1
|
Roesel R, Strati F, Basso C, Epistolio S, Spina P, Djordjevic J, Sorrenti E, Villa M, Cianfarani A, Mongelli F, Galafassi J, Popeskou SG, Facciotti F, Caprera C, Melle F, Majno-Hurst PE, Franzetti-Pellanda A, De Dosso S, Bonfiglio F, Frattini M, Christoforidis D, Iezzi G. Combined tumor-associated microbiome and immune gene expression profiling predict response to neoadjuvant chemo-radiotherapy in locally advanced rectal cancer. Oncoimmunology 2025; 14:2465015. [PMID: 39992705 PMCID: PMC11853554 DOI: 10.1080/2162402x.2025.2465015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Locally advanced rectal cancer (LARC) is treated with neoadjuvant chemo-radiotherapy (nCRT) followed by surgery. A minority of patients show complete response (CR) to nCRT and may avoid surgery and its functional consequences. Instead, most patients show non-complete response (non-CR) and may benefit from additional treatments to increase CR rates. Reliable predictive markers are lacking. Aim of this study was to identify novel signatures predicting nCRT responsiveness. We performed a combined analysis of tumor-associated microbiome and immune gene expression profiling of diagnostic biopsies from 70 patients undergoing nCRT followed by rectal resection, including 16 with CR and 54 with non-CR. Findings were validated by an independent cohort of 49 patients, including 7 with CR and 42 with non-CR. Intratumoral microbiota significantly differed between CR and non-CR groups at genus and species level. Colonization by bacterial species of Ruminococcus genera was consistently associated with CR, whereas abundance of Fusobacterium, Porhpyromonas, and Oscillibacter species predicted non-CR. Immune gene profiling revealed a panel of 59 differentially expressed genes and significant upregulation of IFN-gamma and -alpha response in patients with CR. Integrated microbiome and immune gene profiling analysis unraveled clustering of microbial taxa with each other and with immune cell-related genes and allowed the identification of a combined signature correctly identifying non-CRS in both cohorts. Thus, combined intratumoral microbiome-immune profiling improves the prediction of response to nCRT. Correct identification of unresponsive patients and of bacteria promoting responsiveness might lead to innovative therapeutic approaches based on gut microbiota pre-conditioning to increase nCRT effectiveness in LARC.
Collapse
Affiliation(s)
- Raffaello Roesel
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Francesco Strati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Camilla Basso
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Samantha Epistolio
- Laboratory of Molecular Pathology, Institute of Pathology, Locarno, Switzerland
| | - Paolo Spina
- Laboratory of Molecular Pathology, Institute of Pathology, Locarno, Switzerland
| | - Julija Djordjevic
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Elisa Sorrenti
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Martina Villa
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Agnese Cianfarani
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Francesco Mongelli
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Jacopo Galafassi
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Sotirios G. Popeskou
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Federica Facciotti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Cecilia Caprera
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Melle
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Pietro Edoardo Majno-Hurst
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | | | - Sara De Dosso
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Ferdinando Bonfiglio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples, Naples, Italy
- CEINGE Advanced Biotechnology Franco Salvatore, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Milo Frattini
- Laboratory of Molecular Pathology, Institute of Pathology, Locarno, Switzerland
| | - Dimitrios Christoforidis
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Visceral Surgery, CHUV, University of Lausanne, Lausanne, Switzerland
| | - Giandomenica Iezzi
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
2
|
Liu IC, Gearhart S, Hu C, Chung H, Gabre-Kidan A, Najjar P, Christenson ES, Azad NS, Lee V, Zaheer A, Birkness-Gartman JE, Narang AK, Meyer J. Sustained Organ Preservation in Patients With Rectal Cancer After Sequential Short-Course Radiation Therapy and Chemotherapy. Am Surg 2025:31348251341945. [PMID: 40340411 DOI: 10.1177/00031348251341945] [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: 05/10/2025]
Abstract
BackgroundNonoperative management in patients with rectal cancer with complete response to radiation therapy and chemotherapy is of increasing interest. Most of the data on nonoperative management have centered around patients treated with long-course chemoradiotherapy. The ability of short-course radiation-based treatment courses to achieve durable complete responses with sustained organ preservation is less defined. This study updates our institution's long-term experience with nonoperative management following upfront short-course radiation therapy and sequential/consolidation chemotherapy.MethodsWe retrospectively reviewed patients with nonmetastatic rectal cancer treated with sequential short-course radiation therapy and chemotherapy who reached complete response and were subsequently followed with nonoperative management. We report on disease control outcomes, including rates of regrowth and results of salvage surgery. We investigated characteristics associated with local tumor regrowth.ResultsOur study included 52 patients. The 2-year freedom from local regrowth for the entire cohort was 75%. Notably, patients with high-risk disease characteristics at diagnosis exhibited a trend toward a higher rate of local tumor regrowth. No patient with sustained clinical complete response developed metastatic disease. Of the twelve patients undergoing surgical salvage for regrowth of disease, ten were resected with complete/near-complete total mesorectal surgical specimens with negative margins.ConclusionsThe optimal approach to achieving sustained organ preservation through the use of radiation therapy and chemotherapy is not well defined. Our findings show the viability of neoadjuvant therapy incorporating short-course radiation therapy to achieve durable complete responses.
Collapse
Affiliation(s)
- I-Chia Liu
- Department of Radiation Oncology & Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susan Gearhart
- Department of Surgery, Colorectal Research Unit, Ravitch Division of Colorectal Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chen Hu
- Division of Biostatistics and Bioinformatics, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Haniee Chung
- Department of Surgery, Colorectal Research Unit, Ravitch Division of Colorectal Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alodia Gabre-Kidan
- Department of Surgery, Colorectal Research Unit, Ravitch Division of Colorectal Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter Najjar
- Department of Surgery, Colorectal Research Unit, Ravitch Division of Colorectal Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric S Christenson
- Department of Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Nilofer S Azad
- Department of Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Valerie Lee
- Department of Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Atif Zaheer
- Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Amol K Narang
- Department of Radiation Oncology & Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey Meyer
- Department of Radiation Oncology & Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Natout M, Machmouchi A, Hussain H, Chehade L, Abbas N, Turfa R, Kattan J, Temraz S, Tawil A, Elkhaldi M, Jaber O, Amarin R, Alawabdeh T, Charafeddine M, Al Darazi M, Shamseddine A. Assessment of Immunoscore, MRI Tumor Regression Grade, and Neoadjuvant Rectal Score in Predicting Pathologic Response in Locally Advanced Rectal Cancer in the Averectal Study. Diagnostics (Basel) 2025; 15:913. [PMID: 40218263 PMCID: PMC11989207 DOI: 10.3390/diagnostics15070913] [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: 03/05/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Predictive tools are needed to assess the response to treatment and guide treatment decisions for locally advanced rectal cancer (LARC). This study explores the value of combining the immunoscore (IS) and magnetic resonance imaging tumor regression grade (mrTRG) with pathologic and radiologic neoadjuvant rectal (NAR) scores in predicting pathologic complete response (pCRs). Methods: The scores were assessed for patients with LARC enrolled in the Averectal study (NCT03503630), who received five fractions of short-course radiotherapy, followed by six cycles of mFOLFOX-6 plus avelumab, and total mesorectal excision. The IS was calculated using the mean density percentiles of CD3- and CD8-positive T-cells on baseline biopsy samples. Baseline and post-treatment MRIs were reviewed to measure the mrTRG. NAR scores were calculated using the pre-treatment T stage and post-treatment pathologic and radiologic N and T stages. Results: Fifteen out of thirty-five patients whose data were available achieved pCR (42.8%), and seven out of fourteen patients with mrTRG = 1 (complete response) attained pCR. In patients with both a mrTRG = 1 and high IS, the pCR rate was 66.7% (6/9). All of the patients who achieved pCR had a low or intermediate pathologic NAR score with a significant correlation between pCR and pathologic NAR scores (p < 0.0001). Both pathologic and radiologic NAR scores were correlated with overall survival and disease-free survival. Conclusions: The IS can supplement the mrTRG to better predict TNT outcomes, along with the use of the NAR score. This combination could potentially help with patient selection for non-operative management and guide treatment strategies for those with different recurrence risks.
Collapse
Affiliation(s)
- Mustafa Natout
- Department of Diagnostic Radiology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
| | - Ahmad Machmouchi
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute—NKBCI, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (A.M.); (L.C.); (N.A.); (S.T.); (M.C.); (M.A.D.)
| | - Hero Hussain
- Department of Abdominal Radiology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Laudy Chehade
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute—NKBCI, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (A.M.); (L.C.); (N.A.); (S.T.); (M.C.); (M.A.D.)
| | - Noura Abbas
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute—NKBCI, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (A.M.); (L.C.); (N.A.); (S.T.); (M.C.); (M.A.D.)
| | - Rim Turfa
- Department of Internal Medicine, Division of Hematology/Oncology, King Hussein Cancer Center, Amman 11941, Jordan; (R.T.); (R.A.); (T.A.)
| | - Joseph Kattan
- Department of Hematology/Oncology, Hotel-Dieu de France University Hospital, Beirut 166830, Lebanon;
| | - Sally Temraz
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute—NKBCI, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (A.M.); (L.C.); (N.A.); (S.T.); (M.C.); (M.A.D.)
| | - Ayman Tawil
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
| | - Mousa Elkhaldi
- Department of Radiation Oncology, King Hussein Cancer Center, Amman 11941, Jordan;
| | - Omar Jaber
- Department of Pathology, King Hussein Cancer Center, Amman 11941, Jordan;
| | - Rula Amarin
- Department of Internal Medicine, Division of Hematology/Oncology, King Hussein Cancer Center, Amman 11941, Jordan; (R.T.); (R.A.); (T.A.)
| | - Tala Alawabdeh
- Department of Internal Medicine, Division of Hematology/Oncology, King Hussein Cancer Center, Amman 11941, Jordan; (R.T.); (R.A.); (T.A.)
| | - Maya Charafeddine
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute—NKBCI, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (A.M.); (L.C.); (N.A.); (S.T.); (M.C.); (M.A.D.)
| | - Monita Al Darazi
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute—NKBCI, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (A.M.); (L.C.); (N.A.); (S.T.); (M.C.); (M.A.D.)
| | - Ali Shamseddine
- Department of Internal Medicine, Division of Hematology/Oncology, Naef K. Basile Cancer Institute—NKBCI, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (A.M.); (L.C.); (N.A.); (S.T.); (M.C.); (M.A.D.)
| |
Collapse
|
4
|
Westwood AC, Wilson BI, Laye J, Grabsch HI, Mueller W, Magee DR, Quirke P, West NP. Deep-learning enabled combined measurement of tumour cell density and tumour infiltrating lymphocyte density as a prognostic biomarker in colorectal cancer. BJC REPORTS 2025; 3:12. [PMID: 40033106 DOI: 10.1038/s44276-025-00123-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/20/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Within the colorectal cancer (CRC) tumour microenvironment, tumour infiltrating lymphocytes (TILs) and tumour cell density (TCD) are recognised prognostic markers. Measurement of TILs and TCD using deep-learning (DL) on haematoxylin and eosin (HE) whole slide images (WSIs) could aid management. METHODS HE WSIs from the primary tumours of 127 CRC patients were included. DL was used to quantify TILs across different regions of the tumour and TCD at the luminal surface. The relationship between TILs, TCD, and cancer-specific survival was analysed. RESULTS Median TIL density was higher at the invasive margin than the luminal surface (963 vs 795 TILs/mm2, P = 0.010). TILs and TCD were independently prognostic in multivariate analyses (HR 4.28, 95% CI 1.87-11.71, P = 0.004; HR 2.72, 95% CI 1.19-6.17, P = 0.017, respectively). Patients with both low TCD and low TILs had the poorest survival (HR 10.0, 95% CI 2.51-39.78, P = 0.001), when compared to those with a high TCD and TILs score. CONCLUSIONS DL derived TIL and TCD score were independently prognostic in CRC. Patients with low TILs and TCD are at the highest risk of cancer-specific death. DL quantification of TILs and TCD could be used in combination alongside other validated prognostic biomarkers in routine clinical practice.
