1
|
Schöllnast H. Mucosal Linear Enhancement after Neoadjuvant Chemoradiation Therapy for Rectal Cancer. Radiology 2024; 312:e241483. [PMID: 39136562 DOI: 10.1148/radiol.241483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Affiliation(s)
- Helmut Schöllnast
- From the Institute of Radiology, LKH Graz II, Goestinger Strasse 22, 8020 Graz, Austria; and Department of Radiology, Division of General Radiology, Medical University of Graz, Graz, Austria
| |
Collapse
|
2
|
Tang YL, Li DD, Duan JY, Wang X. Prognostic analysis of rectal cancer patients after neoadjuvant chemoradiotherapy: different prognostic factors in patients with different TRGs. Int J Colorectal Dis 2024; 39:93. [PMID: 38896374 PMCID: PMC11186864 DOI: 10.1007/s00384-024-04666-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE The extent of tumor regression varies widely among locally advanced rectal cancer (LARC) patients who receive neoadjuvant chemoradiotherapy (NCRT) followed by total mesorectal excision (TME). The purpose of this retrospectively study is to assess prognostic factors in LARC patients with NCRT, and further to analyze survival outcomes in patients with different tumor regression grades (TRGs). METHODS This study includes LARC patients who underwent NCRT and TME at our institution. We retrospectively analyzed the clinicopathological characteristics and survival of all patients, and performed subgroup analysis for patients with different TRGs. Survival differences were compared using the Kaplan-Meier method and the log rank test. Additionally, a multiple Cox proportional hazard model was used to identify independent prognostic factors. RESULTS The study included 393 patients, with 21.1%, 26.5%, 45.5%, and 6.9% achieving TRG 0, TRG 1, TRG 2, and TRG 3, respectively. The overall survival (OS) rate and disease-free survival (DFS) rate for all patients were 89.4% and 70.7%, respectively. Patients who achieved TRG 0-3 had different 5-year OS rates (96.9%, 91.1%, 85.2%, and 68.8%, P = 0.001) and 5-year DFS rates (80.8%, 72.4%, 67.0%, 55.8%, P = 0.031), respectively. Multivariate analyses showed that the neoadjuvant rectal (NAR) score was an independent prognostic indicator for both overall survival (OS) (HR = 4.040, 95% CI = 1.792-9.111, P = 0.001) and disease-free survival (DFS) (HR = 1.971, 95% CI = 1.478-2.628, P ˂ 0.001). In the subgroup analyses, the NAR score was found to be associated with DFS in patients with TRG 1 and TRG 2. After conducting multivariate analysis, it was found that ypT stage was a significant predictor of DFS for TRG 1 patients (HR = 4.384, 95% CI = 1.721-11.168, P = 0.002). On the other hand, ypN stage was identified as the dominant prognostic indicator of DFS for TRG 2 patients (HR = 2.795, 95% CI = 1.535-5.091, P = 0.001). However, none of these characteristics was found to be correlated with survival in patients with TRG 0 or TRG 3. CONCLUSION NAR score, in particular, appears to be the most powerful prognostic factor. It is important to consider various prognostic predictors for patients with different TRGs.
Collapse
Affiliation(s)
- Yuan-Ling Tang
- Division of Abdominal Tumor Multimodality Treatment, Department of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital of Sichuan University, No. 37 Guoxue Lane, Wuhou District, Chengdu City, 610041, Sichuan Province, China
| | - Dan-Dan Li
- Division of Abdominal Tumor Multimodality Treatment, Department of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital of Sichuan University, No. 37 Guoxue Lane, Wuhou District, Chengdu City, 610041, Sichuan Province, China
| | - Jia-Yu Duan
- Division of Abdominal Tumor Multimodality Treatment, Department of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital of Sichuan University, No. 37 Guoxue Lane, Wuhou District, Chengdu City, 610041, Sichuan Province, China
| | - Xin Wang
- Division of Abdominal Tumor Multimodality Treatment, Department of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital of Sichuan University, No. 37 Guoxue Lane, Wuhou District, Chengdu City, 610041, Sichuan Province, China.
| |
Collapse
|
3
|
Hillson LVS, McCulloch AK, Edwards J, Dunne PD, O'Cathail SM, Roxburgh CS. Radiation-induced changes in gene expression in rectal cancer specimens. Clin Transl Oncol 2024; 26:1419-1428. [PMID: 38243085 PMCID: PMC11108951 DOI: 10.1007/s12094-023-03361-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/26/2023] [Indexed: 01/21/2024]
Abstract
PURPOSE The standard-of-care for locally advanced rectal cancer is radiotherapy-based neoadjuvant therapy followed by surgical resection. This article reviews the evidence of molecular changes at the transcriptome level induced through radiotherapy in rectal cancer. METHODS The PubMed search "(radiation OR radiotherapy) cancer (transcriptome OR "gene expression") rectal" was used. The studies taken forward utilised gene-expression data on both pre-treatment and post-treatment rectal adenocarcinoma biospecimens from patients treated with RT-based neoadjuvant strategies. RESULTS Twelve publications met the review criteria. There was variation in approaches in terms of design, patient population, cohort size, timing of the post-radiotherapy sampling and method of measuring gene expression. Most of the post-treatment biospecimen retrievals were at resection. The literature indicates a broad upregulation of immune activity through radiotherapy using gene-expression data. CONCLUSION Future studies would benefit from standardised prospective approaches to sampling to enable the inclusion of timepoints relevant to the tumour and immune response.
Collapse
Affiliation(s)
- Lily Victoria Sarah Hillson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
| | - Ashley Kathryn McCulloch
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Joanne Edwards
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Philip David Dunne
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Sean Michael O'Cathail
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Campbell Stuart Roxburgh
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
- Academic Unit of Surgery, Glasgow Royal Infirmary, Glasgow, UK
| |
Collapse
|
4
|
Zwart WH, Temmink SJD, Hospers GAP, Marijnen CAM, Putter H, Nagtegaal ID, Blomqvist L, Kranenbarg EMK, Roodvoets AGH, Martling A, van de Velde CJH, Glimelius B, Peeters KCMJ, van Etten B, Nilsson PJ. Oncological outcomes after a pathological complete response following total neoadjuvant therapy or chemoradiotherapy for high-risk locally advanced rectal cancer in the RAPIDO trial. Eur J Cancer 2024; 204:114044. [PMID: 38636289 DOI: 10.1016/j.ejca.2024.114044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/25/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND A pathological complete response (pCR) following chemoradiation (CRT) or short-course radiotherapy (scRT) leads to a favourable prognosis in patients with rectal cancer. Total neo-adjuvant therapy (TNT) doubles the pCR rate, but it is unknown whether oncological outcomes remain favourable and whether the same characteristics are associated with pCR as after CRT. METHODS Comparison between patients with pCR in the RAPIDO trial in the experimental [EXP] (scRT, chemotherapy, surgery, as TNT) and standard-of-care treatment [STD] (CRT, surgery, postoperative chemotherapy depending on hospital policy) groups. Primary and secondary outcomes were time-to-recurrence (TTR), overall survival (OS) and association between patient, tumour, and treatment characteristics and pCR. RESULTS Among patients with a resection within six months after preoperative treatment, 120/423 (28%) [EXP] and 57/398 (14%) [STD] achieved a pCR. Following pCR, 5-year cumulative TTR and OS rates in the EXP and STD arms were 8% vs. 7% (hazard ratio 1.04, 95%CI 0.32-3.38) and 94% vs. 93% (hazard ratio 1.41, 95%CI 0.51-3.92), respectively. Besides the EXP treatment (odds ratio 2.70, 95%CI 1.83-3.97), pre-treatment carcinoembryonic antigen (CEA) <5, pre-treatment tumour size <40 mm and cT2 were associated with pCR. Distance from the anal verge was the only characteristic with a statistically significant difference in association with pCR between the EXP and STD treatment (Pinteraction=0.042). pCR rates did not increase with prolonged treatment time. CONCLUSIONS The doubled pCR rate of TNT compared to CRT results in similar oncological outcomes. Characteristics associated with pCR are the EXP treatment, normal CEA, and small tumour size.
Collapse
Affiliation(s)
- Wouter H Zwart
- University Medical Center Groningen, Department of Medical Oncology, Groningen, the Netherlands.
| | - Sofieke J D Temmink
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden
| | - Geke A P Hospers
- University Medical Center Groningen, Department of Medical Oncology, Groningen, the Netherlands
| | - Corrie A M Marijnen
- Netherlands Cancer Institute, Department of Radiation Oncology, Amsterdam, the Netherlands; Leiden University Medical Center, Department of Radiation Oncology, Leiden, the Netherlands
| | - Hein Putter
- Leiden University Medical Center, Department of Biomedical Data Sciences, Leiden, the Netherlands
| | - Iris D Nagtegaal
- Radboud University Medical Centre, Department of Pathology, Nijmegen, the Netherlands
| | - Lennart Blomqvist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Annet G H Roodvoets
- Leiden University Medical Center, Department of Surgery, Leiden, the Netherlands
| | - Anna Martling
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden
| | | | - Bengt Glimelius
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Koen C M J Peeters
- Leiden University Medical Center, Department of Surgery, Leiden, the Netherlands
| | - Boudewijn van Etten
- University Medical Center Groningen, Department of Surgery, Groningen, the Netherlands
| | - Per J Nilsson
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden
| |
Collapse
|
5
|
Chen KA, Goffredo P, Butler LR, Joisa CU, Guillem JG, Gomez SM, Kapadia MR. Prediction of Pathologic Complete Response for Rectal Cancer Based on Pretreatment Factors Using Machine Learning. Dis Colon Rectum 2024; 67:387-397. [PMID: 37994445 PMCID: PMC11186794 DOI: 10.1097/dcr.0000000000003038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
BACKGROUND Pathologic complete response after neoadjuvant therapy is an important prognostic indicator for locally advanced rectal cancer and may give insights into which patients might be treated nonoperatively in the future. Existing models for predicting pathologic complete response in the pretreatment setting are limited by small data sets and low accuracy. OBJECTIVE We sought to use machine learning to develop a more generalizable predictive model for pathologic complete response for locally advanced rectal cancer. DESIGN Patients with locally advanced rectal cancer who underwent neoadjuvant therapy followed by surgical resection were identified in the National Cancer Database from years 2010 to 2019 and were split into training, validation, and test sets. Machine learning techniques included random forest, gradient boosting, and artificial neural network. A logistic regression model was also created. Model performance was assessed using an area under the receiver operating characteristic curve. SETTINGS This study used a national, multicenter data set. PATIENTS Patients with locally advanced rectal cancer who underwent neoadjuvant therapy and proctectomy. MAIN OUTCOME MEASURES Pathologic complete response defined as T0/xN0/x. RESULTS The data set included 53,684 patients. Pathologic complete response was experienced by 22.9% of patients. Gradient boosting showed the best performance with an area under the receiver operating characteristic curve of 0.777 (95% CI, 0.773-0.781), compared with 0.684 (95% CI, 0.68-0.688) for logistic regression. The strongest predictors of pathologic complete response were no lymphovascular invasion, no perineural invasion, lower CEA, smaller size of tumor, and microsatellite stability. A concise model including the top 5 variables showed preserved performance. LIMITATIONS The models were not externally validated. CONCLUSIONS Machine learning techniques can be used to accurately predict pathologic complete response for locally advanced rectal cancer in the pretreatment setting. After fine-tuning a data set including patients treated nonoperatively, these models could help clinicians identify the appropriate candidates for a watch-and-wait strategy. See Video Abstract . EL CNCER DE RECTO BASADA EN FACTORES PREVIOS AL TRATAMIENTO MEDIANTE EL APRENDIZAJE AUTOMTICO ANTECEDENTES:La respuesta patológica completa después de la terapia neoadyuvante es un indicador pronóstico importante para el cáncer de recto localmente avanzado y puede dar información sobre qué pacientes podrían ser tratados de forma no quirúrgica en el futuro. Los modelos existentes para predecir la respuesta patológica completa en el entorno previo al tratamiento están limitados por conjuntos de datos pequeños y baja precisión.OBJETIVO:Intentamos utilizar el aprendizaje automático para desarrollar un modelo predictivo más generalizable para la respuesta patológica completa para el cáncer de recto localmente avanzado.DISEÑO:Los pacientes con cáncer de recto localmente avanzado que se sometieron a terapia neoadyuvante seguida de resección quirúrgica se identificaron en la Base de Datos Nacional del Cáncer de los años 2010 a 2019 y se dividieron en conjuntos de capacitación, validación y prueba. Las técnicas de aprendizaje automático incluyeron bosque aleatorio, aumento de gradiente y red neuronal artificial. También se creó un modelo de regresión logística. El rendimiento del modelo se evaluó utilizando el área bajo la curva característica operativa del receptor.ÁMBITO:Este estudio utilizó un conjunto de datos nacional multicéntrico.PACIENTES:Pacientes con cáncer de recto localmente avanzado sometidos a terapia neoadyuvante y proctectomía.PRINCIPALES MEDIDAS DE VALORACIÓN:Respuesta patológica completa definida como T0/xN0/x.RESULTADOS:El conjunto de datos incluyó 53.684 pacientes. El 22,9% de los pacientes experimentaron una respuesta patológica completa. El refuerzo de gradiente mostró el mejor rendimiento con un área bajo la curva característica operativa del receptor de 0,777 (IC del 95%: 0,773 - 0,781), en comparación con 0,684 (IC del 95%: 0,68 - 0,688) para la regresión logística. Los predictores más fuertes de respuesta patológica completa fueron la ausencia de invasión linfovascular, la ausencia de invasión perineural, un CEA más bajo, un tamaño más pequeño del tumor y la estabilidad de los microsatélites. Un modelo conciso que incluye las cinco variables principales mostró un rendimiento preservado.LIMITACIONES:Los modelos no fueron validados externamente.CONCLUSIONES:Las técnicas de aprendizaje automático se pueden utilizar para predecir con precisión la respuesta patológica completa para el cáncer de recto localmente avanzado en el entorno previo al tratamiento. Después de realizar ajustes en un conjunto de datos que incluye pacientes tratados de forma no quirúrgica, estos modelos podrían ayudar a los médicos a identificar a los candidatos adecuados para una estrategia de observar y esperar. (Traducción-Dr. Ingrid Melo ).
