectively. Interestingly, in a subset of patients with cCR followed by early local recurrence, the expression level of miR-21-5p was considerably low, similarly to incomplete responders. We also found SATB1, a miR-21-5p target gene and known multidrug resistance gene, whose expression was inversely correlated with miR-21-5p expression. Finally, we performed functional experiments and showed that miR-21-5p and SATB1 may be directly involved with poor response to nCRT in rectal cancer patients. Conclusions This study suggests miR-21-5p as a promising predictive biomarker, which should aid in the selection of patients with cCR to nCRT that potentially could be spared from radical surgery. Electronic supplementary material The online version of this article (doi:10.1186/s12920-014-0068-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camila Miranda Lopes-Ramos
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, Brazil. .,Fundação Antônio Prudente, São Paulo, Brazil.
| | - Angelita Habr-Gama
- Angelita & Joaquim Gama Institute, São Paulo, Brazil. .,University of São Paulo School of Medicine, São Paulo, Brazil.
| | | | - Natália Mariana Felício
- Angelita & Joaquim Gama Institute, São Paulo, Brazil. .,Hospital Alemão Oswaldo Cruz, São Paulo, Brazil.
| | - Fabiana Bettoni
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, Brazil.
| | | | | | | | - Joaquim Gama-Rodrigues
- Angelita & Joaquim Gama Institute, São Paulo, Brazil. .,University of São Paulo School of Medicine, São Paulo, Brazil.
| | - Anamaria Aranha Camargo
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo, Brazil. .,Ludwig Institute for Cancer Research, São Paulo, Brazil.
| | - Rodrigo Oliva Perez
- Angelita & Joaquim Gama Institute, São Paulo, Brazil. .,University of São Paulo School of Medicine, São Paulo, Brazil. .,Ludwig Institute for Cancer Research, São Paulo, Brazil.
| | | |
Collapse
|
48
|
Meng X, Huang Z, Wang R, Yu J. Prediction of response to preoperative chemoradiotherapy in patients with locally advanced rectal cancer. Biosci Trends 2014; 8:11-23. [PMID: 24647108 DOI: 10.5582/bst.8.11] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Preoperative chemoradiotherapy (CRT) combined with surgery has become a standard treatment strategy for patients with locally advanced rectal cancer (LARC). The pathological response is an important prognostic factor for LARC. The variety of tumor responses has increased the need to find a useful predictive model for the response to CRT to identify patients who will really benefit from this multimodal treatment. Magnetic resonance imaging (MRI), positron emission tomography-computed tomography (PET-CT), serum carcinoembryogenic antigen (CEA), molecular biomarkers analyzed by immunohistochemistry and gene expression profiling are the most used predictive models in LARC. The majority of predictors have yielded encouraging results, but there is still controversy. Diffusion-weighted MRI may be the best model to detect the dynamic changes of rectal cancer and predict the response at an early stage. Gene expression profiling and single nucleotide polymorphisms hold considerable promise to unveil the underlying complex genetics of response to CRT. Because each parameter has its own inherent shortcomings, combined models may be the future trend to predict the response.
Collapse
Affiliation(s)
- Xiangjiao Meng
- Department of Radiation Oncology and Key Laboratory of Radiation Oncology of Shandong Province, Department of Radiation Oncology of Shandong Cancer Hospital and Institute
| | | | | | | |
Collapse
|
49
|
Dou X, Wang RB, Meng XJ, Yan HJ, Jiang SM, Zhu KL, Xu XQ, Chen D, Song XR, Mu DB. PDCD4 as a predictor of sensitivity to neoadjuvant chemoradiotherapy in locally advanced rectal cancer patients. Asian Pac J Cancer Prev 2014; 15:825-30. [PMID: 24568503 DOI: 10.7314/apjcp.2014.15.2.825] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE The purpose of this study was to examine the role of programmed cell death 4 (PDCD4) expression in predicting tumor response to neoadjuvant chemoradiotherapy and outcomes for patients with locally advanced rectal cancer. METHODS Clinicopathological factors and expression of PDCD4 were evaluated in 92 patients with LARC treated with nCRT. After the completion of therapy, 4 cases achieved clinical complete response (cCR), and thus the remaining 88 patients underwent a standardized total mesorectal excision procedure. There were 38 patients (41.3%) with a good response (TRG 3-4) and 54 (58.7%) with a poor one (TRG 0-2). RESULTS Immunohistochemical staining analyses showed that patients with high expression of PDCD4 were more sensitive to nCRT than those with low PDCD4 expression (P=0.02). High PDCD4 expression before nCRT and good response (TRG3-4) were significantly associated with improved 5-year disease-free survival and 5-year overall survival (P<0.05). Multivariate analysis demonstrated that the pretreatment PDCD4 expression was an independent prognostic factor. CONCLUSION Our study demonstrated that high expression of PDCD4 protein is a useful predictive factor for good tumor response to nCRT and good outcomes in patients with LARC.
Collapse
Affiliation(s)
- Xue Dou
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, University of Jinan, Jinan, China E-mail :
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Habr-Gama A, Perez RO. Immediate surgery or clinical follow-up after a complete clinical response? Recent Results Cancer Res 2014; 203:203-10. [PMID: 25103007 DOI: 10.1007/978-3-319-08060-4_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neoadjuvant chemoradiation (CRT) is considered as one of the preferred treatment strategies for patients with locally advanced rectal cancer. This treatment strategy may lead to significant tumor regression, ultimately leading to complete pathological response in up to 42% of patients. Assessment of tumor response following CRT and before radical surgery may identify patients with complete clinical response that could be managed non operatively with strict follow-up (Watch & Wait Strategy).
Collapse
Affiliation(s)
- Angelita Habr-Gama
- University of São Paulo School of Medicine, Rua Manoel da Nóbrega 1564, São Paulo, SP, 04001-005, Brazil,
| | | |
Collapse
|