Collapse
Affiliation(s)
- Alice C Westwood
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | | | - Jon Laye
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Heike I Grabsch
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, Netherlands
| | | | | | - Phillip Quirke
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Nicholas P West
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.
| |
Collapse
|
5
|
Zeng Q, Yang YX, Tang Y, Li N, Lu NN, Wang SL, Li YX, Jin J, Zou SM, Liu WY. Prognostic and Predictive Value of a Modified Diagnostic Biopsy-Adapted Immunoscore in Patients with Rectal Cancer After Neoadjuvant Treatment: A Translational Study From the STELLAR Trial. Int J Radiat Oncol Biol Phys 2025; 121:640-650. [PMID: 39426571 DOI: 10.1016/j.ijrobp.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/28/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE The purpose of this study was to assess the prognostic significance of the modified diagnostic biopsy-adapted immunoscore (mISb) in determining the outcomes for patients with locally advanced rectal cancer (LARC) in a neoadjuvant setting. METHODS AND MATERIALS We included 181 patients with LARC from a single subcenter of a prospective study comparing total neoadjuvant therapy (TNT) based on short-course radiation therapy with long-term chemoradiation therapy (CRT). Tumor biopsies at baseline were stained for CD8+ and CD3+ T-cell densities. The mISb was developed using mean percentile of CD8+ T-cell density and CD8/CD3 ratio. Patients were classified into low (0%-25%), intermediate (>25%-70%), and high (>70%-100%) in both groups. The relativity among different lymphocytes and their correlation with survival were illustrated. Survival analyses and Cox regression models were used to compare the prognostic values of mISb and diagnostic biopsy immunoscore for survival outcomes and to assess the role of mISb in TNT and CRT subgroups, respectively. RESULTS In this study, 151 (83.4%) patients received surgery, and 30 (16.6%) followed a watch and wait strategy. A strong correlation was found between CD8+ and CD3+ T-cell densities (R = 0.86; P < .001), whereas a weak correlation was witnessed between CD8+ and CD8/CD3 ratio (R = 0.45). The 3-year disease-free survival for the entire cohort was 69.9%, with 57.2%, 68.6%, and 85.5% for the low, intermediate, and high mISb groups, respectively (P = .01), whereas diagnostic biopsy immunoscore failed to distinguish survival outcomes. Multivariate analysis revealed mISb to be an independent prognostic factor for disease-free survival in surgically treated patients (P = .01). Specifically, patients with high mISb score showed longer progression-free survival than other subgroups in the TNT cohort (P = .049), but no significant difference was found in the CRT population. CONCLUSIONS In this study, mISb demonstrated significant prognostic value in patients with LARC receiving preoperative therapies, especially in the TNT subgroup. These findings may help tailor the intensity of neoadjuvant therapy for patients.
Collapse
Affiliation(s)
- Qiang Zeng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue-Xin Yang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yuan Tang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning-Ning Lu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Lian Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye-Xiong Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.
| | - Shuang-Mei Zou
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Wen-Yang Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Zhao R, Shen W, Zhao W, Peng W, Wan L, Chen S, Liu X, Wang S, Zou S, Zhang R, Zhang H. Integrating radiomics, pathomics, and biopsy-adapted immunoscore for predicting distant metastasis in locally advanced rectal cancer. ESMO Open 2025; 10:104102. [PMID: 39951928 PMCID: PMC11874550 DOI: 10.1016/j.esmoop.2024.104102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND This study aimed to develop and validate a nomogram that utilized macro- and microscopic tumor characteristics at baseline, including radiomics, pathomics, and biopsy-adapted immunoscore (ISB), to accurately predict distant metastasis (DM) in patients with locally advanced rectal cancer (LARC) who underwent neoadjuvant chemoradiotherapy (nCRT). MATERIALS AND METHODS In total, 201 patients with LARC (91 months of median follow-up) were enrolled. Radiomics features were extracted from apparent diffusion coefficient maps and T2-weighted images. Pathomics features including global pattern (features of the entire image) and local pattern (features of the tumor nuclei) were extracted from whole-slide images of hematoxylin-eosin-stained biopsy specimens. ISB was calculated from the densities of CD3+ and CD8+ T cells in the tumor region using immunohistochemistry on biopsy specimens. The construction of a predictive model was carried out using the least absolute shrinkage and selection operator-Cox analysis, with performance metrics including the area under the curve (AUC) and concordance index (C-index) utilized for evaluation. RESULTS Compared with patients with moderate and high ISB, patients with low ISB exhibited significantly higher risk scores for radiomics and pathomics signatures. The nomogram showed respective C-indexes of 0.902 and 0.848 for 5-year DM-free survival in the training and test sets, along with corresponding AUC values of 0.950 and 0.872. Patients could be efficiently categorized into low- and high-risk groups for developing DM using the nomogram. CONCLUSIONS The nomogram integrating macroscopic radiological information and microscopic pathological information is effective for risk stratification at baseline in LARC treated with nCRT.
Collapse
Affiliation(s)
- R Zhao
- Departments of Diagnositic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - W Shen
- Departments of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - W Zhao
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China; The Institute of Translational Medicine, Tianjin Union Medical Center, Nankai University, Tianjin, China; Tianjin Institute of Coloproctology, Tianjin, China
| | - W Peng
- Departments of Diagnositic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - L Wan
- Departments of Diagnositic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - S Chen
- Departments of Diagnositic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - X Liu
- Departments of Diagnositic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - S Wang
- Department of Pharmaceutical Diagnosis, GE Healthcare, Life Sciences, Beijing, China
| | - S Zou
- Departments of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - R Zhang
- Departments of Diagnositic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - H Zhang
- Departments of Diagnositic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
7
|
Xu N, Zhang JX, Zhang JJ, Huang Z, Mao LC, Zhang ZY, Jin WD. The prognostic value of the neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) in colorectal cancer and colorectal anastomotic leakage patients: a retrospective study. BMC Surg 2025; 25:57. [PMID: 39910526 PMCID: PMC11796187 DOI: 10.1186/s12893-024-02708-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/09/2024] [Indexed: 02/07/2025] Open
Abstract
OBJECTIVE The purpose of this study was to investigate the influence and predictive value of preoperative peripheral blood neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) index on the prognosis of colorectal anastomotic leakage (CAL) patients. METHODS This study retrospectively analyzed the clinical data of 1016 patients who underwent radical resection for colorectal cancer at a single center between January 1, 2007 and December 31, 2023. In this study, NLR and PLR were analyzed before surgery. Kaplan-Meier survival analysis was performed according to the postoperative survival status of the patients. Nomogram and calibration curve were established by proportional hazards model (COX) to verify its predictive value. RESULTS A total of 890 patients with colorectal cancer, 102 patients with CAL, and 788 patients with non- anastomotic leakage (AL) colorectal cancer were enrolled for a median follow-up of 96 months (quartile range 33-133). In this study, COX regression analysis showed that preoperative NLR and PLR could predict the prognosis of CAL patients, and the optimal cut-off points of NLR and PLR were 2.89 and 157.62, respectively. Kaplan-Meier survival curve results showed that 5-year overall survival (OS) and disease-free survival (DFS) in the low NLR and PLR group were significantly higher than those in the high NLR and PLR group. OS and DFS were divided into high, low NLR and PLR groups. Finally, based on COX model, a nomogram analysis was conducted to analyze the risk factors affecting OS and DFS, and the accuracy and practicality of the model were verified by calibration curve and decision curve. CONCLUSION Preoperative NLR and PLR can predict the long-term prognosis of colorectal cancer (CRC) and CAL patients, and patients with NLR ≥ 2.89 and PLR ≥ 157.62 have poor survival prognosis. Nomogram and calibration curve analysis will further improve the accuracy of OS and DFS prediction.
Collapse
Affiliation(s)
- Nuo Xu
- Wuhan University of Science and Technology, School of Medicine, Wuhan, Hubei, 430081, China
| | - Jian-Xin Zhang
- Chinese People's Liberation Army Central Theater General Hospital General Surgery Department, Wuhan, Hubei, 4300770, China
| | - Jia-Jie Zhang
- Department of Statistics and Applied Probability, University of California, Santa Barbara, CA, 93106, USA
| | - Zhuo Huang
- Chinese People's Liberation Army Central Theater General Hospital General Surgery Department, Wuhan, Hubei, 4300770, China
| | - Lian-Chun Mao
- Chinese People's Liberation Army Central Theater General Hospital General Surgery Department, Wuhan, Hubei, 4300770, China
| | - Zhi-Yong Zhang
- Chinese People's Liberation Army Central Theater General Hospital General Surgery Department, Wuhan, Hubei, 4300770, China.
| | - Wei-Dong Jin
- Chinese People's Liberation Army Central Theater General Hospital General Surgery Department, Wuhan, Hubei, 4300770, China.
| |
Collapse
|
8
|
Ben Dhia S, Chauviere D, Mitrea D, Schiappa R, Pace Loscos T, Chamorey E, Baron D. Organ preservation, for rectal cancer: general overview of the latest data from phase III randomized trials. Acta Oncol 2025; 64:120-128. [PMID: 39871514 PMCID: PMC11794997 DOI: 10.2340/1651-226x.2025.41057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/09/2025] [Indexed: 01/29/2025]
Abstract
INTRODUCTION Organ preservation (OP) strategies are gaining interest in improving the quality of life in the management of rectal cancer, particularly for tumors located in the distal or middle rectum. The optimal OP protocol is still not standardized and relies on randomized trials. This review summarizes past and ongoing studies on OP protocols for adenocarcinoma of the distal and middle rectum. METHOD We searched for articles and abstracts on randomized clinical trials investigating OP approaches for rectal cancer, including data presented at the LUCARRE Congress held in Nice on November 25, 2023, covering ongoing and recently published trials on rectal preservation. RESULTS Our review's findings are presented in four tables: the first evaluates key trials with overall survival (OS) as the primary endpoint; the second provides an overview of past Phase III trials; the third reviews Phase II/III trials that specifically focus on local excisions (LE); and finally, the fourth summarizes ongoing trials. Each table is accompanied by detailed comments elucidating the significance and implications of the presented data, alongside a review of current guidelines. INTERPRETATION We highlight the growing interest in OP strategies for rectal cancer management to enhance patients' quality of life. Despite the lack of international consensus on the optimal OP protocol, past and ongoing randomized trials provide valuable findings into the evolving management strategies of rectal cancer treatment. The presented data supports the role of randomized phase III trials to provide evidence for a change in clinical practice.
Collapse
Affiliation(s)
- Syrine Ben Dhia
- Department of Radiotherapy, Antoine Lacassagne Center, Nice, France.
| | - Damien Chauviere
- Department of Clinical Research and Innovation, Antoine Lacassagne Center, Nice, France
| | - Diana Mitrea
- Department of Radiotherapy, Antoine Lacassagne Center, Nice, France
| | - Renaud Schiappa
- Department of Epidemiology, Biostatistics and Health Data, Centre Antoine Lacassagne, University of Côte d'Azur, Nice, France
| | - Tanguy Pace Loscos
- Department of Epidemiology, Biostatistics and Health Data, Centre Antoine Lacassagne, University of Côte d'Azur, Nice, France
| | - Emmanuel Chamorey
- Department of Epidemiology, Biostatistics and Health Data, Centre Antoine Lacassagne, University of Côte d'Azur, Nice, France
| | - David Baron
- Department of Radiotherapy, Antoine Lacassagne Center, Nice, France
| |
Collapse
|
9
|
Haddad TS, Bokhorst JM, Berger MD, Dobbelsteen LVD, Simmer F, Ciompi F, Galon J, Laak JVD, Pagès F, Zlobec I, Lugli A, Nagtegaal ID. Combining immunoscore and tumor budding in colon cancer: an insightful prognostication based on the tumor-host interface. J Transl Med 2024; 22:1090. [PMID: 39623479 PMCID: PMC11610196 DOI: 10.1186/s12967-024-05818-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Tumor Budding (TB) and Immunoscore are independent prognostic markers in colon cancer (CC). Given their respective representation of tumor aggressiveness and immune response, we examined their combination in association with patient disease-free survival (DFS) in pTNM stage I-III CC. METHODS In a series of pTNM stage I-III CCs (n = 654), the Immunoscore was computed and TB detected automatically using a deep learning network. Two-tiered systems for both biomarkers were used with cut-offs of 25% and ten buds for Immunoscore and TB according to clinical guidelines, respectively. Associations of Immunoscore with TB with 5-year DFS were examined using Kaplan-Meier survival analysis in addition to multivariable modeling and relative contribution analysis using Cox regression. RESULTS Immunoscore and TB independently are prognostic with hazard ratio (HR) = 2.0, 95% confidence interval (CI) 1.4-2.8 and HR 2.5, with 95% CI 1.4-4.5, respectively; P value < 0.0001. By combining Immunoscore with TB, patients with Immunoscore Low, TB High tumors had a significantly poorer DFS (HR 5.6, 95% CI 2.6-12.0; P value < 0.0001) than those with Immunoscore High, TB Low tumors. The combined Immunoscore with TB score was independently prognostic (P value = 0.009) in comparison to N-stage, T-stage, and MSI. Immunoscore with TB had the highest relative contribution (35%) to DFS in pTNM stage I-II CCs. CONCLUSIONS The association of Immunoscore and TB with patient survival suggests that both biomarkers are complementary and should be interpreted in combination to identify high-risk Stage I-II patients who should be considered for adjuvant therapy or further diagnostic testing.