Collapse
Affiliation(s)
- Kevin A Chen
- Division of Gastrointestinal Surgery, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, 4038 Burnett Womack Building, Chapel Hill, NC 27599
| | - Paolo Goffredo
- Division of Colorectal Surgery, Department of Surgery, University of Minnesota, Minneapolis, MN, 420 Delaware St SE, Minneapolis, MN 55455
| | - Logan R Butler
- Division of Gastrointestinal Surgery, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, 4038 Burnett Womack Building, Chapel Hill, NC 27599
| | - Chinmaya U Joisa
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 10202C Mary Ellen Jones Building, Chapel Hill, NC, 27599
| | - Jose G Guillem
- Division of Gastrointestinal Surgery, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, 4038 Burnett Womack Building, Chapel Hill, NC 27599
| | - Shawn M Gomez
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 10202C Mary Ellen Jones Building, Chapel Hill, NC, 27599
| | - Muneera R Kapadia
- Division of Gastrointestinal Surgery, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, 4038 Burnett Womack Building, Chapel Hill, NC 27599
| |
Collapse
|
6
|
Flom E, Schultz KS, Pantel HJ, Leeds IL. The Predictors of Complete Pathologic Response in Rectal Cancer during the Total Neoadjuvant Therapy Era: A Systematic Review. Cancers (Basel) 2023; 15:5853. [PMID: 38136397 PMCID: PMC10742121 DOI: 10.3390/cancers15245853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
The modern rectal cancer treatment paradigm offers additional opportunities for organ preservation, most notably via total neoadjuvant therapy (TNT) and consideration for a watch-and-wait (WW) surveillance-only approach. A major barrier to widespread implementation of a WW approach to rectal cancer is the potential discordance between a clinical complete response (cCR) and a pathologic complete response (pCR). In the pre-TNT era, the identification of predictors of pCR after neoadjuvant therapy had been previously studied. However, the last meta-analysis to assess the summative evidence on this important treatment decision point predates the acceptance and dissemination of TNT strategies. The purpose of this systematic review was to assess preoperative predictors of pCR after TNT to guide the ideal selection criteria for WW in the current era. An exhaustive literature review was performed and the electronic databases Embase, Ovid, MEDLINE, PubMed, and Cochrane were comprehensively searched up to 27 June 2023. Search terms and their combinations included "rectal neoplasms", "total neoadjuvant therapy", and "pathologic complete response". Only studies in English were included. Randomized clinical trials or prospective/retrospective cohort studies of patients with clinical stage 2 or 3 rectal adenocarcinoma who underwent at least 8 weeks of neoadjuvant chemotherapy in addition to chemoradiotherapy with pCR as a measured study outcome were included. In this systematic review, nine studies were reviewed for characteristics positively or negatively associated with pCR or tumor response after TNT. The results were qualitatively grouped into four categories: (1) biochemical factors; (2) clinical factors; (3) patient demographics; and (4) treatment sequence for TNT. The heterogeneity of studies precluded meta-analysis. The level of evidence was low to very low. There is minimal data to support any clinicopathologic factors that either have a negative or positive relationship to pCR and tumor response after TNT. Additional data from long-term trials using TNT is critical to better inform those considering WW approaches following a cCR.
Collapse
Affiliation(s)
- Emily Flom
- Division of Colon and Rectal Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kurt S Schultz
- Division of Colon and Rectal Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Haddon J Pantel
- Division of Colon and Rectal Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Ira L Leeds
- Division of Colon and Rectal Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
7
|
Sawada N, Mukai S, Takehara Y, Misawa M, Kudo T, Hayashi T, Wakamura K, Enami Y, Miyachi H, Baba T, Ishida F, Kudo SE. The "Watch and Wait" Method After Chemoradiotherapy for Rectal Cancer Requiring Abdominoperineal Resection. Indian J Surg Oncol 2023; 14:765-772. [PMID: 38187830 PMCID: PMC10767130 DOI: 10.1007/s13193-023-01831-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 09/29/2023] [Indexed: 01/09/2024] Open
Abstract
The present study examined the therapeutic effects of preoperative neoadjuvant chemoradiation therapy (NACRT) and predictive factors for complete clinical remission, compared the prognosis and costs of abdominoperineal resection (APR) and the "watch and wait" method (WW), and evaluated the usefulness of WW. In our department, patients with stage II-III lower rectal cancer requiring APR receive NACRT. NACRT was performed as a preoperative treatment (52 Gy + S-1: 80-120 mg/day × 25 days). Eight weeks after the completion of NACRT, rectal examination, endoscopic, computed tomography, and magnetic resonance imaging findings were evaluated to assess its therapeutic effects. APR was indicated for patients in whom endoscopic findings suggested a residual tumor in which a deep ulcer or marginal swelling remained or lymph node metastasis. However, WW was selected for patients who refused APR after informed consent was obtained. In the APR and WW groups, 5- and 20-year treatment costs after CRT were calculated using the Medical Fee Points of Japan in 2020. No significant differences were observed in 3-year disease-free survival rates for either parameter between the two groups. Regarding expenses, treatment costs were lower in the WW group than in the APR group. Organ preservation using active surveillance with CRT for rectal cancer requiring APR is feasible with the achievement of endoluminal complete remission.
Collapse
Affiliation(s)
- Naruhiko Sawada
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Shumpei Mukai
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Yusuke Takehara
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Masashi Misawa
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Toyoki Kudo
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Takemasa Hayashi
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Kunihiko Wakamura
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Yuta Enami
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Hideyuki Miyachi
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Toshiyuki Baba
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Fumio Ishida
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| | - Shin-ei Kudo
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1, Tsuzuki-Ku, Chigasakichuo, Yokohama City, Kanagawa, 224-8503 Japan
| |
Collapse
|
8
|
Liu J, Lee JYT, Bedrikovetski S, Traeger L, Moore JW, Perry JL, Kroon HM, Sammour T. Clinical predictors of rectal cancer response after neo-adjuvant (Chemo)Radiotherapy in Australia and New Zealand: Analysis of the Bi-National Colorectal Cancer Audit (BCCA). EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:107070. [PMID: 37717474 DOI: 10.1016/j.ejso.2023.107070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/22/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023]
Abstract
Pathological complete response (pCR) is observed in 11-26% of locally advanced rectal cancers undergoing neoadjuvant chemoradiotherapy (nCRT). This study aims to determine pCR rates and clinicopathological predictors in the Australian and New Zealand (ANZ) cohort. The Bi-National Colorectal Cancer Audit (BCCA) was interrogated for all rectal cancer patients who underwent nCRT prior to surgical resection between 2007 and 2020. Patients were divided in two groups: pCR (AJCC tumour regression grade 0) and partial/no response (pPR, regression grade 1,2 or 3). In total, 3230 patients were included. Rates of pCR and pPR were 704 (21.8%) and 2526 (78.2%), respectively. Long-course nCRT (p < 0.0001), lower clinical tumour stage (cT; p < 0.0001), and nodal stage (cN; p = 0.003) were associated with pCR on univariate analysis. On multivariable analysis, cN0 stage and long-course nCRT remained independent factors for a pCR. Awareness of these predictors provides valuable information when counseling patients regarding prognosis and treatment options.
Collapse
Affiliation(s)
- Jianliang Liu
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - Justin Y T Lee
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Sergei Bedrikovetski
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Luke Traeger
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - James W Moore
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Joanne L Perry
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Hidde M Kroon
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Tarik Sammour
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
9
|
Kubota S, Miura T, Wakiya T, Yoshizawa T, Goto S, Morohashi H, Sakamoto Y, Tatara Y, Kijima H, Hakamada K. Exploration of Malignant Characteristics in Neoadjuvant Chemotherapy-Resistant Rectal Cancer, Focusing on Extramural Lesions. Ann Surg Oncol 2023; 30:7612-7623. [PMID: 37548833 PMCID: PMC10562322 DOI: 10.1245/s10434-023-13928-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/28/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Extramural vascular invasion (EMVI) and tumor deposits (TD) are poor prognostic factors in rectal cancer (RC), especially when resistant to neoadjuvant chemotherapy (NAC). We aimed to define differential expression in NAC responders and non-responders with concomitant EMVI and TD. METHODS From 52 RC surgical patients, post-NAC resected specimens were extracted, comprising two groups: cases with residual EMVI and TD (NAC-resistant) and cases without (NAC-effective). Proteomic analysis was conducted to define differential protein expression in the two groups. To validate the findings, immunohistochemistry was performed in another cohort that included 58 RC surgical patients. Based on the findings, chemosensitivity and prognosis were compared. RESULTS The NAC-resistant group was associated with a lower 3-year disease-free survival rate than the NAC-effective group (p = 0.041). Discriminative proteins in the NAC-resistant group were highly associated with the sulfur metabolism pathway. Among these pathway constituents, selenium-binding protein 1 (SELENBP1) expression in the NAC-resistant group decreased to less than one-third of that of the NAC-effective group. Immunohistochemistry in another RC cohort consistently validated the relationship between decreased SELENBP1 and poorer NAC sensitivity, in both pre-NAC biopsy and post-NAC surgery specimens. Furthermore, decrease in SELENBP1 was associated with a lower 3-year disease-free survival rate (p = 0.047). CONCLUSIONS We defined one of the differentially expressed proteins in NAC responders and non-responders, concomitant with EMVI and TD. SELENBP1 was suspected to contribute to NAC resistance and poor prognosis in RC.
Collapse
Affiliation(s)
- Shunsuke Kubota
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Takuya Miura
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Taiichi Wakiya
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Tadashi Yoshizawa
- Department of Pathology and Bioscience, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Shintaro Goto
- Department of Pathology and Bioscience, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Hajime Morohashi
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yoshiyuki Sakamoto
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yota Tatara
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Hiroshi Kijima
- Department of Pathology and Bioscience, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kenichi Hakamada
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| |
Collapse
|
10
|
van der Stel SD, van den Berg JG, Snaebjornsson P, Seignette IM, Witteveen M, Grotenhuis BA, Beets GL, Post AL, Ruers TJM. Size and depth of residual tumor after neoadjuvant chemoradiotherapy in rectal cancer - implications for the development of new imaging modalities for response assessment. Front Oncol 2023; 13:1209732. [PMID: 37736547 PMCID: PMC10509550 DOI: 10.3389/fonc.2023.1209732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
With the shift towards organ preserving treatment strategies in rectal cancer it has become increasingly important to accurately discriminate between a complete and good clinical response after neoadjuvant chemoradiotherapy (CRT). Standard of care imaging techniques such as CT and MRI are well equipped for initial staging of rectal tumors, but discrimination between a good clinical and complete response remains difficult due to their limited ability to detect small residual vital tumor fragments. To identify new promising imaging techniques that could fill this gap, it is crucial to know the size and invasion depth of residual vital tumor tissue since this determines the requirements with regard to the resolution and imaging depth of potential new optical imaging techniques. We analyzed 198 pathology slides from 30 rectal cancer patients with a Mandard tumor regression grade 2 or 3 after CRT that underwent surgery. For each patient we determined response pattern, size of the largest vital tumor fragment or bulk and the shortest distance from the vital tumor to the luminal surface. The response pattern was shrinkage in 14 patients and fragmentation in 16 patients. For both groups combined, the largest vital tumor fragment per patient was smaller than 1mm for 38% of patients, below 0.2mm for 12% of patients and for one patient as small as 0.06mm. For 29% of patients the vital tumor remnant was present within the first 0.01mm from the luminal surface and for 87% within 0.5mm. Our results explain why it is difficult to differentiate between a good clinical and complete response in rectal cancer patients using endoscopy and MRI, since in many patients submillimeter tumor fragments remain below the luminal surface. To detect residual vital tumor tissue in all patients included in this study a technique with a spatial resolution of 0.06mm and an imaging depth of 8.9mm would have been required. Optical imaging techniques offer the possibility of detecting majority of these cases due to the potential of both high-resolution imaging and enhanced contrast between tissue types. These techniques could thus serve as a complimentary tool to conventional methods for rectal cancer response assessment.
Collapse
Affiliation(s)
- Stefan D. van der Stel
- Faculty Technische Natuurwetenschappen (TNW), Group Nanobiophysics, Twente University, Enschede, Netherlands
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Petur Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Iris M. Seignette
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Mark Witteveen
- Faculty Technische Natuurwetenschappen (TNW), Group Nanobiophysics, Twente University, Enschede, Netherlands
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Geerard L. Beets
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, Netherlands
- GROW School for Oncology and Developmental Biology, University of Maastricht, Maastricht, Netherlands
| | - Anouk L. Post
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam Universitair Medisch Centrum (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Theo J. M. Ruers
- Faculty Technische Natuurwetenschappen (TNW), Group Nanobiophysics, Twente University, Enschede, Netherlands
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
11
|
Tanaka MD, Geubels BM, Grotenhuis BA, Marijnen CAM, Peters FP, van der Mierden S, Maas M, Couwenberg AM. Validated Pretreatment Prediction Models for Response to Neoadjuvant Therapy in Patients with Rectal Cancer: A Systematic Review and Critical Appraisal. Cancers (Basel) 2023; 15:3945. [PMID: 37568760 PMCID: PMC10417363 DOI: 10.3390/cancers15153945] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Pretreatment response prediction is crucial to select those patients with rectal cancer who will benefit from organ preservation strategies following (intensified) neoadjuvant therapy and to avoid unnecessary toxicity in those who will not. The combination of individual predictors in multivariable prediction models might improve predictive accuracy. The aim of this systematic review was to summarize and critically appraise validated pretreatment prediction models (other than radiomics-based models or image-based deep learning models) for response to neoadjuvant therapy in patients with rectal cancer and provide evidence-based recommendations for future research. MEDLINE via Ovid, Embase.com, and Scopus were searched for eligible studies published up to November 2022. A total of 5006 studies were screened and 16 were included for data extraction and risk of bias assessment using Prediction model Risk Of Bias Assessment Tool (PROBAST). All selected models were unique and grouped into five predictor categories: clinical, combined, genetics, metabolites, and pathology. Studies generally included patients with intermediate or advanced tumor stages who were treated with neoadjuvant chemoradiotherapy. Evaluated outcomes were pathological complete response and pathological tumor response. All studies were considered to have a high risk of bias and none of the models were externally validated in an independent study. Discriminative performances, estimated with the area under the curve (AUC), ranged per predictor category from 0.60 to 0.70 (clinical), 0.78 to 0.81 (combined), 0.66 to 0.91 (genetics), 0.54 to 0.80 (metabolites), and 0.71 to 0.91 (pathology). Model calibration outcomes were reported in five studies. Two collagen feature-based models showed the best predictive performance (AUCs 0.83-0.91 and good calibration). In conclusion, some pretreatment models for response prediction in rectal cancer show encouraging predictive potential but, given the high risk of bias in these studies, their value should be evaluated in future, well-designed studies.