Collapse
Affiliation(s)
- T S Haddad
- Radboud University Medical Center, Nijmegen, Netherlands
| | - J M Bokhorst
- Radboud University Medical Center, Nijmegen, Netherlands
| | - M D Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - F Simmer
- Radboud University Medical Center, Nijmegen, Netherlands
| | - F Ciompi
- Radboud University Medical Center, Nijmegen, Netherlands
| | - J Galon
- Centre de Recherche Des Cordeliers, Sorbonne Université, Université Paris Cité, 75006, Paris, France
| | - J V D Laak
- Radboud University Medical Center, Nijmegen, Netherlands
| | - F Pagès
- Centre de Recherche Des Cordeliers, Sorbonne Université, Université Paris Cité, 75006, Paris, France
| | - I Zlobec
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - A Lugli
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - I D Nagtegaal
- Radboud University Medical Center, Nijmegen, Netherlands.
- Department of Pathology, RadboudUMC, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
10
|
Montauti E, Oh DY, Fong L. CD4 + T cells in antitumor immunity. Trends Cancer 2024; 10:969-985. [PMID: 39242276 PMCID: PMC11464182 DOI: 10.1016/j.trecan.2024.07.009] [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: 06/04/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 09/09/2024]
Abstract
Advances in cancer immunotherapy have transformed cancer care and realized unprecedented responses in many patients. The growing arsenal of novel therapeutics - including immune checkpoint inhibition (ICI), adoptive T cell therapies (ACTs), and cancer vaccines - reflects the success of cancer immunotherapy. The therapeutic benefits of these treatment modalities are generally attributed to the enhanced quantity and quality of antitumor CD8+ T cell responses. Nevertheless, CD4+ T cells are now recognized to play key roles in both the priming and effector phases of the antitumor immune response. In addition to providing T cell help through co-stimulation and cytokine production, CD4+ T cells can also possess cytotoxicity either directly on MHC class II-expressing tumor cells or to other cells within the tumor microenvironment (TME). The presence of specific populations of CD4+ T cells, and their intrinsic plasticity, within the TME can represent an important determinant of clinical response to immune checkpoint inhibitors, vaccines, and chimeric antigen receptor (CAR) T cell therapies. Understanding how the antitumor functions of specific CD4+ T cell types are induced while limiting their protumorigenic attributes will enable more successful immunotherapies.
Collapse
Affiliation(s)
- Elena Montauti
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - David Y Oh
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA; Immunotherapy Integrated Research Center, Fred Hutchison Cancer Center, Seattle, WA, USA.
| |
Collapse
|
11
|
Li B, Chen L, Huang Y, Wu M, Fang W, Zou X, Zheng Y, Xiao Q. Are the tumor microenvironment characteristics of pretreatment biopsy specimens of colorectal cancer really effectively predict the efficacy of neoadjuvant therapy: A retrospective multicenter study. Medicine (Baltimore) 2024; 103:e39429. [PMID: 39213237 PMCID: PMC11365683 DOI: 10.1097/md.0000000000039429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/20/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
More and more studies had pointed out that the tumor microenvironment characteristics based on colorectal cancer (CRC) pretreatment biopsy specimens could effectively predict the efficacy of neoadjuvant therapy, but under hematoxylin and eosin (HE) staining, whether the tumor microenvironment characteristics observed by pathologists could predict the efficacy of neoadjuvant therapy remains to be discussed. We collected 106 CRC patients who received neoadjuvant treatment and surgical resection from 3 hospitals. The number of mitosis, inflammation degree, desmoplastic reaction (DR), necrosis, tumor-stroma ratio (TSR) and tumor budding (TB) of CRC pretreatment biopsy specimens were observed under HE staining, and the degree of tumor pathological remission of CRC surgical specimens after neoadjuvant treatment was evaluated. According to the tumor regression grade (TRG), patients were divided into good-responders (TRG 0-1) and non-responders (TRG 2-3). All data were analyzed with SPSS software (version 23.0) to evaluate the correlation between the number of mitosis, inflammation degree, DR, necrosis, TSR and TB in pretreatment biopsy samples and the treatment effect. In univariate analysis, mitosis (P = .442), inflammation degree (P = .951), DR (P = .186), necrosis (P = .306), TSR (P = .672), and TB (P = .327) were not associated with the response to neoadjuvant therapy. However, we found that for colon cancer, rectal cancer was more likely to benefit from neoadjuvant therapy (P = .024). In addition, we further analyzed the impact of mitosis, inflammation degree, DR, necrosis, TSR and TB on neoadjuvant therapy in rectal cancer, and found that there was no predictive effect. By analyzing the characteristics of tumor microenvironment of CRC pretreatment biopsy specimens under HE staining, such as mitosis, inflammation degree, DR, necrosis, TSR and TB, it was impossible to effectively predict the efficacy of neoadjuvant therapy for CRC.
Collapse
Affiliation(s)
- Bingbing Li
- Department of Pathology, Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Longjiao Chen
- Department of Pathology, Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Yichun Huang
- Department of Pathology, Ganzhou People’s Hospital, Ganzhou, China
| | - Meng Wu
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Weilan Fang
- Department of Pathology, Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Xin Zou
- Department of Pathology, Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Yihong Zheng
- Department of Pathology, Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Qiuxiang Xiao
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Graduate School, China Medical University, Shenyang, China
| |
Collapse
|
12
|
Beyaert S, Borys A, Baldin P, Dahou H, Magremanne M, Mahy P, Renwart W, Machiels JP, Schmitz S. Study of the PD-L1 expression, T-cells density and immunoscore in paired baseline tumor biopsies and surgical specimens in squamous cell carcinoma of the oral cavity. Oral Oncol 2024; 154:106869. [PMID: 38820890 DOI: 10.1016/j.oraloncology.2024.106869] [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: 03/21/2024] [Revised: 05/13/2024] [Accepted: 05/25/2024] [Indexed: 06/02/2024]
Abstract
OBJECTIVES Primary objective was to evaluate the correlation between immune marker expression in baseline tumor biopsies and their respective surgical specimens in squamous cell carcinoma of the oral cavity (OCSCC). Secondary objective was to assess the impact of these markers on overall (OS) and disease-free survival (DFS). MATERIALS AND METHODS Patients with a histological diagnosis of oral squamous cell carcinoma treated surgically between 2012 and 2020 were included in this retrospective, translational monocentric study. The expression of PD-L1, T-cells markers and an OCSCC-adapted immunoscore were evaluated by multiplex immunohistochemistry. RESULTS One hundred and four patients (mean: 58 years) were included. Seventy patients had paired samples available. Poor correlation was highlighted for PD-L1-positive surface expression (r = 0.29) and combined positive score (CPS). For CPS ≥ 20 and CPS ≥ 1, correlation coefficient r was 0.24 and 0.46 respectively. T-cells density showed also poor correlation with a r of 0.57 and 0.31 for CD3 and CD8 T-cells, respectively. Univariate survival analyses showed significant better OS and DFS (P < 0.05) for patients with stage III-IV OCSCC with a high compared to a low immunoscore, based on surgical samples only. CONCLUSION Our study showed poor correlation in PD-L1 expression, CPS, T-cells density and immunoscore between baseline tumor biopsies and surgical resection specimens. In addition, the immunoscore may emerge as a potential prognostic factor in advanced squamous cell carcinoma of the oral cavity. If surgical specimens are available, they may be of interest for clinical practice decision.
Collapse
Affiliation(s)
- Simon Beyaert
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Head & Neck Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Aléna Borys
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pamela Baldin
- Department of Pathology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Hajar Dahou
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Michèle Magremanne
- Department of Oral and Maxillofacial Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Pierre Mahy
- Department of Oral and Maxillofacial Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - William Renwart
- Department of Head & Neck Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Pascal Machiels
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Medical Oncology, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Sandra Schmitz
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Head & Neck Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
13
|
Domingo E, Kelly C, Hay J, Sansom O, Maka N, Oien K, Iveson T, Saunders M, Kerr R, Tomlinson I, Edwards J, Harkin A, Nowak M, Koelzer V, Easton A, Boukovinas I, Moustou E, Messaritakis I, Chondrozoumaki M, Karagianni M, Pagès F, Arnoux F, Lautard C, Lovera Y, Boquet I, Catteau A, Galon J, Souglakos I, Church DN, TransSCOT Consortium. Prognostic and Predictive Value of Immunoscore in Stage III Colorectal Cancer: Pooled Analysis of Cases From the SCOT and IDEA-HORG Studies. J Clin Oncol 2024; 42:2207-2218. [PMID: 38484206 PMCID: PMC11185918 DOI: 10.1200/jco.23.01648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 06/16/2024] Open
Abstract
PURPOSE Immunoscore (IS) is prognostic in stage III colorectal cancer (CRC) and may predict benefit of duration (6 v 3 months) of adjuvant infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX) chemotherapy. We sought to determine IS prognostic and predictive value in stage-III CRC treated with adjuvant FOLFOX or oral capecitabine and infusional oxaliplatin (CAPOX) in the SCOT and IDEA-HORG trials. METHODS Three thousand sixty-one cases had tumor samples, of which 2,643 (1,792 CAPOX) were eligible for IS testing. Predefined cutoffs (IS-Low and IS-High) were used to classify cases into two groups for analysis of disease-free survival (3-year DFS) and multivariable-adjusted hazard ratios (mvHRs) by Cox regression. RESULTS IS was determined in 2,608 (99.5%) eligible cases, with 877 (33.7%) samples classified as IS-Low. IS-Low tumors were more commonly high-risk (T4 and/or N2; 52.9% IS-Low v 42.2% IS-High; P < .001) and in younger patients (P = .024). Patients with IS-Low tumors had significantly shorter DFS in the CAPOX, FOLFOX, and combined cohorts (mvHR, 1.52 [95% CI, 1.28 to 1.82]; mvHR, 1.58 [95% CI, 1.22 to 2.04]; and mvHR, 1.55 [95% CI, 1.34 to 1.79], respectively; P < .001 all comparisons), regardless of sex, BMI, clinical risk group, tumor location, treatment duration, or chemotherapy regimen. IS prognostic value was greater in younger (≤65 years) than older (>65 years) patients in the CAPOX cohort (mvHR, 1.92 [95% CI, 1.50 to 2.46] v 1.28 [95% CI, 1.01 to 1.63], PINTERACTION = .026), and in DNA mismatch repair proficient than deficient mismatch repair disease (mvHR, 1.68 [95% CI, 1.41 to 2.00] v 0.67 [95% CI, 0.30 to 1.49], PINTERACTION = .03), although these exploratory analyses were uncorrected for multiple testing. Adding IS to a model containing all clinical variables significantly improved prediction of DFS (likelihood ratio test, P < .001) regardless of MMR status. CONCLUSION IS is prognostic in stage III CRC treated with FOLFOX or CAPOX, including within clinically relevant tumor subgroups. Possible variation in IS prognostic value by age and MMR status, and prediction of benefit from extended adjuvant therapy merit validation.
Collapse
Affiliation(s)
- Enric Domingo
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- CRUK Beatson Institute of Cancer Research, Garscube Estate, Glasgow, United Kingdom
| | - Caroline Kelly
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer Hay
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Owen Sansom
- CRUK Beatson Institute of Cancer Research, Garscube Estate, Glasgow, United Kingdom
| | - Noori Maka
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Karin Oien
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Tim Iveson
- University of Southampton, Southampton, United Kingdom
| | - Mark Saunders
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Rachel Kerr
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ian Tomlinson
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Joanne Edwards
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andrea Harkin
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, United Kingdom
| | - Marta Nowak
- Department of Pathology and Molecular Pathology, Zurich, Switzerland
| | - Viktor Koelzer
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- Department of Pathology and Molecular Pathology, Zurich, Switzerland
| | - Alistair Easton
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ioannis Boukovinas
- Medical Oncology Unit Department, Bioclinic Oncology Unit of Thessaloniki, Thessaloniki, Greece
| | - Eleni Moustou
- Pathology, University Hospital of Heraklion, Crete, Greece
| | - Ippokratis Messaritakis
- Laboratory of Translational Oncology, University of Crete—School of Medicine, Heraklion, Greece
| | - Maria Chondrozoumaki
- Laboratory of Tumor Cell Biology, University of Crete - School of Medicine, Heraklion, Greece
| | - Michaela Karagianni
- Laboratory of Translational Oncology, University of Crete—School of Medicine, Heraklion, Greece
| | - Franck Pagès
- INSERM, Laboratory of Integrative Cancer Immunology, Sorbonne Université, Université de Paris Cité, Cordeliers Research Center, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Immunomonitoring Platform, Georges Pompidou European Hospital, Paris, France
| | | | | | | | | | | | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Sorbonne Université, Université de Paris Cité, Cordeliers Research Center, Paris, France
- VERACYTE, Marseille, France
| | - Ioannis Souglakos
- Laboratory of Translational Oncology, University of Crete—School of Medicine, Heraklion, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, Greece
| | - David N. Church
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Comprehensive Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | | |
Collapse
|
14
|
Hijazi A, Galon J. Principles of risk assessment in colon cancer: immunity is key. Oncoimmunology 2024; 13:2347441. [PMID: 38694625 PMCID: PMC11062361 DOI: 10.1080/2162402x.2024.2347441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/16/2024] [Indexed: 05/04/2024] Open
Abstract
In clinical practice, the administration of adjuvant chemotherapy (ACT) following tumor surgical resection raises a critical dilemma for stage II colon cancer (CC) patients. The prognostic features used to identify high-risk CC patients rely on the pathological assessment of tumor cells. Currently, these factors are considered for stratifying patients who may benefit from ACT at early CC stages. However, the extent to which these factors predict clinical outcomes (i.e. recurrence, survival) remains highly controversial, also uncertainty persists regarding patients' response to treatment, necessitating further investigation. Therefore, an imperious need is to explore novel biomarkers that can reliably stratify patients at risk, to optimize adjuvant treatment decisions. Recently, we evaluated the prognostic and predictive value of Immunoscore (IS), an immune digital-pathology assay, in stage II CC patients. IS emerged as the sole significant parameter for predicting disease-free survival (DFS) in high-risk patients. Moreover, IS effectively stratified patients who would benefit most from ACT based on their risk of recurrence, thus predicting their outcomes. Notably, our findings revealed that digital IS outperformed the visual quantitative assessment of the immune response conducted by expert pathologists. The latest edition of the WHO classification for digestive tumor has introduced the evaluation of the immune response, as assessed by IS, as desirable and essential diagnostic criterion. This supports the revision of current cancer guidelines and strongly recommends the implementation of IS into clinical practice as a patient stratification tool, to guide CC treatment decisions. This approach may provide appropriate personalized therapeutic decisions that could critically impact early-stage CC patient care.
Collapse
Affiliation(s)
- Assia Hijazi
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
- Veracyte, Marseille, France
| |
Collapse
|
15
|
Hijazi A, Bifulco C, Baldin P, Galon J. Digital Pathology for Better Clinical Practice. Cancers (Basel) 2024; 16:1686. [PMID: 38730638 PMCID: PMC11083211 DOI: 10.3390/cancers16091686] [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: 04/08/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
(1) Background: Digital pathology (DP) is transforming the landscape of clinical practice, offering a revolutionary approach to traditional pathology analysis and diagnosis. (2) Methods: This innovative technology involves the digitization of traditional glass slides which enables pathologists to access, analyze, and share high-resolution whole-slide images (WSI) of tissue specimens in a digital format. By integrating cutting-edge imaging technology with advanced software, DP promises to enhance clinical practice in numerous ways. DP not only improves quality assurance and standardization but also allows remote collaboration among experts for a more accurate diagnosis. Artificial intelligence (AI) in pathology significantly improves cancer diagnosis, classification, and prognosis by automating various tasks. It also enhances the spatial analysis of tumor microenvironment (TME) and enables the discovery of new biomarkers, advancing their translation for therapeutic applications. (3) Results: The AI-driven immune assays, Immunoscore (IS) and Immunoscore-Immune Checkpoint (IS-IC), have emerged as powerful tools for improving cancer diagnosis, prognosis, and treatment selection by assessing the tumor immune contexture in cancer patients. Digital IS quantitative assessment performed on hematoxylin-eosin (H&E) and CD3+/CD8+ stained slides from colon cancer patients has proven to be more reproducible, concordant, and reliable than expert pathologists' evaluation of immune response. Outperforming traditional staging systems, IS demonstrated robust potential to enhance treatment efficiency in clinical practice, ultimately advancing cancer patient care. Certainly, addressing the challenges DP has encountered is essential to ensure its successful integration into clinical guidelines and its implementation into clinical use. (4) Conclusion: The ongoing progress in DP holds the potential to revolutionize pathology practices, emphasizing the need to incorporate powerful AI technologies, including IS, into clinical settings to enhance personalized cancer therapy.
Collapse
Affiliation(s)
- Assia Hijazi
- The French National Institute of Health & Medical Research (INSERM), Laboratory of Integrative Cancer Immunology, F-75006 Paris, France;
- Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
| | - Carlo Bifulco
- Providence Genomics, Portland, OR 02912, USA;
- Earle A Chiles Research Institute, Portland, OR 97213, USA
| | - Pamela Baldin
- Department of Pathology, Cliniques Universitaires Saint Luc, UCLouvain, 1200 Brussels, Belgium;
| | - Jérôme Galon
- The French National Institute of Health & Medical Research (INSERM), Laboratory of Integrative Cancer Immunology, F-75006 Paris, France;
- Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
- Veracyte, 13009 Marseille, France
| |
Collapse
|
16
|
Safont MJ, García-Figueiras R, Hernando-Requejo O, Jimenez-Rodriguez R, Lopez-Vicente J, Machado I, Ayuso JR, Bustamante-Balén M, De Torres-Olombrada MV, Domínguez Tristancho JL, Fernández-Aceñero MJ, Suarez J, Vera R. Interdisciplinary Spanish consensus on a watch-and-wait approach for rectal cancer. Clin Transl Oncol 2024; 26:825-835. [PMID: 37787973 DOI: 10.1007/s12094-023-03322-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/07/2023] [Indexed: 10/04/2023]
Abstract
Watch-and-wait has emerged as a new strategy for the management of rectal cancer when a complete clinical response is achieved after neoadjuvant therapy. In an attempt to standardize this new clinical approach, initiated by the Spanish Cooperative Group for the Treatment of Digestive Tumors (TTD), and with the participation of the Spanish Association of Coloproctology (AECP), the Spanish Society of Pathology (SEAP), the Spanish Society of Gastrointestinal Endoscopy (SEED), the Spanish Society of Radiation Oncology (SEOR), and the Spanish Society of Medical Radiology (SERAM), we present herein a consensus on a watch-and-wait approach for the management of rectal cancer. We have focused on patient selection, the treatment schemes evaluated, the optimal timing for evaluating the clinical complete response, the oncologic outcomes after the implementation of this strategy, and a protocol for surveillance of these patients.
Collapse
Affiliation(s)
- Maria Jose Safont
- Oncology Department, Consorcio Hospital General Universitario de Valencia. Valencia University, Av. de les Tres Creus, 2, 46014, València, Spain.
| | - Roberto García-Figueiras
- Radiology Department, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | - Jorge Lopez-Vicente
- Gastroenterology Department, Hospital Universitario de Mostoles, Mósteles, Spain
| | - Isidro Machado
- Instituto Valenciano de Oncología, Valencia, Spain
- Pathology Department, Patologika Laboratory QuironSalud, Valencia, Spain
- Pathology Department, University of Valencia, Valencia, Spain
| | | | - Marco Bustamante-Balén
- Gastrointestinal Endoscopy Unit, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | | | | | - Mª Jesús Fernández-Aceñero
- Surgical Pathology Department, Hospital Clínico San Carlos, IdiSSC, Universidad Complutense de Madrid, Madrid, Spain
| | - Javier Suarez
- General Surgery Department, Hospital Universitario de Navarra, Pamplona, Spain
| | - Ruth Vera
- Medical Oncology Department, Hospital Universitario de Navarra, Instituto de Investigación (Idisna), Pamplona, Spain
| |
Collapse
|
17
|
Rejeb SB, Elfekih S, Kouki N, Boulma R, Khouni H. Immunochemistry-based quantification of tumor-infiltrating lymphocytes and immunoscore as prognostic biomarkers in bladder cancer. J Egypt Natl Canc Inst 2024; 36:9. [PMID: 38523233 DOI: 10.1186/s43046-024-00212-8] [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: 10/30/2023] [Accepted: 03/06/2024] [Indexed: 03/26/2024] Open
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs) and the derived immunoscore (IS) have gained considerable attention over the last decade as prognostic markers in many solid cancers. However, in bladder cancer (BC), their prognostic value is not clearly established. METHODS The present study aimed to quantify the TILs rates in BC, assess the derived immunoscore, and investigate their prognostic value. An immunochemistry-based quantification of the different subtypes of TILS was performed on paraffin-embedded blocks from patients with invasive urothelial carcinoma of the bladder. We have assessed the rates of TILs, respectively, on peri-tumoral (PT) and intra-tumoral (IT) areas and calculated for each case the corresponding IS which is the index: CD8+/CD3+ TILs. The IS was then classified as low (I0, I1) or high (I2, I3, I4). We included 30 cases in the analysis. RESULTS The median age of patients was 65 years with a sex ratio of 9. TILs densities and distribution were significantly variable between IT and PT areas CD3+ (p = 0.03) and CD8+ (p = 0.004) with the highest rates on the PT areas. In univariate analysis, a low density of CD8+ TILs was significantly associated with an advanced age (p = 0.05), with the presence of lympho-vascular invasion (p = 0.02) and with the absence of specific histological subtype (p = 0.05). A low immunoscore was significantly associated with the presence of lympho-vascular invasion (p = 0.004). No significant association was found between TILs subpopulations, the IS, and the other clinicopathological and survival data. The overall survival (OS) and disease-free survival (DFS) medians were slightly superior in highly T (CD3+/CD8+)-cell infiltrated tumors as well as tumors with a high IS densities. However, the univariate analysis showed that TILs and immunoscore did not impact overall survival (OS) and disease-free survival (DFS). CONCLUSION TILs and immunoscore might be effective prognostic tools in BC. However, standardized quantification methods and further investigation on larger samples are highly recommended to definitively attest the prognostic value of TILs and IS in BC.
Collapse
Affiliation(s)
- Sarra Ben Rejeb
- Department of Pathology, Hopital des forces de sécurité intérieure de la Marsa, Tunis, Tunisia
- Faculty of Medecine of Tunis, Tunis Manar University, Tunis, Tunisia
| | - Sirine Elfekih
- Department of Pathology, Hopital des forces de sécurité intérieure de la Marsa, Tunis, Tunisia.
- Faculty of Medecine of Tunis, Tunis Manar University, Tunis, Tunisia.
| | - Nadia Kouki
- Department of Pathology, Hopital des forces de sécurité intérieure de la Marsa, Tunis, Tunisia
- Faculty of Medecine of Tunis, Tunis Manar University, Tunis, Tunisia
| | - Rami Boulma
- Department of Urology, Hopital des forces de sécurité intérieure de la Marsa, Tunis, Tunisia
- Faculty of Medecine of Tunis, Tunis Manar University, Tunis, Tunisia
| | - Hassen Khouni
- Department of Urology, Hopital des forces de sécurité intérieure de la Marsa, Tunis, Tunisia
- Faculty of Medecine of Tunis, Tunis Manar University, Tunis, Tunisia
| |
Collapse
|
18
|
Kuznetsova O, Fedyanin M, Zavalishina L, Moskvina L, Kuznetsova O, Lebedeva A, Tryakin A, Kireeva G, Borshchev G, Tjulandin S, Ignatova E. Prognostic and predictive role of immune microenvironment in colorectal cancer. World J Gastrointest Oncol 2024; 16:643-652. [PMID: 38577454 PMCID: PMC10989368 DOI: 10.4251/wjgo.v16.i3.643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/20/2023] [Accepted: 01/22/2024] [Indexed: 03/12/2024] Open
Abstract
Colorectal cancer (CRC) represents a molecularly heterogeneous disease and one of the most frequent causes of cancer-related death worldwide. The traditional classification of CRC is based on pathomorphological and molecular characteristics of tumor cells (mucinous, ring-cell carcinomas, etc.), analysis of mechanisms of carcinogenesis involved (chromosomal instability, microsatellite instability, CpG island methylator phenotype) and mutational statuses of commonly altered genes (KRAS, NRAS, BRAF, APC, etc.), as well as expression signatures (CMS 1-4). It is also suggested that the tumor microenvironment is a key player in tumor progression and metastasis in CRC. According to the latest data, the immune microenvironment can also be predictive of the response to immune checkpoint inhibitors. In this review, we highlight how the immune environment influences CRC prognosis and sensitivity to systemic therapy.