Collapse
Affiliation(s)
- Max D. Tanaka
- Department of Radiation Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Barbara M. Geubels
- Department of Surgery, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Department of Surgery, Catharina Hospital, 5602 ZA Eindhoven, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Brechtje A. Grotenhuis
- Department of Surgery, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Corrie A. M. Marijnen
- Department of Radiation Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Department of Radiation Oncology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Femke P. Peters
- Department of Radiation Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Stevie van der Mierden
- Scientific Information Service, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Monique Maas
- GROW School for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department of Radiology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Alice M. Couwenberg
- Department of Radiation Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
12
|
Lee TH, Jang B, Chang JH, Kim E, Park JH, Chie EK. Genomic landscape of locally advanced rectal adenocarcinoma: Comparison between before and after neoadjuvant chemoradiation and effects of genetic biomarkers on clinical outcomes and tumor response. Cancer Med 2023; 12:15664-15675. [PMID: 37260182 PMCID: PMC10417181 DOI: 10.1002/cam4.6169] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 03/05/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
PURPOSE To explore genomic biomarkers in rectal cancer by performing whole-exome sequencing. MATERIALS AND METHODS Pre-chemoradiation (CRT) biopsy and post-CRT surgical specimens were obtained from 27 patients undergoing neoadjuvant CRT followed by definitive resection. Exomes were sequenced to a mean coverage of 30×. Somatic single-nucleotide variants (SNVs) and insertions/deletions (indels) were identified. Tumor mutational burden was defined as the number of SNVs or indels. Mutational signatures were extracted and fitted to COSMIC reference signatures. Tumor heterogeneity was quantified with a mutant-allele tumor heterogeneity (MATH) score. Genetic biomarkers and frequently occurred copy number alterations (CNAs) were compared between pre- and post-CRT specimens. Their associations with tumor regression grade (TRG) and clinical outcomes were explored. RESULTS Top five mutated genes were APC, TP53, NF1, KRAS, and NOTCH1 for pre-CRT samples and APC, TP53, NF1, CREBBP, and ATM for post-CRT samples. Several gene mutations including RUNX1, EGFR, and TP53 in pre-CRT samples showed significant association with clinical outcomes, but not with TRG. However, no such association was found in post-CRT samples. Discordance of driver mutation status was found between pre- and post-CRT samples. In tumor mutational burden analysis, higher number of SNVs or indels was associated with worse treatment outcomes. Six single-base substitution (SBS) signatures identified were SBS1, SBS30, SBS29, SBS49, SBS3, and SBS44. The MATH score decreased after CRT on paired analysis. Less than half of CNAs frequent in post-CRT samples were present in pre-CRT samples. CONCLUSION Pre- and post-CRT samples showed different genomic landscape. Potential genetic biomarkers of pre-CRT samples found in the current analysis call for external validation.
Collapse
Affiliation(s)
- Tae Hoon Lee
- Department of Radiation OncologySeoul National University HospitalSeoulRepublic of Korea
- Department of Clinical Medical ScienceSeoul National University College of MedicineSeoulRepublic of Korea
| | - Bum‐Sup Jang
- Department of Radiation OncologySeoul National University HospitalSeoulRepublic of Korea
| | - Ji Hyun Chang
- Department of Radiation OncologySeoul National University HospitalSeoulRepublic of Korea
| | - Eunji Kim
- Department of Radiation OncologySeoul Metropolitan Government‐Seoul National University Boramae Medical CenterSeoulRepublic of Korea
| | - Jeong Hwan Park
- Department of PathologySeoul Metropolitan Government‐Seoul National University Boramae Medical CenterSeoulRepublic of Korea
| | - Eui Kyu Chie
- Department of Radiation OncologySeoul National University HospitalSeoulRepublic of Korea
- Department of Clinical Medical ScienceSeoul National University College of MedicineSeoulRepublic of Korea
- Department of Radiation OncologySeoul National University College of MedicineSeoulRepublic of Korea
- Medical Research Center, Institute of Radiation MedicineSeoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
13
|
Chen Y, Sun J, Dong X, Sun D, Qu Y. Significance of ypTNM stage in determining the prognosis and therapy after surgery for locally advanced rectal cancer. World J Surg Oncol 2023; 21:174. [PMID: 37287039 DOI: 10.1186/s12957-023-03059-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/02/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND In the current NCCN guidelines, the prognosis and adjuvant chemotherapy of patients who underwent neoadjuvant chemoradiotherapy (nCRT) are based on pre-radiotherapy clinical TNM (cTNM) stage. However, the value of neoadjuvant pathologic TNM (ypTNM) stage is not clearly described. METHODS This retrospective study investigated the prognosis and adjuvant chemotherapy which based on ypTNM stage compared to cTNM stage. Between 2010 and 2015, a total of 316 rectal cancer patients who underwent nCRT, followed by total mesorectal excision (TME), were included for analysis. RESULTS Our findings revealed that cTNM stage was the only significant independent factor in the pCR group (HR = 6.917, 95% CI: 1.133-42.216, P = 0.038). In the non-pCR group, ypTNM stage was more important than cTNM stage in prognosis (HR = 2.704, 95% CI: 1.811-4.038, P < 0.001). In ypTNM III stage group, there was a statistically significant difference in prognosis between the patients with and without adjuvant chemotherapy (HR = 1.943, 95% CI: 1.015-3.722, P = 0.040), but there was no significant difference in cTNM III stage group (HR = 1.430, 95% CI: 0.728-2.806, P = 0.294). CONCLUSIONS We concluded that ypTNM stage, rather than cTNM stage, might be a more important factor in the prognosis and adjuvant chemotherapy of patients with rectal cancer who underwent nCRT.
Collapse
Affiliation(s)
- Yue Chen
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, China
| | - Jiayu Sun
- Department of Colorectal Surgery, Department of Internal-Medicine, Health Company of Artillery Brigade Service Support Battalion of the 78Th Group Army of the Northern Theater of Chinese People's Liberation Army, No.999 Mail Box, Beishan Road, Siping Economic Development Zone, Tiedong District, Siping City, Jilin Province, 136001, China
| | - Xinxin Dong
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, China
| | - Deyu Sun
- Department of Radiation Oncology Gastrointestinal and Urinary and Musculoskeletal Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, China.
| | - Yanli Qu
- Department of Abdominal and Lymphoma Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, China.
| |
Collapse
|
14
|
Qin S, Lu S, Liu K, Zhou Y, Wang Q, Chen Y, Zhang E, Wang H, Lang N. Radiomics from Mesorectal Blood Vessels and Lymph Nodes: A Novel Prognostic Predictor for Rectal Cancer with Neoadjuvant Therapy. Diagnostics (Basel) 2023; 13:1987. [PMID: 37370882 DOI: 10.3390/diagnostics13121987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/24/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
The objective of our study is to investigate the predictive value of various combinations of radiomic features from intratumoral and different peritumoral regions of interest (ROIs) for achieving a good pathological response (pGR) following neoadjuvant chemoradiotherapy (nCRT) in patients with locally advanced rectal cancer (LARC). This retrospective study was conducted using data from LARC patients who underwent nCRT between 2013 and 2021. Patients were divided into training and validation cohorts at a ratio of 4:1. Intratumoral ROIs (ROIITU) were segmented on T2-weighted imaging, while peritumoral ROIs were segmented using two methods: ROIPTU_2mm, ROIPTU_4mm, and ROIPTU_6mm, obtained by dilating the boundary of ROIITU by 2 mm, 4 mm, and 6 mm, respectively; and ROIMR_F and ROIMR_BVLN, obtained by separating the fat and blood vessels + lymph nodes in the mesorectum. After feature extraction and selection, 12 logistic regression models were established using radiomics features derived from different ROIs or ROI combinations, and five-fold cross-validation was performed. The average area under the receiver operating characteristic curve (AUC) was used to evaluate the performance of the models. The study included 209 patients, consisting of 118 pGR and 91 non-pGR patients. The model that integrated ROIITU and ROIMR_BVLN features demonstrated the highest predictive ability, with an AUC (95% confidence interval) of 0.936 (0.904-0.972) in the training cohort and 0.859 (0.745-0.974) in the validation cohort. This model outperformed models that utilized ROIITU alone (AUC = 0.779), ROIMR_BVLN alone (AUC = 0.758), and other models. The radscore derived from the optimal model can predict the treatment response and prognosis after nCRT. Our findings validated that the integration of intratumoral and peritumoral radiomic features, especially those associated with mesorectal blood vessels and lymph nodes, serves as a potent predictor of pGR to nCRT in patients with LARC. Pending further corroboration in future research, these insights could provide novel imaging markers for refining therapeutic strategies.
Collapse
Affiliation(s)
- Siyuan Qin
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Siyi Lu
- Department of General Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Ke Liu
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Yan Zhou
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Qizheng Wang
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Yongye Chen
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Enlong Zhang
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
- Department of Radiology, Peking University International Hospital, Life Park Road No. 1 Life Science Park of Zhong Guancun, Chang Ping District, Beijing 102206, China
| | - Hao Wang
- Department of Radiation Oncology, Cancer Center, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Ning Lang
- Department of Radiology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| |
Collapse
|
15
|
van der Zijden CJ, Lagarde SM, Hermus M, Kranenburg LW, van Lanschot JJB, Mostert B, Nuyttens JJME, Oudijk L, van der Sluis PC, Spaander MCW, Valkema MJ, Valkema R, Wijnhoven BPL. A prospective cohort study on active surveillance after neoadjuvant chemoradiotherapy for esophageal cancer: protocol of Surgery As Needed for Oesophageal cancer-2. BMC Cancer 2023; 23:327. [PMID: 37038138 PMCID: PMC10084614 DOI: 10.1186/s12885-023-10747-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 03/15/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (nCRT) followed by esophagectomy is a standard treatment for potentially curable esophageal cancer. Active surveillance in patients with a clinically complete response (cCR) 12 weeks after nCRT is regarded as possible alternative to standard surgery. The aim of this study is to monitor the safety, adherence and effectiveness of active surveillance in patients outside a randomized trial. METHODS This nationwide prospective cohort study aims to accrue operable patients with non-metastatic histologically proven adenocarcinoma or squamous cell carcinoma of the esophagus or esophagogastric junction. Patients receive nCRT and response evaluation consists of upper endoscopy with bite-on-bite biopsies, endoscopic ultrasonography plus fine-needle aspiration of suspicious lymph nodes and 18F-fluorodeoxyglucose positron emission tomography/computed tomography scan. When residue or regrowth of tumor in the absence of distant metastases is detected, surgical resection is advised. Patients with cCR after nCRT are suitable to undergo active surveillance. Patients can consult an independent physician or psychologist to support decision-making. Primary endpoint is the number and severity of adverse events in patients with cCR undergoing active surveillance, defined as complications from response evaluations, delayed surgery and the development of distant metastases. Secondary endpoints include timing and quality of diagnostic modalities, overall survival, progression-free survival, fear of cancer recurrence and decisional regret. DISCUSSION Active surveillance after nCRT may be an alternative to standard surgery in patients with esophageal cancer. Similar to organ-sparing approaches applied in other cancer types, the safety and efficacy of active surveillance needs monitoring before data from randomized trials are available. TRIAL REGISTRATION The SANO-2 study has been registered at ClinicalTrials.gov as NCT04886635 (May 14, 2021) - Retrospectively registered.
Collapse
Affiliation(s)
- Charlène J van der Zijden
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015GD, Rotterdam, the Netherlands.
| | - Sjoerd M Lagarde
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Merel Hermus
- Department of Medical Psychology and Psychotherapy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Leonieke W Kranenburg
- Department of Medical Psychology and Psychotherapy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J Jan B van Lanschot
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Joost J M E Nuyttens
- Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lindsey Oudijk
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Pieter C van der Sluis
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maria J Valkema
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Roelf Valkema
- Department of Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bas P L Wijnhoven
- Department of Surgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| |
Collapse
|
16
|
Chou Y, Peng SH, Lin HY, Lan TL, Jiang JK, Liang WY, Hu YW, Wang LW. Radiomic features derived from pretherapeutic MRI predict chemoradiation response in locally advanced rectal cancer. J Chin Med Assoc 2023; 86:399-408. [PMID: 36727777 DOI: 10.1097/jcma.0000000000000887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The standard treatment for locally advanced rectal cancer (LARC) is neoadjuvant concurrent chemoradiotherapy (CRT) followed by surgical excision. Current evidence suggests a favorable prognosis for those with pathological complete response (pCR), and surgery may be spared for them. We trained and validated regression models for CRT response prediction with selected radiomic features extracted from pretreatment magnetic resonance (MR) images to recruit potential candidates for this watch-and-wait strategy. METHODS We retrospectively enrolled patients with LARC who underwent pre-CRT MR imaging between 2010 and 2019. Pathological complete response in surgical specimens after CRT was defined as the ground truth. Quantitative features derived from both unfiltered and filtered images were extracted from manually segmented region of interests on T2-weighted images and selected using variance threshold, univariate statistical tests, and cross-validation least absolute shrinkage and selection operator (Lasso) regression. Finally, a regression model using selected features with high coefficients was optimized and evaluated. Model performance was measured by classification accuracies and area under the receiver operating characteristic (AUROC). RESULTS We extracted 1223 radiomic features from each MRI study of 133 enrolled patients. After tumor excision, 34 (26 %) of 133 patients had pCR in resected specimens. When 25 image-derived features were selected from univariate analysis, classification AUROC was 0.86 and 0.79 with the addition of six clinical features on the hold-out internal validation dataset. When 11 image-derived features were used, the optimized linear regression model had an AUROC value of 0.79 and 0.65 with the addition of six clinical features on the hold-out dataset. Among the radiomic features, texture features including gray level variance, strength, and cluster prominence had the highest coefficient by Lasso regression. CONCLUSION Radiomic features derived from pretreatment MR images demonstrated promising efficacy in predicting pCR after CRT. However, radiomic features combined with clinical features did not result in remarkable improvement in model performance.
Collapse
Affiliation(s)
- Yen Chou
- Department of Medical Imaging, Fu Jen Catholic University Hospital, New Taipei City, Taiwan, ROC
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, ROC
| | - Szu-Hsiang Peng
- Department of Medical Imaging, Far Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
| | - Hsuan-Yin Lin
- Division of Radiology, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| | - Tien-Li Lan
- Division of Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Jeng-Kae Jiang
- Division of Colorectal Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Wen-Yih Liang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Pathology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yu-Wen Hu
- Division of Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Ling-Wei Wang
- Division of Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| |
Collapse
|
17
|
Lv T, Shen L, Xu X, Yao Y, Mu P, Zhang H, Wan J, Wang Y, Guan R, Li X, Fu G, Zhang L, Wang Y, Xia F, Hu C, Clevers H, Zhang Z, Hua G. Patient-derived tumor organoids predict responses to irinotecan-based neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer. Int J Cancer 2023; 152:524-535. [PMID: 36161653 DOI: 10.1002/ijc.34302] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/18/2022] [Accepted: 09/05/2022] [Indexed: 02/01/2023]
Abstract
Adding irinotecan to neoadjuvant chemoradiotherapy (nCRT) in locally advanced rectal cancer (LARC) increases the pathologic complete response (pCR) rate but brings more toxicities. Robust biomarkers to predict response to irinotecan-based nCRT are extremely necessary for selecting the right patients. Our previous study suggests that patient-derived tumor organoids (PDTOs) sensitivity to chemoradiotherapy matches patient responses. In this study, we investigated whether PDTOs sensitivity to irinotecan can predict complete response (CR) and survival. Eligible patients receiving irinotecan-based nCRT between April 5, 2017 and December 11, 2020 were enrolled in the training cohort (n = 91) for response prediction and survival analysis. Patients receiving nCRT between February 21, 2021 and September 17, 2021 were included in the validation cohort (n = 27). Predictive performances of irinotecan organoid size ratio (OSR) for CR or pCR were evaluated. The irinotecan-sensitive groups had higher response rates compared with the insensitive groups (training cohort: 71.8% vs 24.4%, P < .0001; validation cohort, 81.8% vs 18.8%, P = .002). Moreover, the irinotecan-sensitive group had higher rates of 3-year disease-free survival (DFS: 71.6% vs 55.5%, P = .034) and distant metastasis-free survival (DMFS, 77.9% vs 57.2%, P = .015) than the irinotecan-insensitive group. 5-FU and irradiation sensitivities failed to predict 3-year DFS (5-FU: 65.4% vs 61.9%, P = .643; irradiation: 84.8% vs 57.8%; P = .072). Performances of irinotecan OSR to predict CR or pCR were good in the training cohort (CR: AUC = 0.828; 95% CI = 0.723-0.932; pCR: AUC = 0.864; 95% CI = 0.759-0.961). The validation showed robust predictive ability (CR: AUC = 0.796, 95% CI = 0.5974-0.9952; pCR: AUC = 0.917, 95% CI = 0.7921-1.0000). Irinotecan sensitivity in PDTOs was a predictive and prognostic factor in LARC.
Collapse
Affiliation(s)
- Tao Lv
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoya Xu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ye Yao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peiyuan Mu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Juefeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruoyu Guan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaomeng Li
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guoxiang Fu
- Research and Early Development, D1Med Technology (Shanghai) Inc, Shanghai, China
| | - Long Zhang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Cancer institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yaqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Hu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hans Clevers
- Hubrecht Institute, KNAW and University Medical Center Utrecht, Utrecht, The Netherlands.,Pharma, Research and Early Development (pRED) of F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guoqiang Hua
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Cancer institute, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
18
|
Kmeid M, Brar R, Sullivan L, Arslan ME, Shrestha N, Lee EC, Chen A, Jennings TA, Lee H. Diagnostic yield and repeat biopsies in rectal and nonrectal colorectal adenocarcinoma: Are we hedging on rectal biopsies? Acad Pathol 2023; 10:100063. [PMID: 36970329 PMCID: PMC10031322 DOI: 10.1016/j.acpath.2022.100063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/09/2022] [Accepted: 11/12/2022] [Indexed: 02/05/2023] Open
Abstract
Patients with rectal cancer undergo more repeat biopsies compared to those with nonrectal colon cancer prior to management. We investigated the factors driving the higher frequency of repeat biopsies in patients with rectal cancer. We compared clinicopathologic features of diagnostic and nondiagnostic (in regard to invasion) rectal (n = 64) and colonic (n = 57) biopsies from colorectal cancer patients and characterized corresponding resections. Despite similar diagnostic yield, repeat biopsy was more common in rectal carcinoma, especially in patients receiving neoadjuvant therapy (p < 0.05). The presence of desmoplasia (odds ratio 12.9, p < 0.05) was a strong predictor of making a diagnosis of invasion in both rectal and nonrectal colon cancer biopsies. Diagnostic biopsies had more desmoplasia, intramucosal carcinoma component and marked inflammation, and less low-grade dysplasia component (p < 0.05). Diagnostic yield of biopsy was higher for tumors with high-grade tumor budding, mucosal involvement by high-grade dysplasia/intramucosal carcinoma without low-grade dysplasia and diffuse surface desmoplasia irrespective of tumor location. Sample size, amount of benign tissue, appearance, and T stage did not affect diagnostic yield. Repeat biopsy of rectal cancer is primarily driven by management implications. Diagnostic yield in colorectal cancer biopsies is multifactorial and is not due to differing pathologists' diagnostic approach per tumor site. For rectal tumors, a multidisciplinary strategic approach is warranted to avoid repeat biopsy when unnecessary.
Collapse
Affiliation(s)
- Michel Kmeid
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | - Rupinder Brar
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | - Luz Sullivan
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | | | | | - Edward C. Lee
- Department of Surgery, Albany Medical Center, Albany, NY, USA
| | - Anne Chen
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | | | - Hwajeong Lee
- Department of Pathology, Albany Medical Center, Albany, NY, USA
- Corresponding author. Department of Pathology, Albany Medical Center, 47 New Scotland Ave., MC81, Albany, NY 12208, USA.
| |
Collapse
|
19
|
Predicting Neoadjuvant Treatment Response in Rectal Cancer Using Machine Learning: Evaluation of MRI-Based Radiomic and Clinical Models. J Gastrointest Surg 2023; 27:122-130. [PMID: 36271199 DOI: 10.1007/s11605-022-05477-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/25/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Radiomics is an approach to medical imaging that quantifies the features normally translated into visual display. While both radiomic and clinical markers have shown promise in predicting response to neoadjuvant chemoradiation therapy (nCRT) for rectal cancer, the interrelationship is not yet clear. METHODS A retrospective, single-institution study of patients treated with nCRT for locally advanced rectal cancer was performed. Clinical and radiomic features were extracted from electronic medical record and pre-treatment magnetic resonance imaging, respectively. Machine learning models were created and assessed for complete response and positive treatment effect using the area under the receiver operating curves. RESULTS Of 131 rectal cancer patients evaluated, 68 (51.9%) were identified to have a positive treatment effect and 35 (26.7%) had a complete response. On univariate analysis, clinical T-stage (OR 0.46, p = 0.02), lymphovascular/perineural invasion (OR 0.11, p = 0.03), and statin use (OR 2.45, p = 0.049) were associated with a complete response. Clinical T-stage (OR 0.37, p = 0.01), lymphovascular/perineural invasion (OR 0.16, p = 0.001), and abnormal carcinoembryonic antigen level (OR 0.28, p = 0.002) were significantly associated with a positive treatment effect. The clinical model was the strongest individual predictor of both positive treatment effect (AUC = 0.64) and complete response (AUC = 0.69). The predictive ability of a positive treatment effect increased by adding tumor and mesorectal radiomic features to the clinical model (AUC = 0.73). CONCLUSIONS The use of a combined model with both clinical and radiomic features resulted in the strongest predictive capability. With the eventual goal of tailoring treatment to the individual, both clinical and radiologic markers offer insight into identifying patients likely to respond favorably to nCRT.
Collapse
|
20
|
Montori A, Germani A, Ferri M, Milano A, Ranalli TV, Piane M, Pilozzi E. Somatic NGS Analysis of DNA Damage Response (DDR) Genes ATM, MRE11A, RAD50, NBN, and ATR in Locally Advanced Rectal Cancer Treated with Neoadjuvant Chemo-Radiotherapy. Biomedicines 2022; 10:biomedicines10123247. [PMID: 36552003 PMCID: PMC9775018 DOI: 10.3390/biomedicines10123247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/25/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neoadjuvant chemo-radiotherapy (nCRT) represents the standard of care for locally advanced rectal cancer (LARC); however, there exists no biomarker that can predict the cancer's response to treatment as less than 20% of patients experience pathological complete response (pCR). Ionizing radiations induce double strand breaks (DSBs) and trigger a DNA damage response (DDR) involving ATM, ATR, and the MRN complex (MRE11, Rad50, and NBS1). In this study, we performed an extensive mutational analysis of the genes involved in the DDR pathway in LARC patients who have undergone nCRT. METHODS 13 LARC patients with pCR and 11 LARC patients with partial response (pPR) were investigated using a NGS dedicated panel, designed for formalin-fixed paraffin-embedded (FFPE) samples, containing ATR, ATM, and MRE11-RAD50-NBN genes. The identified variants were classified according to guidelines' recommendations. RESULTS Eight non-benign variants, six of which were observed in 3 (23%) out of 13 pCR patients, were identified. In particular, a pCR patient carried out a pathogenetic frameshift mutation in exon 21 of the RAD50 gene. The two remaining non-benign missense variants were found in 2 (18%) out of 11 patients in the pPR group. CONCLUSIONS Our data show that the genes involved in the Homologous Recombination (HR) pathway are rarely mutated in LARC; however, given the identification of a missense mutation in RAD 50 in one case of pCR, it could be worth exploring its potential role as a biomarker in larger series.
Collapse
Affiliation(s)
- Andrea Montori
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Unit of Pathologic Morphological and Molecular Anatomy, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Mario Ferri
- Department of Medical-Surgical Sciences and Translational Medicine, “Sapienza” University of Rome, Piazzale Aldo Moro 5, 00189 Rome, Italy
- Unit of Gastrointestinal Surgery, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Annalisa Milano
- Unit of Oncology, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
| | | | - Maria Piane
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Unit of Medical Genetics and Advanced Cellular Diagnostic, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Unit of Pathologic Morphological and Molecular Anatomy, Sant’Andrea University Hospital, Via di Grottarossa 1035, 00189 Rome, Italy
- Correspondence:
| |
Collapse
|
21
|
Tojo M, Horie H, Koinuma K, Miyato H, Tsukui H, Kaneko Y, Futoh Y, Kimura Y, Takahashi K, Saito A, Ohzawa H, Yamaguchi H, Lefor AK, Sata N, Kitayama J. Programmed cell death ligand 1 expression on monocytes is inversely correlated with tumour response to preoperative chemoradiotherapy for locally advanced rectal cancer. Colorectal Dis 2022; 24:1140-1149. [PMID: 35502766 PMCID: PMC9790410 DOI: 10.1111/codi.16167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/17/2022] [Accepted: 04/23/2022] [Indexed: 12/30/2022]
Abstract
AIM The clinical efficacy of chemoradiotherapy (CRT) is largely dependent on host immune status. The aim of this study was to identify possible markers expressed on circulating mononuclear cells to predict tumour response in patients with locally advanced rectal cancer (LARC). METHODS Peripheral blood samples were obtained from 47 patients diagnosed with LARC before and after CRT. The numbers of lymphocytes and monocyte subsets were analysed using flow cytometry. Based on clinical and pathological findings, patients were classified as high or low responders. RESULTS Lymphocyte counts were markedly decreased after CRT. Total numbers of lymphocytes (p = 0.030) and CD4(+) T cells (p = 0.041) in post-CRT samples were significantly lower in low responders than in high responders. In contrast, monocyte counts were not reduced and the number of CD14dim (+) CD16(+) nonclassical (patrolling) monocytes were somewhat increased after CRT (p = 0.050). Moreover, the ratios of programmed cell death ligand 1 (PD-L1) (+) cells on patrolling monocytes before and after CRT were significantly higher in low responders than in high responders (p = 0.0046, p = 0.0006). The same trend was observed for classical and intermediate monocytes. The expression of PD-L1 on patrolling monocytes before CRT correlated inversely with the number of T cells and natural killer (NK) cells after CRT. PD-L1(+) ratio in patrolling monocytes was an independent predictor for response to CRT. CONCLUSION Programmed cell death ligand 1 (PD-L1) expression on patrolling monocytes suppresses cell-mediated immunity in patients receiving CRT which could be related to tumour response, and may be a useful biomarker for decision-making in the management of patients with LARC.
Collapse
Affiliation(s)
- Mineyuki Tojo
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Hisanaga Horie
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Koji Koinuma
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Hideyo Miyato
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan,Jichi Medical University HospitalDivision of Translational Research, Center for Clinical ResearchShimotsukeTochigiJapan
| | - Hidenori Tsukui
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Yuki Kaneko
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Yurie Futoh
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Yuki Kimura
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Kazuya Takahashi
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Akira Saito
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Hideyuki Ohzawa
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan,Department of Clinical OncologyJichi Medical UniversityShimotsukeTochigiJapan
| | - Hironori Yamaguchi
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan,Department of Clinical OncologyJichi Medical UniversityShimotsukeTochigiJapan
| | | | - Naohiro Sata
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan
| | - Joji Kitayama
- Department of SurgeryJichi Medical UniversityShimotsukeTochigiJapan,Jichi Medical University HospitalDivision of Translational Research, Center for Clinical ResearchShimotsukeTochigiJapan
| |
Collapse
|
22
|
Yang F, Hill J, Abraham A, Ghosh S, Steed T, Kurtz C, Joseph K, Yun J, Warkentin B, Thai J, Nijjar T, Severin D, Tankel K, Fairchild A, Usmani N. Tumor Volume Predicts for Pathologic Complete Response in Rectal Cancer Patients Treated With Neoadjuvant Chemoradiation. Am J Clin Oncol 2022; 45:405-409. [PMID: 36106894 DOI: 10.1097/coc.0000000000000942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Nonoperative management (NOM) of locally advanced rectal cancer is an emerging approach allowing patients to preserve their anal sphincter. Identifying clinical factors associated with pathologic complete response (pCR) is essential for physicians and patients considering NOM. MATERIALS AND METHODS In total, 412 locally advanced rectal cancer patients were included in this retrospective analysis. Tumor volumes were derived from pretreatment MRI. Clinical parameters such as tumor volume, stage, and location were analyzed by univariate and multivariate analysis, against pCR. A receiver operator characteristic curve was generated to identify a tumor volume cut-off with the highest clinically relevant Youden index for predicting pCR. RESULTS Seventy-five of 412 patients (18%) achieved pCR. A tumor volume threshold of 37.3 cm 3 was identified as predictive for pCR. On regression analysis, a tumor volume >37.3 cm 3 was associated with a greater than 78% probability of not achieving pCR. On multivariate analysis, a GTV <37.3 cm 3 [odds ratio (OR)=3.7, P <0.0001] was significantly associated with an increased pCR rate, whereas tumor length > 4.85 cm was associated with pCR on univariate (OR=3.03, P <0.01) but not on multivariate analysis (OR=1.45, P =0.261). Other clinical parameters did not impact pCR rates. CONCLUSIONS A tumor volume threshold of 37.3 cm 3 was identified as predictive for pCR in locally advanced rectal cancer patients receiving neoadjuvant chemoradiation. Tumors above this volume threshold corresponded to a greater than 78% probability of not achieving pCR. This information will be helpful at diagnosis for clinicians who are considering potential candidates for NOM.
Collapse
Affiliation(s)
- Fan Yang
- Division of Radiation Oncology, Cross Cancer Institute
| | - Jordan Hill
- Division of Radiation Oncology, Cross Cancer Institute
| | - Aswin Abraham
- Division of Radiation Oncology, Cross Cancer Institute
| | - Sunita Ghosh
- Division of Radiation Oncology, Cross Cancer Institute
| | - Tanner Steed
- Division of Radiation Oncology, Cross Cancer Institute
| | - Clay Kurtz
- Undergraduate Medical Program, Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Kurian Joseph
- Division of Radiation Oncology, Cross Cancer Institute
| | - Jihyun Yun
- Division of Radiation Oncology, Cross Cancer Institute
| | | | - JoAnn Thai
- Undergraduate Medical Program, Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Tirath Nijjar
- Division of Radiation Oncology, Cross Cancer Institute
| | - Diane Severin
- Division of Radiation Oncology, Cross Cancer Institute
| | - Keith Tankel
- Division of Radiation Oncology, Cross Cancer Institute
| | | | - Nawaid Usmani
- Division of Radiation Oncology, Cross Cancer Institute
| |
Collapse
|
23
|
Latif A, Shirkhoda M, Rouhollahi MR, Nemati S, Yahyazadeh SH, Zendehdel K, Soroush AR, Yaghoobi Notash A. Predicting Factors of Complete Pathological Response in Locally Advanced Rectal Cancer. Middle East J Dig Dis 2022; 14:443-451. [PMID: 37547496 PMCID: PMC10404107 DOI: 10.34172/mejdd.2022.306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/20/2022] [Indexed: 08/08/2023] Open
Abstract
Background: Current treatment of choice for locally advanced rectal cancer is neoadjuvant chemoradiotherapy (neo-CRT) followed by surgical resection and adjuvant chemotherapy. Some patients may experience complete pathological response (cPR) after the neoadjuvant treatment. However, the predicting factors are still debated. Methods: In this registry-based retrospective cohort study, 258 patients with locally advanced rectal cancer were included. Patients were categorized into two groups with or without cPR. Logistic regression analysis was recruited to investigate the odds ratio for all independent variables, and those with significant results were included in multivariate regression analysis. Results: Achievement of cPR was 21.3%. The odds ratio of cPR was significantly lower when the tumor distance from the anal verge was>10 centimeters (OR=0.24, P=0.040). Also, the odds of cPR with N1 involvement in comparison with N0 involvement decreased for 0.41 (P=0.043). It was also true for patients with N2 involvement in comparison with N0 involvement (OR=0.31, P=0.031). Higher odds ratio of cPR was observed in patients who underwent surgery in>12 weeks after neo-CRT (OR=2.9, P=0.022). Furthermore, the odds of cPR decreased for 0.9 with increasing in carcinoembryonic antigen (CEA) level (P=0.044). Conclusion: Patients with rectal cancer in clinical stage II or lower, without the involvement of the lymphatic system at diagnosis, and with tumors located in the lower parts of the rectum, with lower levels of CEA, and longer duration between neo-CRT and surgery were more likely to achieve cPR after neo-CRT. With the current knowledge, the "wait and watch policy" is still debated and needs to be defined more precisely by upcoming studies.