Collapse
Affiliation(s)
- Olesya Kuznetsova
- Department of Chemotherapy, Federal State Budgetary Institution (N.N. Blokhin National Medical Research Center of Oncology) of the Ministry of Health of the Russian Federation, Moscow 115478, Russia
| | - Mikhail Fedyanin
- Department of Chemotherapy, Federal State Budgetary Institution (N.N. Blokhin National Medical Research Center of Oncology) of the Ministry of Health of the Russian Federation, Moscow 115478, Russia
| | - Larisa Zavalishina
- Department of Pathology, Russian Medical Academy of Continuous Professional Education, Moscow 123242, Russia
| | - Larisa Moskvina
- Department of Pathology, Russian Medical Academy of Continuous Professional Education, Moscow 123242, Russia
| | - Olga Kuznetsova
- Department of Pathology, Russian Medical Academy of Continuous Professional Education, Moscow 123242, Russia
| | | | - Alexey Tryakin
- Department of Chemotherapy, Federal State Budgetary Institution (N.N. Blokhin National Medical Research Center of Oncology) of the Ministry of Health of the Russian Federation, Moscow 115478, Russia
| | - Galina Kireeva
- Federal State Budgetary Institution “National Medical and Surgical Center named after N.I. Pirogov” of the Ministry of Health of the Russian Federation, Moscow 105203, Russia
| | - Gleb Borshchev
- Federal State Budgetary Institution “National Medical and Surgical Center named after N.I. Pirogov” of the Ministry of Health of the Russian Federation, Moscow 105203, Russia
| | - Sergei Tjulandin
- Department of Chemotherapy, Federal State Budgetary Institution (N.N. Blokhin National Medical Research Center of Oncology) of the Ministry of Health of the Russian Federation, Moscow 115478, Russia
| | | |
Collapse
|
19
|
Jepsen DNM, Høeg H, Bzorek M, Orhan A, Eriksen JO, Gögenur I, Reiss B, Fiehn AMK. Digitally assessed lymphocyte infiltration in rectal cancer biopsies is associated with pathological response to neoadjuvant therapy. Hum Pathol 2024; 144:61-70. [PMID: 38157991 DOI: 10.1016/j.humpath.2023.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
A frequently used treatment strategy in locally advanced rectal cancer (RC) is neoadjuvant therapy followed by surgery. Patients treated with neoadjuvant therapy achieve varying pathological response, and currently, predicting the degree of response is challenging. This study examined the association between digitally assessed histopathological features in the diagnostic biopsies and pathological response to neoadjuvant therapy, aiming to find potential predictive biomarkers. 50 patients with RC treated with neoadjuvant chemotherapy and/or radiotherapy followed by surgery were included. Deep learning-based digital algorithms were used to assess the epithelium tumor area percentage (ETP) based on H&E-stained slides, and to quantify the density of CD3+ and CD8+ lymphocytes, as well as the CD8+/CD3+ lymphocyte percentage, based on immunohistochemically stained slides, from the diagnostic tumor biopsies. Pathological response was assessed according to the Mandard method. A good pathological response was defined as tumor regression grade (TRG) 1-2, and a complete pathological response was defined as Mandard TRG 1. Associations between the ETP and lymphocyte densities in the diagnostic biopsies and the pathological response were examined. The density of CD8+ lymphocytes, and the CD8+/CD3+ lymphocyte percentage, were associated with both good and complete response to neoadjuvant therapy, while the density of CD3+ lymphocytes was associated with complete response. The ETP did not correlate with response to neoadjuvant therapy. It is well-known that infiltration of lymphocytes in colorectal cancer is a prognostic biomarker. However, assessment of CD8+ and CD3+ lymphocytes in the diagnostic tumor biopsies of patients with RC may also be useful in predicting response to neoadjuvant therapy.
Collapse
Affiliation(s)
- Dea Natalie Munch Jepsen
- Department of Pathology, Zealand University Hospital, Denmark; Center for Surgical Science, Department of Surgery, Zealand University Hospital, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark.
| | | | - Michael Bzorek
- Department of Pathology, Zealand University Hospital, Denmark.
| | - Adile Orhan
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Denmark; Department of Clinical Oncology, Zealand University Hospital, Denmark.
| | | | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark.
| | | | - Anne-Marie Kanstrup Fiehn
- Department of Pathology, Zealand University Hospital, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark.
| |
Collapse
|
20
|
El Sissy C, Kirilovsky A, Lagorce Pagès C, Marliot F, Custers PA, Dizdarevic E, Sroussi M, Castillo-Martin M, Haicheur N, Dermani M, Loche N, Buttard B, Musina AM, Anitei MG, van den Berg JG, Broeks A, Iseas S, Coraglio M, Loria FS, Romero A, Laurent-Puig P, de Reyniès A, Fernandez LM, Karoui M, Tougeron D, Vaccaro CA, Santino JP, Poulsen LØ, Lindebjerg J, O'Connor JM, Scripcariu V, Dimofte MG, Gérard JP, Chalabi M, Figueiredo N, Perez RO, Habr-Gama A, Galon J, Hansen TF, Jensen LH, Beets G, Zeitoun G, Pagès F. International Validation of the Immunoscore Biopsy in Patients With Rectal Cancer Managed by a Watch-and-Wait Strategy. J Clin Oncol 2024; 42:70-80. [PMID: 37788410 PMCID: PMC10730081 DOI: 10.1200/jco.23.00586] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/06/2023] [Accepted: 07/24/2023] [Indexed: 10/05/2023] Open
Abstract
PURPOSE No biomarker capable of improving selection and monitoring of patients with rectal cancer managed by watch-and-wait (W&W) strategy is currently available. Prognostic performance of the Immunoscore biopsy (ISB) was recently suggested in a preliminary study. METHODS This international validation study included 249 patients with clinical complete response (cCR) managed by W&W strategy. Intratumoral CD3+ and CD8+ T cells were quantified on pretreatment rectal biopsies by digital pathology and converted to ISB. The primary end point was time to recurrence (TTR; the time from the end of neoadjuvant treatment to the date of local regrowth or distant metastasis). Associations between ISB and outcomes were analyzed by stratified Cox regression adjusted for confounders. Immune status of tumor-draining lymph nodes (n = 161) of 17 additional patients treated by neoadjuvant chemoradiotherapy and surgery was investigated by 3'RNA-Seq and immunofluorescence. RESULTS Recurrence-free rates at 5 years were 91.3% (82.4%-100.0%), 62.5% (53.2%-73.3%), and 53.1% (42.4%-66.5%) with ISB High, ISB Intermediate, and ISB Low, respectively (hazard ratio [HR; Low v High], 6.51; 95% CI, 1.99 to 21.28; log-rank P = .0004). ISB was also significantly associated with disease-free survival (log-rank P = .0002), and predicted both local regrowth and distant metastasis. In multivariate analysis, ISB was independent of patient age, sex, tumor location, cT stage (T, primary tumor; c, clinical), cN stage (N, regional lymph node; c, clinical), and was the strongest predictor for TTR (HR [ISB High v Low], 6.93; 95% CI, 2.08 to 23.15; P = .0017). The addition of ISB to a clinical-based model significantly improved the prediction of recurrence. Finally, B-cell proliferation and memory in draining lymph nodes was evidenced in the draining lymph nodes of patients with cCR. CONCLUSION The ISB is validated as a biomarker to predict both local regrowth and distant metastasis, with a gradual scaling of the risk of pejorative outcome.
Collapse
Affiliation(s)
- Carine El Sissy
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| | - Amos Kirilovsky
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| | - Christine Lagorce Pagès
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
- Department of Pathology, AP-HP, Georges Pompidou European Hospital, Paris, France
| | - Florence Marliot
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| | - Petra A. Custers
- Department of Surgery, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Edina Dizdarevic
- Department of Oncology, Danish Colorectal Cancer Center South, University Hospital of Southern Denmark, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Marine Sroussi
- Cordeliers Research Center, Sorbonne University, INSERM, University Paris Cité, SIRIC CARPEM, Paris, France
- Chemistry Biology Innovation Institute, BioChimie Laboratory, ESPCI, UMR8231 CNRS, University PSL, Paris, France
| | | | - Nacilla Haicheur
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| | - Mohamed Dermani
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| | - Nicolas Loche
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| | - Bénedicte Buttard
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
| | - Ana Maria Musina
- Department of Surgical Oncology, Regional Institute of Oncology (IRO), Iasi, Romania
- “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Maria Gabriela Anitei
- “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- Surgical Oncology Unit, Regional Institute of Oncology, Iasi, Romania
| | - José G. van den Berg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Soledad Iseas
- Oncology Unit, Gastroenterology Hospital Carlos Bonorino Udaondo, Buenos Aires, Argentina
| | - Mariana Coraglio
- Oncology Unit, Gastroenterology Hospital Carlos Bonorino Udaondo, Buenos Aires, Argentina
| | - Fernando Sanchez Loria
- GI Clinical Oncology and GI Surgical Oncology, Instituto Alexander Fleming, Buenos Aires, Argentina
| | - Alfredo Romero
- Department of Clinical Oncology, British Hospital of Buenos Aires, Buenos Aires, Argentina
| | - Pierre Laurent-Puig
- Cordeliers Research Center, Sorbonne University, University Paris Cité, INSERM UMR1138, Paris, France
- Department of Biology, Cancer Institute Paris CARPEM, AP-HP, Georges Pompidou European Hospital, Paris, France
| | - Aurélien de Reyniès
- Cordeliers Research Center, University Paris Cité, INSERM UMRS1138, Paris, France
- AP-HP, SeqOIA Genomic Medicine Laboratory—IT Platform, Paris, France
| | - Laura M. Fernandez
- Colorectal Surgery, Digestive Department, Champalimaud Foundation, Lisbon, Portugal
| | - Mehdi Karoui
- Department of Digestive and Oncologic Surgery, AP-HP, Georges Pompidou European Hospital, Paris, France
| | - David Tougeron
- Department of Hepato-Gastro-Enterology and Nutritional Assistance, University Hospital of Poitiers, Poitiers, France
| | - Carlos A. Vaccaro
- Department of General Surgery, Hospital Italiano, Buenos Aires, Argentina
| | - Juan P. Santino
- Department of General Surgery, Hospital Italiano, Buenos Aires, Argentina
| | - Laurids Østergaard Poulsen
- Department of Oncology, Danish Colorectal Cancer Center South, University Hospital of Southern Denmark, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Jan Lindebjerg
- Department of Oncology, Danish Colorectal Cancer Center South, University Hospital of Southern Denmark, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Juan Manuel O'Connor
- GI Clinical Oncology and GI Surgical Oncology, Instituto Alexander Fleming, Buenos Aires, Argentina
| | - Viorel Scripcariu
- “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
- Surgical Oncology Unit, Regional Institute of Oncology, Iasi, Romania
| | - Mihail-Gabriel Dimofte
- Department of Surgical Oncology, Regional Institute of Oncology (IRO), Iasi, Romania
- “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | | | - Myriam Chalabi
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nuno Figueiredo
- Colorectal Surgery, Digestive Department, Champalimaud Foundation, Lisbon, Portugal
| | - Rodrigo O. Perez
- Division of Colorectal Surgery, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
- Department of Surgical Oncology, Beneficencia Portuguesa Hospital, São Paulo, Brazil
- Angelita and Joaquim Gama Institute, São Paulo, Brazil
| | - Angelita Habr-Gama
- Division of Colorectal Surgery, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
- Department of Surgical Oncology, Beneficencia Portuguesa Hospital, São Paulo, Brazil
- Angelita and Joaquim Gama Institute, São Paulo, Brazil
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
| | - Torben Frøstrup Hansen
- Department of Oncology, Danish Colorectal Cancer Center South, University Hospital of Southern Denmark, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Lars Henrik Jensen
- Department of Oncology, Danish Colorectal Cancer Center South, University Hospital of Southern Denmark, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Geerard Beets
- Department of Surgery, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Guy Zeitoun
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| | - Franck Pagès
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Cordeliers Research Center, Sorbonne University, University Paris Cité, Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, Assistance Publique-Hôpitaux de Paris (AP-HP), Georges Pompidou European Hospital, Paris, France
| |
Collapse
|
21
|
Pennings AJ, van der Velden BA, Kloft M, Kooreman LFS, Kleijnen JMP, Breukink SO, Beets GL, Grabsch HI, Melenhorst J. The Role of Nonmetastatic Lymph Nodes in the Survival of Colorectal Cancer: A Systematic Review. ANNALS OF SURGERY OPEN 2023; 4:e336. [PMID: 38144501 PMCID: PMC10735087 DOI: 10.1097/as9.0000000000000336] [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: 05/02/2023] [Accepted: 08/11/2023] [Indexed: 12/26/2023] Open
Abstract
Objective In this review, we aim to provide an overview of literature on lymph node (LN) histomorphological features and their relationship with the prognosis in colorectal cancer (CRC). Background Lymph nodes play a crucial role in the treatment and prognosis of CRC. The presence of LN metastases considerably worsens the prognosis in CRC patients. Literature has shown that the total number of LNs and the number negative LNs (LNnegs) has prognostic value in CRC patients. In esophageal carcinoma, LN size seems to be surrogate of the host antitumor response and a potentially clinically useful new prognostic biomarker for (y)pN0 esophageal carcinoma. Methods A comprehensive search was performed in Pubmed, Embase, Medline, CINAHL, and the Cochrane library in March 2021. The PRISMA guidelines were followed. Only studies focusing on histomorphological features and LN size and their relation to overall survival were selected. Results A total of 9 unique articles met all inclusion criteria and were therefore included in this systematic review. Six of these studies investigated HMF (eg, paracortical hyperplasia, germinal center predominance, and sinus histiocytosis) and 4 studies LNneg size and their relationship with overall survival. The presence of paracortical hyperplasia and an increased number of large LNnegs is related to a more favorable prognosis in CRC. Conclusion The results of this systematic review seem to support the hypothesis that there is a relationship between the host antitumor response reflected in different histomorphological reaction patterns visible in LNnegs and LNneg size related to survival in CRC patients.