Collapse
Affiliation(s)
- AmirHossein Latif
- Department of General Surgery, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shirkhoda
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saeed Nemati
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kazem Zendehdel
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Soroush
- Department of General Surgery, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Aidin Yaghoobi Notash
- Department of General Surgery, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Akbulut S, Hargura AS, Garzali IU, Aloun A, Colak C. Clinical presentation, management, screening and surveillance for colorectal cancer during the COVID-19 pandemic. World J Clin Cases 2022; 10:9228-9240. [PMID: 36159422 PMCID: PMC9477669 DOI: 10.12998/wjcc.v10.i26.9228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/29/2022] [Accepted: 08/05/2022] [Indexed: 02/05/2023] Open
Abstract
Management of colorectal cancer (CRC) was severely affected by the changes implemented during the pandemic, and this resulted in delayed elective presentation, increased emergency presentation, reduced screening and delayed definitive therapy. This review was conducted to analyze the impact of the coronavirus disease 2019 (COVID-19) pandemic on management of CRC and to identify the changes made in order to adapt to the pandemic. We performed a literature search in PubMed, Medline, Index Medicus, EMBASE, SCOPUS, Reference Citation Analysis (https://www.referencecitationanalysis.com/) and Google Scholar using the following keywords in various combinations: Colorectal cancer, elective surgery, emergency surgery, stage upgrading, screening, surveillance and the COVID-19 pandemic. Only studies published in English were included. To curtail the spread of COVID-19 infection, there were modifications made in the management of CRC. Screening was limited to high risk individuals, and the screening tests of choice during the pandemic were fecal occult blood test, fecal immunochemical test and stool DNA testing. The use of capsule colonoscopy and open access colonoscopy was also encouraged. Blood-based tests like serum methylated septin 9 were also encouraged for screening of CRC during the pandemic. The presentation of CRC was also affected by the pandemic with more patients presenting with emergencies like obstruction and perforation. Stage migration was also observed during the pandemic with more patients presenting with more advanced tumors. The operative therapy of CRC was altered by the pandemic as more emergencies surgeries were done, which may require exteriorization by stoma. This was to reduce the morbidity associated with anastomosis and encourage early discharge from the hospital. There was also an initial reduction in laparoscopic surgical procedures due to the fear of aerosols and COVID-19 infection. As we gradually come out of the pandemic, we should remember the lessons learned and continue to apply them even after the pandemic passes.
Collapse
Affiliation(s)
- Sami Akbulut
- Department of Surgery, Inonu University Faculty of Medicine, Malatya 44280, Turkey
- Biostatistics and Medical Informatics, Inonu University Faculty of Medicine, Malatya 44280, Turkey
| | - Abdirahman Sakulen Hargura
- Department of Surgery, Inonu University Faculty of Medicine, Malatya 44280, Turkey
- Department of Surgery, Kenyatta University Teaching, Referral and Research Hospital, Nairobi 00100, Kenya
| | - Ibrahim Umar Garzali
- Department of Surgery, Inonu University Faculty of Medicine, Malatya 44280, Turkey
- Department of Surgery, Aminu Kano Teaching Hospital, Kano 700101, Nigeria
| | - Ali Aloun
- Department of Surgery, King Hussein Medical Center, Amman 11855, Jordan
| | - Cemil Colak
- Biostatistics and Medical Informatics, Inonu University Faculty of Medicine, Malatya 44280, Turkey
| |
Collapse
|
25
|
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
|
26
|
Lee SY, Kim DW, Lee J, Park HM, Kim CH, Lee KH, Oh HK, Kang SB, Kim HR. Association between microsatellite instability and tumor response to neoadjuvant chemoradiotherapy for rectal cancer. Ann Surg Treat Res 2022; 103:176-182. [PMID: 36128037 PMCID: PMC9478427 DOI: 10.4174/astr.2022.103.3.176] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose The relationship between microsatellite instability (MSI) and tumor response after neoadjuvant chemoradiotherapy (nCRT) in rectal cancer remains unclear. The present study aimed to evaluate the association between MSI and tumor response to nCRT in rectal cancer treatment. Methods Patients with rectal cancer from 2 tertiary hospitals who underwent nCRT, followed by radical surgery, were included. The microsatellite status was determined using a PCR-based Bethesda panel. Tumors with a Dworak’s tumor regression grade of 3 or 4 were considered to have a good response. Predictive factors for a good response to nCRT were analyzed. Results Of the 1,401 patients included, 910 (65.0%) had MSI results and 1.5% (14 of 910) showed MSI-H. Among all the patients, 519 (37.0%) showed a good response to nCRT. A univariate analysis showed that MSI-H tended to be negatively associated with a good response to nCRT, but no statistical significance was observed (7.1% vs. 24.1%, P = 0.208). Multivariate analysis showed that well-differentiated tumors were the only predictive factor for good response to nCRT (odds ratio [OR], 2.241; 95% confidence interval [CI], 1.492–3.364; P < 0.001). MSI status tended to be associated with the response to nCRT (OR, 0.215; 95% CI, 0.027–1.681; P = 0.143). Conclusion MSI-H was not associated with response to nCRT in patients with rectal cancer.
Collapse
Affiliation(s)
- Soo Young Lee
- Department of Surgery, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Duck-Woo Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jaram Lee
- Department of Surgery, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Hyeong-Min Park
- Department of Surgery, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Chang Hyun Kim
- Department of Surgery, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Heung-Kwon Oh
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Bum Kang
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyeong Rok Kim
- Department of Surgery, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
27
|
Ghadimi M, Rödel C, Hofheinz R, Flebbe H, Grade M. Multimodal Treatment of Rectal Cancer. DEUTSCHES ARZTEBLATT INTERNATIONAL 2022; 119:570-580. [PMID: 35791271 PMCID: PMC9743213 DOI: 10.3238/arztebl.m2022.0254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/03/2022] [Accepted: 06/14/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Colorectal cancer is one of the three most common types of cancer in Germany. Approximately 30% of these cancers are located in the rectum, corresponding to about 18 000 new cases per year. METHODS This review is based on publications retrieved by a selective search in the PubMed database, including current guidelines and recommendations. RESULTS Specialized imaging, particularly magnetic resonance imaging, is essential for treatment planning. In very early stages of this disease, tumors without risk factors can be excised locally. Otherwise, radical surgical resection with lymphadenectomy remains the standard treatment, and can be performed either minimally invasive or open. At present, neoadjuvant treatment plans are evolving in the direction of total neoadjuvant therapy. In addition, recent studies investigate whether the improved efficacy of neoadjuvant therapy might now enable patients with a complete clinical remission to be spared from surgical resection (organ-preserving watch-and-wait strategy). CONCLUSION The treatment of rectal cancer is a prime example of an interdisciplinary, multimodal approach. In the past, the focus was mainly on improving oncologic outcomes; at present, increasing attention is being devoted to the patients' quality of life as well and the functional aspects of the various modes of treatment.
Collapse
Affiliation(s)
- Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen
| | - Claus Rödel
- Department of Radiotherapy and Oncology, Goethe University Frankfurt am Main
| | - Ralf Hofheinz
- Department of Medical Hematology and Oncology, University Hospital Mannheim, University of Heidelberg
| | - Hannah Flebbe
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen
| | - Marian Grade
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen,*Universitätsmedizin Göttingen Klinik für Allgemein-, Viszeral- und Kinderchirurgie Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| |
Collapse
|
28
|
Wanigasooriya K, Barros-Silva JD, Tee L, El-asrag ME, Stodolna A, Pickles OJ, Stockton J, Bryer C, Hoare R, Whalley CM, Tyler R, Sillo T, Yau C, Ismail T, Beggs AD. Patient Derived Organoids Confirm That PI3K/AKT Signalling Is an Escape Pathway for Radioresistance and a Target for Therapy in Rectal Cancer. Front Oncol 2022; 12:920444. [PMID: 35860583 PMCID: PMC9289101 DOI: 10.3389/fonc.2022.920444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
Objectives Partial or total resistance to preoperative chemoradiotherapy occurs in more than half of locally advanced rectal cancer patients. Several novel or repurposed drugs have been trialled to improve cancer cell sensitivity to radiotherapy, with limited success. We aimed to understand the mechanisms of resistance to chemoradiotherapy in rectal cancer using patient derived organoid models. Design To understand the mechanisms underlying this resistance, we compared the pre-treatment transcriptomes of patient-derived organoids (PDO) with measured radiotherapy sensitivity to identify biological pathways involved in radiation resistance coupled with single cell sequencing, genome wide CRISPR-Cas9 and targeted drug screens. Results RNA sequencing enrichment analysis revealed upregulation of PI3K/AKT/mTOR and epithelial mesenchymal transition pathway genes in radioresistant PDOs. Single-cell sequencing of pre & post-irradiation PDOs showed mTORC1 and PI3K/AKT upregulation, which was confirmed by a genome-wide CRSIPR-Cas9 knockout screen using irradiated colorectal cancer (CRC) cell lines. We then tested the efficiency of dual PI3K/mTOR inhibitors in improving cancer cell sensitivity to radiotherapy. After irradiation, significant AKT phosphorylation was detected (p=0.027) which was abrogated with dual PI3K/mTOR inhibitors and lead to significant radiosensitisation of the HCT116 cell line and radiation resistant PDO lines. Conclusions The PI3K/AKT/mTOR pathway upregulation contributes to radioresistance and its targeted pharmacological inhibition leads to significant radiosensitisation in CRC organoids, making it a potential target for clinical trials.
Collapse
Affiliation(s)
- Kasun Wanigasooriya
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- Department of Surgery, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
| | - Joao D. Barros-Silva
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
| | - Louise Tee
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
| | - Mohammed E. El-asrag
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
| | - Agata Stodolna
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
| | - Oliver J. Pickles
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- Department of Surgery, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
| | - Joanne Stockton
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
| | - Claire Bryer
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
| | - Rachel Hoare
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
| | - Celina M. Whalley
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
| | - Robert Tyler
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- Department of Surgery, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
| | - Toritseju Sillo
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- Department of Surgery, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
| | - Christopher Yau
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
| | - Tariq Ismail
- Department of Surgery, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
| | - Andrew D. Beggs
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Birmingham, United Kingdom
- Department of Surgery, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- *Correspondence: Andrew D. Beggs,
| |
Collapse
|
29
|
Mbanu P, Saunders MP, Mistry H, Mercer J, Malcomson L, Yousif S, Price G, Kochhar R, Renehan AG, van Herk M, Osorio EV. Clinical and radiomics prediction of complete response in rectal cancer pre-chemoradiotherapy. Phys Imaging Radiat Oncol 2022; 23:48-53. [PMID: 35800297 PMCID: PMC9253904 DOI: 10.1016/j.phro.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 11/11/2022] Open
Abstract
Background and purpose Patients with rectal cancer could avoid major surgery if they achieve clinical complete response (cCR) post neoadjuvant treatment. Therefore, prediction of treatment outcomes before treatment has become necessary to select the best neo-adjuvant treatment option. This study investigates clinical and radiomics variables' ability to predict cCR in patients pre chemoradiotherapy. Materials and methods Using the OnCoRe database, we recruited a matched cohort of 304 patients (152 with cCR; 152 without cCR) deriving training (N = 200) and validation (N = 104) sets. We collected pre-treatment MR (magnetic resonance) images, demographics and blood parameters (haemoglobin, neutrophil, lymphocyte, alkaline phosphate and albumin). We segmented the gross tumour volume on T2 Weighted MR Images and extracted 1430 stable radiomics features per patient. We used principal component analysis (PCA) and receiver operating characteristic area under the curve (ROC AUC) to reduce dimensionality and evaluate the models produced. Results Using Logistic regression analysis, PCA-derived combined model (radiomics plus clinical variables) gave a ROC AUC of 0.76 (95% CI: 0.69-0.83) in the training set and 0.68 (95% CI 0.57-0.79) in the validation set. The clinical only model achieved an AUC of 0.73 (95% CI 0.66-0.80) and 0.62 (95% CI 0.51-0.74) in the training and validation set, respectively. The radiomics model had an AUC of 0.68 (95% CI 0.61-0.75) and 0.66 (95% CI 0.56-0.77) in the training and validation sets. Conclusion The predictive characteristics of both clinical and radiomics variables for clinical complete response remain modest but radiomics predictability is improved with addition of clinical variables.
Collapse
Affiliation(s)
- Peter Mbanu
- Department of Clinical Oncology, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Mark P. Saunders
- Department of Clinical Oncology, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Hitesh Mistry
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
- Division of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - Joe Mercer
- Department of Radiological Oncology, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Lee Malcomson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
- Colorectal and Peritoneal Oncology Centre, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Saif Yousif
- Department of Clinical Oncology, Lancashire Teaching Hospital, Preston, United Kingdom
| | - Gareth Price
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Rohit Kochhar
- Department of Radiological Oncology, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Andrew G. Renehan
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
- Colorectal and Peritoneal Oncology Centre, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Marcel van Herk
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Eliana Vasquez Osorio
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
30
|
Zwart WH, Hotca A, Hospers GAP, Goodman KA, Garcia-Aguilar J. The Multimodal Management of Locally Advanced Rectal Cancer: Making Sense of the New Data. Am Soc Clin Oncol Educ Book 2022; 42:1-14. [PMID: 35561302 DOI: 10.1200/edbk_351411] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the past 40 years, the treatment of locally advanced rectal cancer has evolved with the addition of radiotherapy or chemoradiotherapy and providing (neo)adjuvant systemic chemotherapy to major surgery. However, recent trends have focused on improving our ability to risk-stratify patients and tailoring treatment to achieve the best oncologic outcome while limiting the impact on long-term quality of life. Therefore, there has been increasing interest in pursuing a watch-and-wait approach to achieve organ preservation. Several retro- and prospective studies suggest safety of the watch-and-wait approach, though it is still considered controversial due to limited clinical evidence, concerns about tumor regrowth, and subsequent distant progression. To further reduce treatment, MRI risk stratification, together with patient characteristics and patient preferences, can guide personalized treatment and reserve radiation and chemotherapy for a select patient population. Ultimately, improved options for reassessment during neoadjuvant treatment may allow for more adaptive therapy options based on treatment response. This article provides an overview of some major developments in the multimodal treatment of locally advanced rectal cancer. It reviews some relevant, controversial issues of the watch-and-wait approach and opportunities to personally tailor and reduce treatment. It also reviews the overall neoadjuvant treatment, including total neoadjuvant therapy trials, and how to best optimize for a potential complete response. Finally, it provides an algorithm as an example of how such a personalized, tailored, adaptive, and reduced treatment could look like in the future.
Collapse
Affiliation(s)
- Wouter H Zwart
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Geke A P Hospers
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | | |
Collapse
|
31
|
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
|
32
|
Fleischmann M, Diefenhardt M, Nicolas AM, Rödel F, Ghadimi M, Hofheinz RD, Greten FR, Rödel C, Fokas E. ACO/ARO/AIO-21 - Capecitabine-based chemoradiotherapy in combination with the IL-1 receptor antagonist anakinra for rectal cancer Patients: A phase I trial of the German rectal cancer study group. Clin Transl Radiat Oncol 2022; 34:99-106. [PMID: 35449546 PMCID: PMC9018120 DOI: 10.1016/j.ctro.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/04/2022] [Indexed: 01/04/2023] Open
Abstract
Response to chemoradiotherapy in rectal cancer is highly heterogeneous, ranging from complete response to tumor progression. Interleukin-1 signaling polarizes cancer-associated fibroblasts (CAF) towards an inflammatory phenotype and predisposes iCAFs to irradiation-induced senescence. Targeting interleukin-1 could potentially reconstitute the tumor microenvironment and improve therapy response. The ACO/ARO-AIO-21 phase I trial is testing the interleukin-1 receptor antagonist anakinra in combination with fluoropyrimidine-based chemoradiotherapy in rectal cancer.
Purpose Methods/Design Discussion
Collapse
|
33
|
Takenaka IKTM, Bartelli TF, Defelicibus A, Sendoya JM, Golubicki M, Robbio J, Serpa MS, Branco GP, Santos LBC, Claro LCL, Dos Santos GO, Kupper BEC, da Silva IT, Llera AS, de Mello CAL, Riechelmann RP, Dias-Neto E, Iseas S, Aguiar S, Nunes DN. Exome and Tissue-Associated Microbiota as Predictive Markers of Response to Neoadjuvant Treatment in Locally Advanced Rectal Cancer. Front Oncol 2022; 12:809441. [PMID: 35392220 PMCID: PMC8982181 DOI: 10.3389/fonc.2022.809441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical and pathological responses to multimodal neoadjuvant therapy in locally advanced rectal cancers (LARCs) remain unpredictable, and robust biomarkers are still lacking. Recent studies have shown that tumors present somatic molecular alterations related to better treatment response, and it is also clear that tumor-associated bacteria are modulators of chemotherapy and immunotherapy efficacy, therefore having implications for long-term survivorship and a good potential as the biomarkers of outcome. Here, we performed whole exome sequencing and 16S ribosomal RNA (rRNA) amplicon sequencing from 44 pre-treatment LARC biopsies from Argentinian and Brazilian patients, treated with neoadjuvant chemoradiotherapy or total neoadjuvant treatment, searching for predictive biomarkers of response (responders, n = 17; non-responders, n = 27). In general, the somatic landscape of LARC was not capable to predict a response; however, a significant enrichment in mutational signature SBS5 was observed in non-responders (p = 0.0021), as well as the co-occurrence of APC and FAT4 mutations (p < 0.05). Microbiota studies revealed a similar alpha and beta diversity of bacteria between response groups. Yet, the linear discriminant analysis (LDA) of effect size indicated an enrichment of Hungatella, Flavonifractor, and Methanosphaera (LDA score ≥3) in the pre-treatment biopsies of responders, while non-responders had a higher abundance of Enhydrobacter, Paraprevotella (LDA score ≥3) and Finegoldia (LDA score ≥4). Altogether, the evaluation of these biomarkers in pre-treatment biopsies could eventually predict a neoadjuvant treatment response, while in post-treatment samples, it could help in guiding non-operative treatment strategies.
Collapse
Affiliation(s)
| | - Thais F Bartelli
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Alexandre Defelicibus
- Laboratory of Bioinformatics and Computational Biology, International Center for Research, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Juan M Sendoya
- Laboratorio de Terapia Molecular y Celular - Genomics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mariano Golubicki
- Oncology Unit, Hospital de Gastroenterología Carlos Bonorino Udaondo, Buenos Aires, Argentina.,Clinical Oncology, Intergrupo Argentino para el Tratamiento de los Tumores Gastrointestinales (IATTGI), Buenos Aires, Argentina
| | - Juan Robbio
- Clinical Oncology, Intergrupo Argentino para el Tratamiento de los Tumores Gastrointestinales (IATTGI), Buenos Aires, Argentina
| | - Marianna S Serpa
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Gabriela P Branco
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Luana B C Santos
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Laura C L Claro
- Department of Pathology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | | | - Bruna E C Kupper
- Colorectal Cancer Department, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Israel T da Silva
- Laboratory of Bioinformatics and Computational Biology, International Center for Research, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Andrea S Llera
- Laboratorio de Terapia Molecular y Celular - Genomics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Celso A L de Mello
- Department of Clinical Oncology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | | | - Emmanuel Dias-Neto
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil.,Laboratory of Neurosciences (LIM-27) Alzira Denise Hertzog Silva, Institute of Psychiatry, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Soledad Iseas
- Oncology Unit, Hospital de Gastroenterología Carlos Bonorino Udaondo, Buenos Aires, Argentina
| | - Samuel Aguiar
- Colorectal Cancer Department, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Diana Noronha Nunes
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil.,National Institute of Science and Technology in Oncogenomics and Therapeutic Innovation (INCITO), São Paulo, Brazil
| |
Collapse
|
34
|
Chiloiro G, Boldrini L, Preziosi F, Cusumano D, Yadav P, Romano A, Placidi L, Lenkowicz J, Dinapoli N, Bassetti MF, Gambacorta MA, Valentini V. A Predictive Model of 2yDFS During MR-Guided RT Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer Patients. Front Oncol 2022; 12:831712. [PMID: 35280799 PMCID: PMC8907443 DOI: 10.3389/fonc.2022.831712] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/31/2022] [Indexed: 01/04/2023] Open
Abstract
Purpose Distant metastasis is the main cause of treatment failure in locally advanced rectal cancer (LARC) patients, despite the recent improvement in treatment strategies. This study aims to evaluate the “delta radiomics” approach in patients undergoing neoadjuvant chemoradiotherapy (nCRT) treated with 0.35-T magnetic resonance-guided radiotherapy (MRgRT), developing a logistic regression model able to predict 2-year disease-free-survival (2yDFS). Methods Patients affected by LARC were enrolled in this multi-institutional study. A predictive model of 2yDFS was developed taking into account both clinical and radiomics variables. Gross tumour volume (GTV) was delineated on the magnetic resonance (MR) images acquired during MRgRT, and 1,067 radiomic features (RF) were extracted using the MODDICOM platform. The performance of RF in predicting 2yDFS was investigated in terms of the Wilcoxon–Mann–Whitney test and area under receiver operating characteristic (ROC) curve (AUC). Results 48 patients have been retrospectively enrolled, with 8 patients (16.7%) developing distant metastases at the 2-year follow-up. A total of 1,099 variables (1,067 RF and 32 clinical variables) were evaluated in two different models: radiomics and radiomics/clinical. The best-performing 2yDFS predictive model was a delta radiomics one, based on the variation in terms of area/surface ratio between biologically effective doses (BED) at 54 Gy and simulation (AUC of 0.92). Conclusions The results of this study suggest a promising role of delta radiomics analysis on 0.35-T MR images in predicting 2yDFS for LARC patients. Further analyses including larger cohorts of patients and an external validation are needed to confirm these preliminary results.
Collapse
Affiliation(s)
- Giuditta Chiloiro
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Luca Boldrini
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Francesco Preziosi
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Davide Cusumano
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Poonam Yadav
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Angela Romano
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Lorenzo Placidi
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Jacopo Lenkowicz
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Nicola Dinapoli
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Michael F Bassetti
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Maria Antonietta Gambacorta
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Vincenzo Valentini
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
35
|
Fernandes MC, Gollub MJ, Brown G. The importance of MRI for rectal cancer evaluation. Surg Oncol 2022; 43:101739. [PMID: 35339339 PMCID: PMC9464708 DOI: 10.1016/j.suronc.2022.101739] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 02/20/2022] [Indexed: 12/19/2022]
Abstract
Magnetic resonance imaging (MRI) has gained increasing importance in the management of rectal cancer over the last two decades. The role of MRI in patients with rectal cancer has expanded beyond the tumor-node-metastasis (TNM) system in both staging and restaging scenarios and has contributed to identifying "high" and "low" risk features that can be used to tailor and personalize patient treatment; for instance, selecting the patients for neoadjuvant chemoradiation (NCRT) before the total mesorectal excision (TME) surgery based on risk of recurrence. Among those features, the status of the circumferential resection margin (CRM), extramural vascular invasion (EMVI), and tumor deposits (TD) have stood out. Moreover, MRI also has played a role in surgical planning, especially when the tumor is located in the low rectum, when the relationship between tumor and the anal canal is important to choose the best surgical approach, and in cases of locally advanced or recurrent tumors invading adjacent pelvic organs that may require more complex surgeries such as pelvic exenteration. As approaches using organ preservation emerge, including transanal local excision and "watch-and-wait", MRI may help in the patient selection for those treatments, follow up, and detection of tumor regrowth. Additionally, potential MRI-based prognostic and predictive biomarkers, such as quantitative and semi-quantitative metrics derived from functional sequences like diffusion-weighted imaging (DWI) and dynamic contrast-enhanced (DCE), and radiomics, are under investigation. This review provides an overview of the current role of MRI in rectal cancer in staging and restaging and highlights the main areas under investigation and future perspectives.
Collapse
|
36
|
Mbanu P, Osorio EV, Mistry H, Malcomson L, Yousif S, Aznar M, Kochhar R, Van Herk M, Renehan AG, Saunders MP. Clinico-pathological predictors of clinical complete response in rectal cancer. Cancer Treat Res Commun 2022; 31:100540. [PMID: 35231874 DOI: 10.1016/j.ctarc.2022.100540] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 02/17/2022] [Accepted: 02/19/2022] [Indexed: 06/14/2023]
Abstract
PURPOSE Prediction of clinical complete response in rectal cancer before neoadjuvant chemo-radiotherapy treatment enables treatment selection. Patients predicted to have complete response could have chemo-radiotherapy, and others could have additional doublet chemotherapy at this stage of their treatment to improve their overall outcome. This work investigates the role of clinical variables in predicting clinical complete response. METHOD Using the UK-based OnCoRe database (2008 to 2019), we performed a propensity-score matched study of 322 patients who received neoadjuvant chemoradiotherapy. We collected pre-treatment clinic-pathological, inflammatory and radiotherapy-related characteristics. We determined the odds for the occurrence of cCR using conditional logistic regression models. We derived the post-model Area under the Curve (AUC) as an indicator of discrimination performance and stated a priori that an AUC of 0.75 or greater was required for potential clinical utility. RESULTS Pre-treatment tumour diameter, mrT-stage, haemoglobin, alkaline phosphate and total radiotherapy depths were associated with cCR on univariable and multivariable analysis. Additionally, neutrophil to lymphocyte ratio (NLR), neutrophil-monocyte to lymphocyte ratio (NMLR), lymphocyte count and albumin were all significantly associated with cCR on multivariable analysis. A nomogram using the above parameters was developed with a resulting ROC AUC of 0.75. CONCLUSION We identified routine clinic-pathological, inflammatory and radiotherapy-related variables which are independently associated with cCR. A nomogram was developed to predict cCR. The performance characteristics from this model were on the prior clinical utility threshold. Additional research is required to develop more associated variables to better select patients with rectal cancer undergoing chemoradiotherapy who may benefit from pursuing a W&W strategy.
Collapse
Affiliation(s)
- P Mbanu
- Department of Clinical Oncology, Christie NHS Foundation Trust, Manchester, United Kingdom.
| | - E Vasquez Osorio
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - H Mistry
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom; Division of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - L Malcomson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom; Colorectal and Peritoneal Oncology Centre, Christie NHS Foundation Trust, Manchester, United Kingdom
| | - S Yousif
- Department of Clinical Oncology, Lancashire Teaching Hospital, Preston, United Kingdom
| | - M Aznar
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - R Kochhar
- Department of Radiological Oncology, Christie NHS Foundation Trust, Manchester, United Kingdom
| | - M Van Herk
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - A G Renehan
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom; Colorectal and Peritoneal Oncology Centre, Christie NHS Foundation Trust, Manchester, United Kingdom
| | - M P Saunders
- Department of Clinical Oncology, Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
37
|
Contemporary Management of Locally Advanced and Recurrent Rectal Cancer: Views from the PelvEx Collaborative. Cancers (Basel) 2022; 14:1161. [PMID: 35267469 PMCID: PMC8909015 DOI: 10.3390/cancers14051161] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Pelvic exenteration is a complex operation performed for locally advanced and recurrent pelvic cancers. The goal of surgery is to achieve clear margins, therefore identifying adjacent or involved organs, bone, muscle, nerves and/or vascular structures that may need resection. While these extensive resections are potentially curative, they can be associated with substantial morbidity. Recently, there has been a move to centralize care to specialized units, as this facilitates better multidisciplinary care input. Advancements in pelvic oncology and surgical innovation have redefined the boundaries of pelvic exenterative surgery. Combined with improved neoadjuvant therapies, advances in diagnostics, and better reconstructive techniques have provided quicker recovery and better quality of life outcomes, with improved survival This article provides highlights of the current management of advanced pelvic cancers in terms of surgical strategy and potential future developments.
Collapse
|
38
|
Li M, Xiao Q, Venkatachalam N, Hofheinz RD, Veldwijk MR, Herskind C, Ebert MP, Zhan T. Predicting response to neoadjuvant chemoradiotherapy in rectal cancer: from biomarkers to tumor models. Ther Adv Med Oncol 2022; 14:17588359221077972. [PMID: 35222695 PMCID: PMC8864271 DOI: 10.1177/17588359221077972] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/14/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a major contributor to cancer-associated morbidity worldwide and over one-third of CRC is located in the rectum. Neoadjuvant chemoradiotherapy (nCRT) followed by surgical resection is commonly applied to treat locally advanced rectal cancer (LARC). In this review, we summarize current and novel concepts of neoadjuvant therapy for LARC such as total neoadjuvant therapy and describe how these developments impact treatment response. Moreover, as response to nCRT is highly divergent in rectal cancers, we discuss the role of potential predictive biomarkers. We review recent advances in biomarker discovery, from a clinical as well as a histopathological and molecular perspective. Furthermore, the role of emerging predictive biomarkers derived from the tumor environment such as immune cell composition and gut microbiome is presented. Finally, we describe how different tumor models such as patient-derived cancer organoids are used to identify novel predictive biomarkers for chemoradiotherapy (CRT) in rectal cancer.