Collapse
Affiliation(s)
- Alexander J. Pennings
- From the Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Brecht A. van der Velden
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Maximilian Kloft
- Department of Pathology, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Internal Medicine IV, Justus-Liebig-University, University Hospital Giessen and Marburg, Giessen, Germany
| | - Loes F. S. Kooreman
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of Pathology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jos M. P. Kleijnen
- Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Stephanie O. Breukink
- From the Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Geerard L. Beets
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Heike I. Grabsch
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of Pathology, Maastricht University Medical Center, Maastricht, the Netherlands
- Pathology & Data Analytics, Leeds Institute of Research at St James’s, University of Leeds, Leeds, United Kingdom
| | - Jarno Melenhorst
- From the Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
22
|
Schulz S, Jesinghaus M, Foersch S. [Multistain deep learning as a prognostic and predictive biomarker in colorectal cancer]. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:104-108. [PMID: 37987821 DOI: 10.1007/s00292-023-01280-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 11/22/2023]
Abstract
The tumor immune microenvironment (TIME) plays a crucial prognostic and predictive role in solid malignancies such as colorectal cancer (CRC). Nevertheless, scoring systems based on TIME such as the Immunoscore (IS) are rarely used in clinical practice. Among other reasons, this might be due to the additional time required for manual quantification of tumor-associated immune cells or costs associated with proprietary/commercial solutions. To address these issues, we developed a multistain deep learning model (MSDLM) and trained, validated, and tested it on immunohistochemical image data of different immune cell subtypes from over 1000 patients with CRC. Our model showed high prognostic accuracy and outperformed other clinical, molecular, and immune cell-based parameters. It might also be used for therapy response prediction in rectal cancer patients undergoing neoadjuvant therapy. Leveraging artificial intelligence interpretability/explainability methods, we ascertained that the MSDLM's predictions align with recognized antitumor immune response patterns. Consequently, the AImmunoscore (AIS) could emerge as a potential TIME-based decision-making tool for clinicians.
Collapse
Affiliation(s)
- Stefan Schulz
- Institut für Pathologie, Universitätsmedizin Mainz, Langenbeckstr. 1, Mainz, Deutschland
| | - Moritz Jesinghaus
- Institut für Pathologie, Universitätsklinikum Marburg, Marburg, Deutschland
| | - Sebastian Foersch
- Institut für Pathologie, Universitätsmedizin Mainz, Langenbeckstr. 1, Mainz, Deutschland.
| |
Collapse
|
23
|
Sano S, Akiyoshi T, Yamamoto N, Hiyoshi Y, Mukai T, Yamaguchi T, Nagasaki T, Taketomi A, Fukunaga Y, Kawachi H. Intratumoral Budding and CD8-Positive T-cell Density in Pretreatment Biopsies as a Predictor of Response to Neoadjuvant Chemoradiotherapy in Advanced Rectal Cancer. Clin Colorectal Cancer 2023; 22:411-420.e1. [PMID: 37516615 DOI: 10.1016/j.clcc.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/31/2023]
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (CRT) is the standard treatment for advanced rectal cancer. Yet, the response to CRT varies from complete response to zero tumor regression. MATERIALS AND METHODS The impact of intratumoral budding (ITB) and intratumoral CD8+ cell density on response to CRT and survival were evaluated in biopsy samples from 266 patients with advanced rectal cancer who were treated with long-course neoadjuvant CRT. The expression of epithelial-mesenchymal transition (EMT) markers was compared between patients with high and low ITB, using data from 174 patients with RNA sequencing. RESULTS High ITB was observed in 62 patients (23.3%). There was no association between ITB and CD8+ cell density. The multivariable logistic regression analysis showed that high CD8+ cell density (OR, 2.69; 95% CI, 1.45-4.98; P = .002) was associated with good response to CRT, whereas high ITB (OR, 0.33; 95% CI, 0.14-0.80; P = .014) was associated with poor response. Multivariable Cox regression analysis for survival showed that high CD8+ cell density was associated with better recurrence-free survival (HR, 0.41; 95% CI, 0.24-0.72; P = .002) and overall survival (HR, 0.36; 95% CI, 0.17-0.74; P = .005), but significance values for ITB were marginal (P = .104 for recurrence-free survival and P = .163 for overall survival). The expression of EMT-related genes was not significantly different between patients with high and low ITB. CONCLUSION ITB and CD8+ cell density in biopsy samples may serve as useful biomarkers to predict therapy response in patients with rectal cancer treated with neoadjuvant CRT.
Collapse
Affiliation(s)
- Shuhei Sano
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Akiyoshi
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Noriko Yamamoto
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yukiharu Hiyoshi
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiki Mukai
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomohiro Yamaguchi
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiya Nagasaki
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yosuke Fukunaga
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroshi Kawachi
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
24
|
Schurink NW, van Kranen SR, van Griethuysen JJM, Roberti S, Snaebjornsson P, Bakers FCH, de Bie SH, Bosma GPT, Cappendijk VC, Geenen RWF, Neijenhuis PA, Peterson GM, Veeken CJ, Vliegen RFA, Peters FP, Bogveradze N, El Khababi N, Lahaye MJ, Maas M, Beets GL, Beets-Tan RGH, Lambregts DMJ. Development and multicenter validation of a multiparametric imaging model to predict treatment response in rectal cancer. Eur Radiol 2023; 33:8889-8898. [PMID: 37452176 PMCID: PMC10667134 DOI: 10.1007/s00330-023-09920-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
OBJECTIVES To develop and validate a multiparametric model to predict neoadjuvant treatment response in rectal cancer at baseline using a heterogeneous multicenter MRI dataset. METHODS Baseline staging MRIs (T2W (T2-weighted)-MRI, diffusion-weighted imaging (DWI) / apparent diffusion coefficient (ADC)) of 509 patients (9 centres) treated with neoadjuvant chemoradiotherapy (CRT) were collected. Response was defined as (1) complete versus incomplete response, or (2) good (Mandard tumor regression grade (TRG) 1-2) versus poor response (TRG3-5). Prediction models were developed using combinations of the following variable groups: (1) Non-imaging: age/sex/tumor-location/tumor-morphology/CRT-surgery interval (2) Basic staging: cT-stage/cN-stage/mesorectal fascia involvement, derived from (2a) original staging reports, or (2b) expert re-evaluation (3) Advanced staging: variables from 2b combined with cTN-substaging/invasion depth/extramural vascular invasion/tumor length (4) Quantitative imaging: tumour volume + first-order histogram features (from T2W-MRI and DWI/ADC) Models were developed with data from 6 centers (n = 412) using logistic regression with the Least Absolute Shrinkage and Selector Operator (LASSO) feature selection, internally validated using repeated (n = 100) random hold-out validation, and externally validated using data from 3 centers (n = 97). RESULTS After external validation, the best model (including non-imaging and advanced staging variables) achieved an area under the curve of 0.60 (95%CI=0.48-0.72) to predict complete response and 0.65 (95%CI=0.53-0.76) to predict a good response. Quantitative variables did not improve model performance. Basic staging variables consistently achieved lower performance compared to advanced staging variables. CONCLUSIONS Overall model performance was moderate. Best results were obtained using advanced staging variables, highlighting the importance of good-quality staging according to current guidelines. Quantitative imaging features had no added value (in this heterogeneous dataset). CLINICAL RELEVANCE STATEMENT Predicting tumour response at baseline could aid in tailoring neoadjuvant therapies for rectal cancer. This study shows that image-based prediction models are promising, though are negatively affected by variations in staging quality and MRI acquisition, urging the need for harmonization. KEY POINTS This multicenter study combining clinical information and features derived from MRI rendered disappointing performance to predict response to neoadjuvant treatment in rectal cancer. Best results were obtained with the combination of clinical baseline information and state-of-the-art image-based staging variables, highlighting the importance of good quality staging according to current guidelines and staging templates. No added value was found for quantitative imaging features in this multicenter retrospective study. This is likely related to acquisition variations, which is a major problem for feature reproducibility and thus model generalizability.
Collapse
Affiliation(s)
- Niels W Schurink
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology & Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| | - Simon R van Kranen
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joost J M van Griethuysen
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology & Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| | - Sander Roberti
- Department of Epidemiology and Biostatistics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petur Snaebjornsson
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frans C H Bakers
- Department of Radiology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Shira H de Bie
- Department of Radiology, Deventer Ziekenhuis, Schalkhaar, The Netherlands
| | - Gerlof P T Bosma
- Department of Interventional Radiology, Elisabeth Tweesteden Hospital, Tilburg, The Netherlands
| | - Vincent C Cappendijk
- Department of Radiology, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | - Remy W F Geenen
- Department of Radiology, Northwest Clinics, Alkmaar, The Netherlands
| | | | | | - Cornelis J Veeken
- Department of Radiology, IJsselland Hospital, Capelle aan den IJssel, The Netherlands
| | - Roy F A Vliegen
- Department of Radiology, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Femke P Peters
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nino Bogveradze
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology & Developmental Biology, University of Maastricht, Maastricht, The Netherlands
- Department of Radiology, Acad. F. Todua Medical Center, Research Institute of Clinical Medicine, Tbilisi, Georgia
| | - Najim El Khababi
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology & Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| | - Max J Lahaye
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology & Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| | - Monique Maas
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology & Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| | - Geerard L Beets
- GROW School for Oncology & Developmental Biology, University of Maastricht, Maastricht, The Netherlands
- Department of Surgery, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Regina G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology & Developmental Biology, University of Maastricht, Maastricht, The Netherlands
- Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Doenja M J Lambregts
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- GROW School for Oncology & Developmental Biology, University of Maastricht, Maastricht, The Netherlands.
| |
Collapse
|
25
|
Amintas S, Giraud N, Fernandez B, Dupin C, Denost Q, Garant A, Frulio N, Smith D, Rullier A, Rullier E, Vuong T, Dabernat S, Vendrely V. The Crying Need for a Better Response Assessment in Rectal Cancer. Curr Treat Options Oncol 2023; 24:1507-1523. [PMID: 37702885 PMCID: PMC10643426 DOI: 10.1007/s11864-023-01125-9] [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] [Accepted: 07/09/2023] [Indexed: 09/14/2023]
Abstract
OPINION STATEMENT Since total neoadjuvant treatment achieves almost 30% pathologic complete response, organ preservation has been increasingly debated for good responders after neoadjuvant treatment for patients diagnosed with rectal cancer. Two organ preservation strategies are available: a watch and wait strategy and a local excision strategy including patients with a near clinical complete response. A major issue is the selection of patients according to the initial tumor staging or the response assessment. Despite modern imaging improvement, identifying complete response remains challenging. A better selection could be possible by radiomics analyses, exploiting numerous image features to feed data characterization algorithms. The subsequent step is to include baseline and/or pre-therapeutic MRI, PET-CT, and CT radiomics added to the patients' clinicopathological data, inside machine learning (ML) prediction models, with predictive or prognostic purposes. These models could be further improved by the addition of new biomarkers such as circulating tumor biomarkers, molecular profiling, or pathological immune biomarkers.