Collapse
Affiliation(s)
- Moying Li
- Medical Faculty Mannheim, Heidelberg
University, Mannheim
| | - Qiyun Xiao
- Department of Medicine II, Mannheim University
Hospital, Medical Faculty Mannheim, Heidelberg University, Mannheim,
Germany
| | - Nachiyappan Venkatachalam
- Department of Medicine II, Mannheim University
Hospital, Medical Faculty Mannheim, Heidelberg University, Mannheim,
Germany
| | - Ralf-Dieter Hofheinz
- Department of Medicine III, Mannheim University
Hospital, Medical Faculty Mannheim, Heidelberg University, Mannheim,
GermanyMannheim Cancer Center, Medical Faculty Mannheim, Heidelberg
University, Mannheim, Germany
| | - Marlon R. Veldwijk
- Department of Radiation Oncology, Mannheim
University Hospital, Medical Faculty Mannheim, Heidelberg University,
Mannheim, Germany
| | - Carsten Herskind
- Department of Radiation Oncology, Mannheim
University Hospital, Medical Faculty Mannheim, Heidelberg University,
Mannheim, Germany
| | - Matthias P. Ebert
- Department of Medicine II, Mannheim University
Hospital, Medical Faculty Mannheim, Heidelberg University, Mannheim,
GermanyMannheim Cancer Center, Medical Faculty Mannheim, Heidelberg
University, Mannheim, GermanyDKFZ-Hector Cancer Institute, University
Medical Center Mannheim, Mannheim, Germany
| | - Tianzuo Zhan
- Department of Internal Medicine II, Mannheim
University Hospital, Medical Faculty Mannheim, Heidelberg University,
Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, GermanyMannheim Cancer Center,
Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
39
|
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
|
40
|
Iseas S, Sendoya JM, Robbio J, Coraglio M, Kujaruk M, Mikolaitis V, Rizzolo M, Cabanne A, Ruiz G, Salanova R, Gualdrini U, Méndez G, Antelo M, Carballido M, Rotondaro C, Viglino J, Eleta M, Di Sibio A, Podhajcer OL, Roca E, Llera AS, Golubicki M, Abba MC. Prognostic Impact of An Integrative Landscape of Clinical, Immune, and Molecular Features in Non-Metastatic Rectal Cancer. Front Oncol 2022; 11:801880. [PMID: 35071006 PMCID: PMC8777220 DOI: 10.3389/fonc.2021.801880] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Rectal Cancer (RC) is a complex disease that involves highly variable treatment responses. Currently, there is a lack of reliable markers beyond TNM to deliver a personalized treatment in a cancer setting where the goal is a curative treatment. Here, we performed an integrated characterization of the predictive and prognostic role of clinical features, mismatch-repair deficiency markers, HER2, CDX2, PD-L1 expression, and CD3-CD8+ tumor-infiltrating lymphocytes (TILs) coupled with targeted DNA sequencing of 76 non-metastatic RC patients assigned to total mesorectal excision upfront (TME; n = 15) or neoadjuvant chemo-radiotherapy treatment (nCRT; n = 61) followed by TME. Eighty-two percent of RC cases displayed mutations affecting cancer driver genes such as TP53, APC, KRAS, ATM, and PIK3CA. Good response to nCRT treatment was observed in approximately 40% of the RC cases, and poor pathological tumor regression was significantly associated with worse disease-free survival (DFS, HR = 3.45; 95%CI = 1.14-10.4; p = 0.028). High neutrophils-platelets score (NPS) (OR = 10.52; 95%CI=1.34-82.6; p = 0.025) and KRAS mutated cases (OR = 5.49; 95%CI = 1.06-28.4; p = 0.042) were identified as independent predictive factors of poor response to nCRT treatment in a multivariate analysis. Furthermore, a Cox proportional-hazard model showed that the KRAS mutational status was an independent prognostic factor associated with higher risk of local recurrence (HR = 9.68; 95%CI = 1.01-93.2; p <0.05) and shorter DFS (HR = 2.55; 95%CI = 1.05-6.21; p <0.05), while high CEA serum levels were associated with poor DFS (HR = 2.63; 95%CI = 1.01-6.85; p <0.05). Integrated clinical and molecular-based unsupervised analysis allowed us to identify two RC prognostic groups (cluster 1 and cluster 2) associated with disease-specific OS (HR = 20.64; 95%CI = 2.63-162.2; p <0.0001), metastasis-free survival (HR = 3.67; 95%CI = 1.22-11; p = 0.012), local recurrence-free survival (HR = 3.34; 95%CI = 0.96-11.6; p = 0.043) and worse DFS (HR = 2.68; 95%CI = 1.18-6.06; p = 0.012). The worst prognosis cluster 2 was enriched by stage III high-risk clinical tumors, poor responders to nCRT, with low TILs density and high frequency of KRAS and TP53 mutated cases compared with the best prognosis cluster 1 (p <0.05). Overall, this study provides a comprehensive and integrated characterization of non-metastatic RC cases as a new insight to deliver a personalized therapeutic approach.
Collapse
Affiliation(s)
- Soledad Iseas
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Juan M. Sendoya
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Juan Robbio
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
- Unidad de Investigación Traslacional, Laboratorio de Biología Molecular GENUIT, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Mariana Coraglio
- Proctology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Mirta Kujaruk
- Pathology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Vanesa Mikolaitis
- Pathology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Mariana Rizzolo
- Pathology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Ana Cabanne
- Pathology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Gonzalo Ruiz
- Biomakers Molecular Pathology and Research, Buenos Aires, Argentina
| | - Rubén Salanova
- Biomakers Molecular Pathology and Research, Buenos Aires, Argentina
| | - Ubaldo Gualdrini
- Proctology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Guillermo Méndez
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Marina Antelo
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Marcela Carballido
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Cecilia Rotondaro
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Julieta Viglino
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Martín Eleta
- Imaxe Image Diagnosis Center, Buenos Aires, Argentina
| | | | - Osvaldo L. Podhajcer
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Enrique Roca
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Andrea S. Llera
- Laboratorio de Terapia Molecular y Celular, Genocan, Fundación Instituto Leloir, IIBBA (CONICET), Buenos Aires, Argentina
| | - Mariano Golubicki
- Oncology Unit, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
- Unidad de Investigación Traslacional, Laboratorio de Biología Molecular GENUIT, Gastroenterology Hospital “Dr. Carlos Bonorino Udaondo”, Buenos Aires, Argentina
| | - Martín Carlos Abba
- Basic and Applied Immunological Research Center, School of Medical Sciences, National University of La Plata, La Plata, Argentina
| |
Collapse
|
41
|
Li JY, Huang XZ, Gao P, Song YX, Chen XW, Lv XE, Fu Y, Xiao Q, Ye SY, Wang ZN. Survival landscape of different tumor regression grades and pathologic complete response in rectal cancer after neoadjuvant therapy based on reconstructed individual patient data. BMC Cancer 2021; 21:1214. [PMID: 34773999 PMCID: PMC8590217 DOI: 10.1186/s12885-021-08922-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/27/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Neoadjuvant therapy can lead to different tumor regression grades (TRG) in rectal cancer after neoadjuvant therapy. The purposes of this study are to investigate the relationships among TRG, pathologic complete response (pCR) and long-term survival, on the basis of reconstructed individual patient data (IPD). METHODS The PubMed, Embase, Ovid and Cochrane CENTRAL databases were searched. The primary endpoint was to evaluate the survival landscape of different TRGs after neoadjuvant therapy and the secondary endpoint was to evaluate the associations between pCR and survival. IPD were reconstructed with Kaplan-Meier curves. RESULTS The 10-year overall survival (OS) and 5-year disease-free survival (DFS) were clearly higher in the pCR group than in the non-pCR (npCR) group (80.5% vs. 48.3, 90.1% vs. 69.8%). Furthermore, the OS and DFS increased with improvement in tumor regression after neoadjuvant therapy. According to the IPD, the pCR group had longer OS (HR = 0.240, 95% CI = 0.177-0.325, p < 0.001) and DFS (HR = 0.274, 95% CI = 0.205-0.367, p < 0.001) than the npCR group. Better tumor regression was associated with better survival outcomes (p < 0.005). Direct calculation of published HR values yielded similar results. CONCLUSIONS Our results indicate a positive relationship between better tumor regressions and improved survival benefits among the npCR group and patients with rectal cancer achieving pCR had much longer OS and DFS than patients achieving npCR, presenting a survival landscape of different TRGs and pCR in rectal cancer after neoadjuvant therapy.
Collapse
Affiliation(s)
- Jia-Yi Li
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Xuan-Zhang Huang
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Peng Gao
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Yong-Xi Song
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Xiao-Wan Chen
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Xing-Er Lv
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Yv Fu
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Qiong Xiao
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Shi-Yv Ye
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Zhen-Ning Wang
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang City, 110001, China.
| |
Collapse
|
42
|
Xu Y, Lou X, Liang Y, Zhang S, Yang S, Chen Q, Xu Z, Zhao M, Li Z, Zhao K, Liu Z. Predicting Neoadjuvant Chemoradiotherapy Response in Locally Advanced Rectal Cancer Using Tumor-Infiltrating Lymphocytes Density. J Inflamm Res 2021; 14:5891-5899. [PMID: 34785927 PMCID: PMC8591410 DOI: 10.2147/jir.s342214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Accumulating evidence revealed the predictive value of tumor-infiltrating lymphocytes (TILs) for neoadjuvant chemoradiotherapy (nCRT) response in solid tumors. This study quantified TILs density using hematoxylin and eosin (H&E) stained whole-slide images (WSIs) and investigated the predictive value of TILs density on nCRT response in locally advanced rectal cancer (LARC) patients. PATIENTS AND METHODS Two hundred and ten patients diagnosed with LARC were enrolled in this study. The density of TILs in the stroma region was quantified by a semi-automatic method in WSIs. Patients were stratified into low-TILs and high-TILs groups using the median value as the threshold. The tumor regression grade (TRG) was used to assess the response to nCRT in tumor resected specimens. Based on TRG, patients were classified into major-responder (TRG 0-1) and non-responder (TRG 2-3) groups. RESULTS The TILs density was significantly correlated with the nCRT response. Specifically, patients with high-TILs tend to have a higher major-responder rate than the low-TILs group (63.8% vs 47.6%, P = 0.026). Univariate analysis showed the TILs density was a predictor for the nCRT response (high vs low, odds ratio [OR] =1.94, 95% confidence interval 1.12-3.37, P = 0.019), and multivariate analysis further confirmed the correlation (adjusted odds ratio [AOR] = 2.41, 1.28-4.56, P = 0.007). CONCLUSION Patients with a high-TIL density have a higher major-responder rate than the low-TILs group, indicating patients with a strong immune response benefit more from nCRT. This semi-automatic method can facilitate the individualized preoperative prediction of the TRG for LARC patients before nCRT.
Collapse
Affiliation(s)
- Yao Xu
- School of Medicine, South China University of Technology, Guangzhou, 510006, People’s Republic of China
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
| | - Xiaoying Lou
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510665, People’s Republic of China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
| | - Shenyan Zhang
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510665, People’s Republic of China
| | - Shangqing Yang
- School of Life Science and Technology, Xidian University, Xian, 710071, People’s Republic of China
| | - Qicong Chen
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, 510006, People’s Republic of China
| | - Zeyan Xu
- School of Medicine, South China University of Technology, Guangzhou, 510006, People’s Republic of China
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
| | - Minning Zhao
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510080, People’s Republic of China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, People’s Republic of China
| | - Ke Zhao
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
| | - Zaiyi Liu
- School of Medicine, South China University of Technology, Guangzhou, 510006, People’s Republic of China
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
| |
Collapse
|
43
|
Morais M, Fonseca T, Machado-Neves R, Honavar M, Coelho AR, Lopes J, Barbosa E, Guerreiro E, Carneiro S. Can pretreatment blood biomarkers predict pathological response to neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer? Future Oncol 2021; 17:4947-4957. [PMID: 34734533 DOI: 10.2217/fon-2021-0707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aims: To investigate the value of previously described pretreatment hematological and biochemical biomarkers as predictors of pathological response. Methods: The authors performed a retrospective analysis of 191 patients with locally advanced rectal cancer who underwent long-course neoadjuvant chemoradiotherapy at two Portuguese centers. The authors performed logistic regression analysis to search for predictive markers of pathological complete and good response. Results: High platelet-neutrophil index (p = 0.042) and clinical tumor stage >2 (p = 0.015) were predictive of poor response. None of the analyzed biomarkers predicted pathological complete response in this study. Conclusion: A high platelet-neutrophil index before neoadjuvant chemoradiotherapy could help predict poorer pathological response in patients with locally advanced rectal cancer. However, no other blood biomarker predicted incomplete or poor response in this study.
Collapse
Affiliation(s)
- Marina Morais
- Surgery Department, Unidade Local de Saúde de Matosinhos
| | - Telma Fonseca
- Surgery Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | | | | | - Ana Rita Coelho
- Pathologic Anatomy Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Joanne Lopes
- Pathologic Anatomy Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Elisabete Barbosa
- Surgery Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | | | - Silvestre Carneiro
- Surgery Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| |
Collapse
|
44
|
Cheng Y, Luo Y, Hu Y, Zhang Z, Wang X, Yu Q, Liu G, Cui E, Yu T, Jiang X. Multiparametric MRI-based Radiomics approaches on predicting response to neoadjuvant chemoradiotherapy (nCRT) in patients with rectal cancer. Abdom Radiol (NY) 2021; 46:5072-5085. [PMID: 34302510 DOI: 10.1007/s00261-021-03219-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/15/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE To investigate the value of multiparametric MRI-based radiomics on predicting response to nCRT in patients with rectal cancer. METHODS This study enrolled 193 patients with pathologically confirmed LARC who received nCRT treatment between Apr. 2014 and Jun. 2018. All patients underwent baseline T1-weighted (T1W), T2-weighted (T2W) and T2-weighted fat-suppression (T2FS) MRI scans before neoadjuvant chemoradiotherapy. Radiomics features were extracted and selected from the MRI data to establish the radiomics signature. Important clinical predictors were identified by Mann-Whitney U test and Chi-square test. The nomogram integrating the radiomics signature and important clinical predictors was constructed using multivariate logistic regression. Prediction capabilities of each model were assessed with receiver operating characteristic (ROC) curve analysis. Performance of the nomogram was evaluated by its calibration and potential clinical usefulness. RESULTS For the prediction of good response (GR) and pathologic complete response (pCR), the developed radiomics signature comprising 10 and 7 features, respectively, were significantly associated with the therapeutic response to nCRT. The nomogram incorporating the radiomics signature and important clinical predictors (CEA and CA19-9 for predicting GR; CEA, posttreatment length and posttreatment thickness for predicting pCR) achieved favorable prediction efficacy, with AUCs of 0.918 (95% confidence interval [CI]: 0.867-0.971, Sen = 0.972, Spe = 0.828) and 0.944 (95% CI: 0.891-0.997, Sen = 0.943, Spe = 0.828) in the training and validation cohort for predicting GR, respectively; with AUCs of 0.959 (95% CI: 0.927-0.991, Sen = 1.000, Spe = 0.833) and 0.912 (95% CI: 0.843-0.982, Sen = 1.000, Spe = 0.815) in the training and validation cohort for predicting pCR, respectively. Decision curve analysis confirmed potential clinical usefulness of our nomogram. CONCLUSIONS This study demonstrated that the MRI-based radiomics nomogram is predictive of response to nCRT and can be considered as a promising tool for facilitating treatment decision-making for patients with LARC.
Collapse
Affiliation(s)
- Yuan Cheng
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yahong Luo
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Yue Hu
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110122, People's Republic of China
| | - Zhaohe Zhang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Xingling Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Qing Yu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Guanyu Liu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Enuo Cui
- School of Computer Science and Engineering, Shenyang University, Shenyang, 110044, People's Republic of China
| | - Tao Yu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Xiran Jiang
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
45
|
Pyo DH, Choi JY, Lee WY, Yun SH, Kim HC, Huh JW, Park YA, Shin JK, Cho YB. A Nomogram for Predicting Pathological Complete Response to Neoadjuvant Chemoradiotherapy Using Semiquantitative Parameters Derived From Sequential PET/CT in Locally Advanced Rectal Cancer. Front Oncol 2021; 11:742728. [PMID: 34676170 PMCID: PMC8523984 DOI: 10.3389/fonc.2021.742728] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/14/2021] [Indexed: 12/25/2022] Open
Abstract
We evaluated the predictive value of semiquantitative volumetric parameters derived from sequential PET/CT and developed a nomogram to predict pathological complete response (pCR) in patients with rectal cancer treated by neoadjuvant chemoradiotherapy (nCRT). From April 2008 to December 2013, among the patients who underwent nCRT, those who were taken sequential PET/CT before and after nCRT were included. MRI-based staging and semiquantitative parameters of PET/CT including standardized uptake value (SUV), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) were evaluated before and after nCRT. Multivariable analysis was performed to select significant predictors to construct a nomogram. Sensitivity, specificity, accuracy, and area under the receiver operating characteristics curve (AUC) of the model were evaluated to determine its performance. Among 137 eligible patients, 17 (12.4%) had pCR. All post-PET/CT parameters showed significant differences between pCR and non-pCR groups. Patients were randomly assigned to a training group (91 patients) and a validation group (46 patients). In multivariable analysis with the training group, post-CEA, post-MRI T staging, post-SUVmax, and post-MTV were significantly associated with pCR. There was no significant pre-nCRT variable for predicting pCR. Using significant predictors, a nomogram was developed. Sensitivity, specificity, accuracy, and AUC of the nomogram were 0.882, 0.808, 0.848, and 0.884 with the training group and 0.857, 0.781, 0.783, and 0.828 with the validation group, respectively. This model showed the better performance than other predictive models that did not contain PET/CT parameters. A nomogram containing semiquantitative post-PET/CT could effectively select candidates for organ-sparing strategies.
Collapse
Affiliation(s)
- Dae Hee Pyo
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Woo Yong Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
| | - Seong Hyeon Yun
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jung Wook Huh
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yoon Ah Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jung Kyong Shin
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea.,Department of Biopharmaceutical Convergence, Sungkyunkwan University, Seoul, South Korea
| |
Collapse
|
46
|
Watch and Wait Approach for Rectal Cancer Following Neoadjuvant Treatment: The Experience of a High Volume Cancer Center. Diagnostics (Basel) 2021; 11:diagnostics11081507. [PMID: 34441441 PMCID: PMC8394713 DOI: 10.3390/diagnostics11081507] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/07/2021] [Accepted: 08/08/2021] [Indexed: 11/17/2022] Open
Abstract
Multimodal treatments for rectal cancer, along with significant research on predictors to response to therapy, have led to more conservative surgical strategies. We describe our experience of the rectal sparing approach in rectal cancer patients with clinical complete response (cCR) after neoadjuvant treatment. We also specifically highlight our clinical and imaging criteria to select patients for the watch and wait strategy (w&w). Data came from 39 out of 670 patients treated for locally advanced rectal cancer between January 2016 until February 2020. The selection criteria were a clinical complete response after neoadjuvant chemotherapy managed with a watch and wait (w&w) strategy. A strict follow-up period was adopted in these selected patients and follow-ups were performed every three months during the first two years and every six months after that. The median follow-up time was 28 months. Six patients had a local recurrence (15.3%); all were salvageable by total mesorectal excision (TME). Five patients had a distant metastasis (12.8%). There was no local unsalvageable disease after w&w strategy. The rectal sparing approach in patients with clinical complete response after neoadjuvant treatment is the best possible treatment and is appropriate to analyze from this perspective. The watch and wait approach after neoadjuvant treatment for rectal cancer can be successfully explored after inflexible and strict patient selection.
Collapse
|
47
|
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
|
48
|
Dayde D, Gunther J, Hirayama Y, Weksberg DC, Boutin A, Parhy G, Aguilar-Bonavides C, Wang H, Katayama H, Abe Y, Do KA, Hara K, Kinoshita T, Komori K, Shimizu Y, Tajika M, Niwa Y, Wang YA, DePinho R, Hanash S, Krishnan S, Taguchi A. Identification of Blood-Based Biomarkers for the Prediction of the Response to Neoadjuvant Chemoradiation in Rectal Cancer. Cancers (Basel) 2021; 13:cancers13143642. [PMID: 34298853 PMCID: PMC8306983 DOI: 10.3390/cancers13143642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Although pathologic complete response (pCR) to neoadjuvant chemoradiation (nCRT) in locally advanced rectal cancer (LARC) is associated with better outcomes, a subset of tumors exhibit resistance to nCRT. Therefore, there is a need of biomarkers to predict the nCRT response and increment efforts for personalized therapeutic options. To this end, we analyzed pretreatment plasma proteome of a mouse model of rectal cancer treated with concurrent chemoradiation, resulting in identification and validation of plasma VEGFR3 as a potential predicting biomarker. In addition, plasma levels of EGFR and COX2, previously validated tissue-based predicting biomarkers, were significantly higher in non-pCR than pCR LARC patients, indicating that EGFR and COX2 can also serve as blood-based biomarkers. The performance of the biomarker panel combining VEGFR3, EGFR, and COX2 were significantly improved compared to that of each marker alone, providing a rationale for further integration and refinement of the biomarker panel and validation in the larger sample sets. Abstract The current standard of care for patients with locally advanced rectal cancer (LARC) is neoadjuvant chemoradiation (nCRT) followed by total mesorectal excision surgery. However, the response to nCRT varies among patients and only about 20% of LARC patients achieve a pathologic complete response (pCR) at the time of surgery. Therefore, there is an unmet need for biomarkers that could predict the response to nCRT at an early time point, allowing for the selection of LARC patients who would or would not benefit from nCRT. To identify blood-based biomarkers for prediction of nCRT response, we performed in-depth quantitative proteomic analysis of pretreatment plasma from mice bearing rectal tumors treated with concurrent chemoradiation, resulting in the quantification of 567 proteins. Among the plasma proteins that increased in mice with residual rectal tumor after chemoradiation compared to mice that achieved regression, we selected three proteins (Vascular endothelial growth factor receptor 3 [VEGFR3], Insulin like growth factor binding protein 4 [IGFBP4], and Cathepsin B [CTSB]) for validation in human plasma samples. In addition, we explored whether four tissue protein biomarkers previously shown to predict response to nCRT (Epidermal growth factor receptor [EGFR], Ki-67, E-cadherin, and Prostaglandin G/H synthase 2 [COX2]) also act as potential blood biomarkers. Using immunoassays for these seven biomarker candidates as well as Carcinoembryonic antigen [CEA] levels on plasma collected before nCRT from 34 patients with LARC (6 pCR and 28 non-pCR), we observed that levels of VEGFR3 (p = 0.0451, AUC = 0.720), EGFR (p = 0.0128, AUC = 0.679), and COX2 (p = 0.0397, AUC = 0.679) were significantly increased in the plasma of non-pCR LARC patients compared to those of pCR LARC patients. The performance of the logistic regression model combining VEGFR3, EGFR, and COX2 was significantly improved compared with the performance of each biomarker, yielding an AUC of 0.869 (sensitivity 43% at 95% specificity). Levels of VEGFR3 and EGFR were significantly decreased 5 to 7 months after tumor resection in plasma from 18 surgically resected rectal cancer patients, suggesting that VEGFR3 and EGFR may emanate from tumors. These findings suggest that circulating VEGFR3 can contribute to the prediction of the nCRT response in LARC patients together with circulating EGFR and COX2.
Collapse
Affiliation(s)
- Delphine Dayde
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.D.); (G.P.)
| | - Jillian Gunther
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.G.); (D.C.W.); (S.K.)
| | - Yutaka Hirayama
- Department of Endoscopy, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan; (Y.H.); (M.T.); (Y.N.)
| | - David C. Weksberg
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.G.); (D.C.W.); (S.K.)
- UPMC Pinnacle Radiation Oncology, Harrisburg, PA 17109, USA
| | - Adam Boutin
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.B.); (A.Y.W.); (R.D.)
| | - Gargy Parhy
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.D.); (G.P.)
| | - Clemente Aguilar-Bonavides
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.A.-B.); (K.-A.D.)
| | - Hong Wang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (H.W.); (H.K.); (S.H.)
- Hangzhou Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou 311200, China
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (H.W.); (H.K.); (S.H.)
| | - Yuichi Abe
- Division of Molecular Diagnostics, Aichi Cancer Center, Nagoya 464-8681, Japan;
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.A.-B.); (K.-A.D.)
| | - Kazuo Hara
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan;
| | - Takashi Kinoshita
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan; (T.K.); (K.K.); (Y.S.)
| | - Koji Komori
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan; (T.K.); (K.K.); (Y.S.)
| | - Yasuhiro Shimizu
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan; (T.K.); (K.K.); (Y.S.)
| | - Masahiro Tajika
- Department of Endoscopy, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan; (Y.H.); (M.T.); (Y.N.)
| | - Yasumasa Niwa
- Department of Endoscopy, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan; (Y.H.); (M.T.); (Y.N.)
| | - Y. Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.B.); (A.Y.W.); (R.D.)
| | - Ronald DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.B.); (A.Y.W.); (R.D.)
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (H.W.); (H.K.); (S.H.)
| | - Sunil Krishnan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.G.); (D.C.W.); (S.K.)
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Ayumu Taguchi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.D.); (G.P.)
- Division of Molecular Diagnostics, Aichi Cancer Center, Nagoya 464-8681, Japan;
- Division of Advanced Cancer Diagnostics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Correspondence: ; Tel.: +81-52-764-9884
| |
Collapse
|
49
|
Mor E, Assaf D, Laks S, Benvenisti H, Schtrechman G, Hazzan D, Segev L, Yaka R, Shacham-Shmueli E, Margalit O, Halpern N, Perelson D, Kaufmann MI, Ben-Yaacov A, Nissan A, Adileh M. Ratio of Pathological Response to Preoperative Chemotherapy in Patients Undergoing Complete Cytoreduction and Hyperthermic Intraperitoneal Chemotherapy for Metastatic Colorectal Cancer Correlates with Survival. Ann Surg Oncol 2021; 28:9138-9147. [PMID: 34232423 DOI: 10.1245/s10434-021-10367-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pathological response of colorectal peritoneal metastasis (CRPM) may affect prognosis. We investigated the relationship between oncological outcomes and pathological response to chemotherapy of CRPM following cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). METHODS We conducted a retrospective analysis of a prospectively maintained Peritoneal Surface Malignancies database between 2015 and 2020. Analysis included patients with CRPM who underwent a CRS/HIPEC procedure (n = 178). The cohort was divided into three groups according to the response ratio (ratio of tumor-positive specimens to the total number of specimens resected): Group A, complete response; Group B, high response ratio, and Group C, low response ratio. RESULTS The group demographics were similar, but the overall complication rate was higher in Group C (65.2%) compared with Groups A (55%) and B (42.8%) [p = 0.03]. Survival correlated to response ratio; the estimated median disease-free survival of Group C was 9.1 months (5.97-12.23), 14.9 months (4.72-25.08) for Group B, and was not reached in Group A (p = 0.001). The estimated median overall survival in Group C was 35 months (26.69-43.31), and was not reached in Groups A and B (p = 0.001). CONCLUSIONS The pathological response ratio to systemic therapy correlates with survival in patients undergoing CRS/HIPEC. This study supports the utilization of preoperative therapy for better patient selection, with a potential impact on survival.
Collapse
Affiliation(s)
- Eyal Mor
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Dan Assaf
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Shachar Laks
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Haggai Benvenisti
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Gal Schtrechman
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - David Hazzan
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Lior Segev
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Ronel Yaka
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Einat Shacham-Shmueli
- The Department of Oncology, Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Ofer Margalit
- The Department of Oncology, Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Naama Halpern
- The Department of Oncology, Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Daria Perelson
- The Department of Anesthesiology, Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Monica-Inda Kaufmann
- The Department of Pathology, Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Almog Ben-Yaacov
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Aviram Nissan
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel
| | - Mohammad Adileh
- The Department of General and Oncological Surgery, Surgery C Sheba Medical Center, Affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Hashomer, Israel.
| |
Collapse
|
50
|
Kumar R, Harilal S, Carradori S, Mathew B. A Comprehensive Overview of Colon Cancer- A Grim Reaper of the 21st Century. Curr Med Chem 2021; 28:2657-2696. [PMID: 33106132 DOI: 10.2174/0929867327666201026143757] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 12/09/2022]
Abstract
A few decades ago, the incidence of colorectal cancer (CRC) was low and is now the fourth in the list of deadly cancers producing nearly a million deaths annually. A population that is aging along with risk factors such as smoking, obesity, sedentary lifestyle with little or no physical activity, and non-healthy food habits of developed countries can increase the risk of colorectal cancer. The balance in gut microbiota and the metabolites produced during bacterial fermentation within the host plays a significant role in regulating intestinal diseases as well as colorectal cancer development. Recent progress in the understanding of illness resulted in multiple treatment options such as surgery, radiation, and chemotherapy, including targeted therapy and multitherapies. The treatment plan for CRC depends on the location, stage and grade of cancer as well as genomic biomarker tests. Despite all the advancements made in the genetic and molecular aspects of the disease, the knowledge seems inadequate as the drug action as well as the wide variation in drug response did not appear strongly correlated with the individual molecular and genetic characteristics, which suggests the requirement of comprehensive molecular understanding of this complex heterogeneous disease. Furthermore, multitherapies or a broad spectrum approach, which is an amalgamation of the various promising as well as effective therapeutic strategies that can tackle heterogeneity and act on several targets of the disease, need to be validated in clinical studies. The latest treatment options have significantly increased the survival of up to three years in the case of advanced disease. The fact that colorectal cancer is developed from a polypoid precursor, as well as the symptoms of the disease that occur at an advanced stage, underlines how screening programs can help early detection and decrease mortality as well as morbidity from CRC.
Collapse
Affiliation(s)
- Rajesh Kumar
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Seetha Harilal
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Simone Carradori
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| |
Collapse
|