Collapse
Affiliation(s)
- Samuel Amintas
- Tumor Biology and Tumor Bank Laboratory, CHU Bordeaux, F-33600, Pessac, France.
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, F-33000, Bordeaux, France.
| | - Nicolas Giraud
- Department of Radiation Oncology, CHU Bordeaux, F-33000, Bordeaux, France
| | | | - Charles Dupin
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, F-33000, Bordeaux, France
- Department of Radiation Oncology, CHU Bordeaux, F-33000, Bordeaux, France
| | - Quentin Denost
- Bordeaux Colorectal Institute, F-33000, Bordeaux, France
| | - Aurelie Garant
- UT Southwestern Department of Radiation Oncology, Dallas, USA
| | - Nora Frulio
- Radiology Department, CHU Bordeaux, F-33600, Pessac, France
| | - Denis Smith
- Department of Digestive Oncology, CHU Bordeaux, F-33600, Pessac, France
| | - Anne Rullier
- Histology Department, CHU Bordeaux, F-33000, Bordeaux, France
| | - Eric Rullier
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, F-33000, Bordeaux, France
- Surgery Department, CHU Bordeaux, F-33600, Pessac, France
| | - Te Vuong
- Department of Radiation Oncology, McGill University, Jewish General Hospital, Montreal, Canada
| | - Sandrine Dabernat
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, F-33000, Bordeaux, France
- Biochemistry Department, CHU Bordeaux, F-33000, Bordeaux, France
| | - Véronique Vendrely
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, F-33000, Bordeaux, France
- Department of Radiation Oncology, CHU Bordeaux, F-33000, Bordeaux, France
| |
Collapse
|
26
|
Zhang X, Zeng Y, Li H, Zhuang Q, Tang L, Wu J, Li J. A Modified NAR Scoring Model Incorporating Immune Infiltration Characteristics to Better Predict Long-Term Survival Following Neoadjuvant Radiotherapy in Rectal Cancer. Life (Basel) 2023; 13:2106. [PMID: 38004246 PMCID: PMC10672442 DOI: 10.3390/life13112106] [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: 09/17/2023] [Revised: 10/15/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
(1) Background: The neoadjuvant rectal (NAR) score has been developed as a prognostic tool for survival in locally advanced rectal cancer (LARC). However, the NAR score only incorporates weighted cT, ypT, and ypN categories. This long-term follow-up study aims to modify a novel prognostic scoring model and identify a short-term endpoint for survival. (2) Methods: The prognostic factors for overall survival (OS) were explored through univariate and multivariate analyses. Based on Cox regression modeling, nomogram plots were constructed. Area under the curve (AUC) and concordance indices were used to evaluate the performance of the nomogram. Receiver operating characteristic (ROC) analysis was conducted to compare the efficiency of the nomogram with other prognostic factors. (3) Results: After a long-term follow-up, the 5-year OS was 67.1%. The mean NAR score was 20.4 ± 16.3. Multivariate analysis indicated that CD8+ T-cell, lymphovascular invasion, and the NAR score were independent predictors of OS. The modified NAR scoring model, incorporating immune infiltration characteristics, exhibited a high C-index of 0.739 for 5-year OS, significantly outperforming any individual factor. Moreover, the predictive value of the nomogram was superior to the AJCC stage and pathological complete regression at 3-year, 5-year, and 10-year time points, respectively. Over time, the model's predictions of long-term survival remained consistent and improved in accuracy. (4) Conclusions: The modified NAR scoring model, incorporating immune infiltration characteristics, demonstrates high accuracy and consistency in predicting OS.
Collapse
Affiliation(s)
| | | | | | | | | | - Junxin Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China; (X.Z.); (Y.Z.); (H.L.); (Q.Z.); (L.T.)
| | - Jinluan Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China; (X.Z.); (Y.Z.); (H.L.); (Q.Z.); (L.T.)
| |
Collapse
|
27
|
Jiang W, Yu X, Dong X, Long C, Chen D, Cheng J, Yan B, Xu S, Lin Z, Chen G, Zhuo S, Yan J. A nomogram based on collagen signature for predicting the immunoscore in colorectal cancer. Front Immunol 2023; 14:1269700. [PMID: 37781377 PMCID: PMC10538535 DOI: 10.3389/fimmu.2023.1269700] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Objectives The Immunoscore can categorize patients into high- and low-risk groups for prognostication in colorectal cancer (CRC). Collagen plays an important role in immunomodulatory functions in the tumor microenvironment (TME). However, the correlation between collagen and the Immunoscore in the TME is unclear. This study aimed to construct a collagen signature to illuminate the relationship between collagen structure and Immunoscore. Methods A total of 327 consecutive patients with stage I-III stage CRC were included in a training cohort. The fully quantitative collagen features were extracted at the tumor center and invasive margin of the specimens using multiphoton imaging. LASSO regression was applied to construct the collagen signature. The association of the collagen signature with Immunoscore was assessed. A collagen nomogram was developed by incorporating the collagen signature and clinicopathological predictors after multivariable logistic regression. The performance of the collagen nomogram was evaluated via calibration, discrimination, and clinical usefulness and then tested in an independent validation cohort. The prognostic values of the collagen nomogram were assessed using Cox regression and the Kaplan-Meier method. Results The collagen signature was constructed based on 16 collagen features, which included 6 collagen features from the tumor center and 10 collagen features from the invasive margin. Patients with a high collagen signature were more likely to show a low Immunoscore (Lo IS) in both cohorts (P<0.001). A collagen nomogram integrating the collagen signature and clinicopathological predictors was developed. The collagen nomogram yielded satisfactory discrimination and calibration, with an AUC of 0.925 (95% CI: 0.895-0.956) in the training cohort and 0.911 (95% CI: 0.872-0.949) in the validation cohort. Decision curve analysis confirmed that the collagen nomogram was clinically useful. Furthermore, the collagen nomogram-predicted subgroup was significantly associated with prognosis. Moreover, patients with a low-probability Lo IS, rather than a high-probability Lo IS, could benefit from chemotherapy in high-risk stage II and stage III CRC patients. Conclusions The collagen signature is significantly associated with the Immunoscore in the TME, and the collagen nomogram has the potential to individualize the prediction of the Immunoscore and identify CRC patients who could benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Wei Jiang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- School of Science, Jimei University, Xiamen, Fujian, China
| | - Xian Yu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Xiaoyu Dong
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chenyan Long
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Dexin Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiaxin Cheng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Botao Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shuoyu Xu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zexi Lin
- School of Science, Jimei University, Xiamen, Fujian, China
| | - Gang Chen
- Department of Pathology, The Affiliated Cancer Hospital of Fujian Medical University, Fujian Provincial Cancer Hospital, Fuzhou, China
- Precision Medicine Center, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Shuangmu Zhuo
- School of Science, Jimei University, Xiamen, Fujian, China
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Stepanyan A, Fassan M, Spolverato G, Castagliuolo I, Scarpa M, Scarpa M. IMMUNOREACT 0: Biopsy-based immune biomarkers as predictors of response to neoadjuvant therapy for rectal cancer-A systematic review and meta-analysis. Cancer Med 2023; 12:17878-17890. [PMID: 37537787 PMCID: PMC10523971 DOI: 10.1002/cam4.6423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 07/03/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND The main therapy for rectal cancer patients is neoadjuvant therapy (NT) followed by surgery. Immune biomarkers are emerging as potential predictors of the response to NT. We performed a meta-analysis to estimate their predictive significance. METHODS A systematic literature search of PubMed, Ovid MEDLINE and EMBASE databases was performed to identify eligible studies. Studies on patients with rectal cancer undergoing NT in which the predictive significance of at least one of the immunological markers of interest was assessed by immunohistochemistry (IHC) in pretreatment biopsies were included. RESULTS Seventeen studies reporting sufficient data met the inclusion criteria for meta-analysis. High levels of total CD3+, CD4+ and CD8+ tumor infiltrating lymphocytes (TILs), as well as stromal and intraepithelial CD8+ compartments, significantly predicted good pathological response to NT. Moreover, high levels of total (tumoral and immune cell expression) PD-L1 resulted associated to a good pathological response. On the contrary, high levels of intraepithelial CD4+ TILs were correlated with poor pathological response. FoxP3+ TILs, tumoral PD-L1 and CTLA-4 were not correlated to the treatment response. CONCLUSION This meta-analysis indicated that high-density TILs might be predictive biomarkers of pathological response in patients that underwent NT for rectal cancer.
Collapse
Affiliation(s)
- Astghik Stepanyan
- UOC Chirurgia Generale 3Azienda Ospedale‐Università PadovaPaduaItaly
| | - Matteo Fassan
- Department of Medicine DIMEDUniversity of PaduaPaduaItaly
- Veneto Institute of Oncology IOV‐IRCCSPaduaItaly
| | - Gaya Spolverato
- UOC Chirurgia Generale 3Azienda Ospedale‐Università PadovaPaduaItaly
| | | | - Melania Scarpa
- Immunology and Molecular Oncology Diagnostics UnitVeneto Institute of Oncology IOV‐IRCCSPaduaItaly
| | - Marco Scarpa
- UOC Chirurgia Generale 3Azienda Ospedale‐Università PadovaPaduaItaly
| |
Collapse
|
29
|
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: 5] [Impact Index Per Article: 2.5] [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
|
30
|
Wyrwicz L, Saunders M, Hall M, Ng J, Hong T, Xu S, Lucas J, Lu X, Lautermilch N, Formenti S, Glynne-Jones R. AN0025, a novel antagonist of PGE2-receptor E-type 4 (EP4), in combination with total neoadjuvant treatment of advanced rectal cancer. Radiother Oncol 2023; 185:109669. [PMID: 37054987 DOI: 10.1016/j.radonc.2023.109669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/15/2023]
Abstract
PURPOSE To assess the safety and efficacy of AN0025 in combination with preoperative radiotherapy and chemotherapy in either short course (SCRT) or long course radiotherapy (LCRT) settings for those with locally advanced rectal cancer. PATIENTS AND METHODS Twenty-eight subjects with locally advanced rectal cancer participated in this multicenter, open-label, Phase Ib trial. Enrolled subjects received either 250 mg or 500 mg of AN0025 once daily for 10 weeks with either LCRT or SCRT with chemotherapy (7 subjects/group). Participants were assessed for safety/efficacy starting from the first dose of study drug administration and were followed for 2 years. RESULTS No treatment-emergent adverse or serious adverse events meeting dose-limiting criteria were observed, with only 3 subjects discontinuing AN0025 treatment due to adverse events. Twenty-five of 28 subjects completed 10 weeks of AN0025 and adjuvant therapy and were evaluated for efficacy. Overall, 36.0% of subjects (9/25 subjects) achieved a pathological complete response or a complete clinical response, including 26.7% of subjects (4/15 subjects who underwent surgery) who achieved a pathological complete response. A total of 65.4% of subjects had magnetic resonance imaging-confirmed down-staging ≤ stage 3 following completion of treatment. With a median follow-up of 30 months. The 12-month disease-free survival and overall survival were 77.5% (95% confidence interval [CI]: 56.6, 89.2) and 96.3% (95% CI: 76.5, 99.5), respectively. CONCLUSIONS Treatment with AN0025 administered for 10 weeks along with preoperative SCRT or LCRT did not appear to worsen the toxicity in subjects with locally advanced rectal cancer, was well-tolerated and showed promise in inducing both a pathological and complete clinical response. These findings suggest its activity deserves further investigation in larger clinical trials.
Collapse
Affiliation(s)
- Lucjan Wyrwicz
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Mark Saunders
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Marcia Hall
- Medical Oncology, Mount Vernon Cancer Centre, Northwood, United Kingdom
| | - John Ng
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Theodore Hong
- Massachusetts General Hospital, Harvard Medical School, Hatfield, United Kingdom
| | - Sherry Xu
- Adlai Nortye USA, North Brunswick, NJ, United States
| | - Justin Lucas
- Adlai Nortye USA, North Brunswick, NJ, United States
| | - Xuyang Lu
- Adlai Nortye USA, North Brunswick, NJ, United States
| | | | - Silvia Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | | |
Collapse
|
31
|
Ascierto PA, Agarwala SS, Warner AB, Ernstoff MS, Fox BA, Gajewski TF, Galon J, Garbe C, Gastman BR, Gershenwald JE, Kalinski P, Krogsgaard M, Leidner RS, Lo RS, Menzies AM, Michielin O, Poulikakos PI, Weber JS, Caracò C, Osman I, Puzanov I, Thurin M. Perspectives in Melanoma: meeting report from the Melanoma Bridge (December 1st-3rd, 2022-Naples, Italy). J Transl Med 2023; 21:508. [PMID: 37507765 PMCID: PMC10375730 DOI: 10.1186/s12967-023-04325-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/01/2023] [Indexed: 07/30/2023] Open
Abstract
Outcomes for patients with melanoma have improved over the past decade with the clinical development and approval of immunotherapies targeting immune checkpoint receptors such as programmed death-1 (PD-1), programmed death ligand 1 (PD-L1) or cytotoxic T lymphocyte antigen-4 (CTLA-4). Combinations of these checkpoint therapies with other agents are now being explored to improve outcomes and enhance benefit-risk profiles of treatment. Alternative inhibitory receptors have been identified that may be targeted for anti-tumor immune therapy, such as lymphocyte-activation gene-3 (LAG-3), as have several potential target oncogenes for molecularly targeted therapy, such as tyrosine kinase inhibitors. Unfortunately, many patients still progress and acquire resistance to immunotherapy and molecularly targeted therapies. To bypass resistance, combination treatment with immunotherapies and single or multiple TKIs have been shown to improve prognosis compared to monotherapy. The number of new combinations treatment under development for melanoma provides options for the number of patients to achieve a therapeutic benefit. Many diagnostic and prognostic assays have begun to show clinical applicability providing additional tools to optimize and individualize treatments. However, the question on the optimal algorithm of first- and later-line therapies and the search for biomarkers to guide these decisions are still under investigation. This year, the Melanoma Bridge Congress (Dec 1st-3rd, 2022, Naples, Italy) addressed the latest advances in melanoma research, focusing on themes of paramount importance for melanoma prevention, diagnosis and treatment. This included sessions dedicated to systems biology on immunotherapy, immunogenicity and gene expression profiling, biomarkers, and combination treatment strategies.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy.
| | | | | | - Marc S Ernstoff
- ImmunoOncology Branch (IOB), Developmental Therapeutics Program, Cancer Therapy and Diagnosis Division, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Bernard A Fox
- Robert W. Franz Cancer Center, Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Thomas F Gajewski
- Department of Pathology and Department of Medicine (Section of Hematology/Oncology), University of Chicago, Chicago, IL, USA
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, 75006, Paris, France
- Centre de Recherche Des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - Brian R Gastman
- Department of Surgery, School of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Michelle Krogsgaard
- Laura and Isaac Perlmutter Cancer Center and Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rom S Leidner
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Roger S Lo
- Jonsson Comprehensive Cancer Center David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Olivier Michielin
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Poulikos I Poulikakos
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeffrey S Weber
- Laura and Isaac Perlmutter Cancer Center, a NCI-Funded Comprehensive Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Corrado Caracò
- Division of Surgery of Melanoma and Skin Cancer, Istituto Nazionale Tumori "Fondazione Pascale" IRCCS, Naples, Italy
| | - Iman Osman
- Rudolf L, Baer, New York University Langone Medical Center, New York, NY, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Magdalena Thurin
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
32
|
Tse BCY, Bergamin S, Steffen P, Hruby G, Pavlakis N, Clarke SJ, Evans J, Engel A, Kneebone A, Molloy MP. CD11c + and IRF8 + cell densities in rectal cancer biopsies predict outcomes of neoadjuvant chemoradiotherapy. Oncoimmunology 2023; 12:2238506. [PMID: 37485033 PMCID: PMC10361136 DOI: 10.1080/2162402x.2023.2238506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 07/09/2023] [Accepted: 07/16/2023] [Indexed: 07/25/2023] Open
Abstract
Approximately 20% of locally advanced rectal cancer (LARC) patients treated preoperatively with chemoradiotherapy (CRT) achieve pathologically confirmed complete regression. However, there are no clinically implemented biomarkers measurable in biopsies that are predictive of tumor regression. Here, we conducted multiplexed immunophenotyping of rectal cancer diagnostic biopsies from 16 LARC patients treated preoperatively with CRT. We identified that patients with greater tumor regression had higher tumor infiltration of pan-T cells and IRF8+HLA-DR+ cells prior to CRT. High IRF8+HLA-DR+ cell density was further associated with prolonged disease-specific survival with 83% survival at 5 y compared to 28% in patients with low infiltration. Contrastingly, low CD11c+ myeloid cell infiltration prior to CRT was a putative biomarker associated with longer 3- and 5-y disease-free survival. The results demonstrate the potential use of rectal cancer diagnostic biopsies to measure IRF8+ HLA-DR+ cells as predictors of CRT-induced tumor regression and CD11c+ myeloid cells as predictors of LARC patient survival.
Collapse
Affiliation(s)
- Benita C. Y. Tse
- Bowel Cancer and Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Sarah Bergamin
- Department of Radiation Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Pascal Steffen
- Bowel Cancer and Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - George Hruby
- Department of Radiation Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
| | - Stephen J. Clarke
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
| | - Justin Evans
- Colorectal Surgical Unit, Royal North Shore Hospital, Sydney, Australia
| | - Alexander Engel
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Colorectal Surgical Unit, Royal North Shore Hospital, Sydney, Australia
| | - Andrew Kneebone
- Department of Radiation Oncology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
| | - Mark P. Molloy
- Bowel Cancer and Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| |
Collapse
|
33
|
Mohan H, Rabie M, Walsh C, Harji D, Sutton P, Geh I, Jackson I, Helbren E, Evans M, Jenkins JT. Patient and multidisciplinary team perspectives on watch and wait in rectal cancer. Colorectal Dis 2023; 25:1489-1497. [PMID: 37477408 DOI: 10.1111/codi.16592] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/21/2023] [Accepted: 03/25/2023] [Indexed: 07/22/2023]
Abstract
This article adopts a multidisciplinary approach, including surgery, oncology, radiology and patient perspectives, to discuss the key points of debate surrounding a watch and wait approach. In an era of shared decision-making, discussion of watch and wait as an option in the context of complete clinical response is appropriate, although it is not the gold standard treatment. Key challenges are the difficulty in assessing for a complete clinical response, prediction of recurrence and access to timely diagnostics for surveillance. Salvage surgery has good results if regrowth is detected early but does have imperfect outcomes, with only a 90% salvage rate. Good communication with patients about the risks and alternatives is essential. Patients undergoing watch and wait should ideally be enrolled in prospective registries or clinical trials.
Collapse
Affiliation(s)
- Helen Mohan
- ACPGBI Advanced Malignancy Subcommittee, London, UK
- The Dukes Club, London, UK
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | | | - Ciaran Walsh
- ACPGBI Multidisciplinary Clinical Committee, London, UK
| | | | | | - Ian Geh
- ACPGBI Multidisciplinary Clinical Committee, London, UK
| | | | - Emma Helbren
- British Society of Gastrointestinal Abdominal Radiology (BSGAR), London, UK
| | - Martyn Evans
- ACPGBI Multidisciplinary Clinical Committee, London, UK
| | - John T Jenkins
- ACPGBI Advanced Malignancy Subcommittee, London, UK
- ACPGBI Multidisciplinary Clinical Committee, London, UK
| |
Collapse
|
34
|
Rezapour A, Rydbeck D, Byvald F, Tasselius V, Danielsson G, Angenete E, Yrlid U. A type I interferon footprint in pre-operative biopsies is an independent biomarker that in combination with CD8 + T cell quantification can improve the prediction of response to neoadjuvant treatment of rectal adenocarcinoma. Oncoimmunology 2023; 12:2209473. [PMID: 37180638 PMCID: PMC10173792 DOI: 10.1080/2162402x.2023.2209473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/16/2023] Open
Abstract
Tailored treatment for patients with rectal cancer requires clinically available markers to predict their response to neoadjuvant treatment. The quantity of tumor-infiltrating lymphocytes (TILs) in pre-operative tumor biopsies has been suggested to predict a favorable response, but opposing results exist. A biopsy-adapted Immunoscore (ISB) based on TILs has recently emerged as a promising predictor of tumor regression and prognosis in (colo)rectal cancer. We aimed to refine the ISB for prediction of response using multiplex immunofluorescence (mIF) on pre-operative rectal cancer biopsies. We combined the distribution and density of conventional T cell subsets and γδT cells with a type I Interferon (IFN)-driven response assessed using Myxovirus resistance protein A (MxA) expression. We found that pathological complete response (pCR) following neoadjuvant treatment was associated with type I IFN. Stratification of patients according to the density of CD8+ in the entire tumor tissue and MxA+ cells in tumor stroma, where equal weight was assigned to both parameters, resulted in improved predictive quality compared to the ISB. This novel stratification approach using these two independent parameters in pre-operative biopsies could potentially aid in identifying patients with a good chance of achieving a pCR following neoadjuvant treatment.
Collapse
Affiliation(s)
- Azar Rezapour
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Rydbeck
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
- Department of Surgery, SSORG - Scandinavian Surgical Outcomes Research Group, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fabian Byvald
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Viktor Tasselius
- Department of Surgery, SSORG - Scandinavian Surgical Outcomes Research Group, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Gustaf Danielsson
- Department of Clinical Pathology and Genetics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eva Angenete
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital/Östra, Gothenburg, Sweden
- Department of Surgery, SSORG - Scandinavian Surgical Outcomes Research Group, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
35
|
Shi J, Sun Z, Gao Z, Huang D, Hong H, Gu J. Radioimmunotherapy in colorectal cancer treatment: present and future. Front Immunol 2023; 14:1105180. [PMID: 37234164 PMCID: PMC10206275 DOI: 10.3389/fimmu.2023.1105180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Colorectal cancer (CRC) is a deadly form of cancer worldwide. Patients with locally advanced rectal cancer and metastatic CRC have a poor long-term prognosis, and rational and effective treatment remains a major challenge. Common treatments include multi-modal combinations of surgery, radiotherapy, and chemotherapy; however, recurrence and metastasis rates remain high. The combination of radiotherapy and immunotherapy (radioimmunotherapy [RIT]) may offer new solutions to this problem, but its prospects remain uncertain. This review aimed to summarize the current applications of radiotherapy and immunotherapy, elaborate on the underlying mechanisms, and systematically review the preliminary results of RIT-related clinical trials for CRC. Studies have identified several key predictors of RIT efficacy. Summarily, rational RIT regimens can improve the outcomes of some patients with CRC, but current study designs have limitations. Further studies on RIT should focus on including larger sample sizes and optimizing the combination therapy regimen based on underlying influencing factors.
Collapse
Affiliation(s)
- Jingyi Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhuang Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhaoya Gao
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Dandan Huang
- Department of Oncology, Peking University Shougang Hospital, Beijing, China
| | - Haopeng Hong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jin Gu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China
- Peking Tsinghua Center for Life Science, Peking University International Cancer Center, Beijing, China
| |
Collapse
|
36
|
Moretto R, Rossini D, Catteau A, Antoniotti C, Giordano M, Boccaccino A, Ugolini C, Proietti A, Conca V, Kassambara A, Pietrantonio F, Salvatore L, Lonardi S, Tamberi S, Tamburini E, Poma AM, Fieschi J, Fontanini G, Masi G, Galon J, Cremolini C. Dissecting tumor lymphocyte infiltration to predict benefit from immune-checkpoint inhibitors in metastatic colorectal cancer: lessons from the AtezoT RIBE study. J Immunother Cancer 2023; 11:jitc-2022-006633. [PMID: 37085190 PMCID: PMC10124320 DOI: 10.1136/jitc-2022-006633] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Tumor immune cells influence the efficacy of immune-checkpoint inhibitors (ICIs) and many efforts aim at identifying features of tumor immune microenvironment able to predict benefit from ICIs in proficient mismatch repair (pMMR)/microsatellite stable (MSS) metastatic colorectal cancer (mCRC). METHODS We characterized tumor immune cell infiltrate, by assessing tumor-infiltrating lymphocytes (TILs), Immunoscore, Immunoscore-IC, and programmed death ligand-1 (PD-L1) expression in tumor samples of patients with mCRC enrolled in the AtezoTRIBE study, a phase II randomized trial comparing FOLFOXIRI/bevacizumab/atezolizumab to FOLFOXIRI/bevacizumab, with the aim of evaluating the prognostic and predictive value of these features. RESULTS Out of 218 patients enrolled, 181 (83%), 77 (35%), 157 (72%) and 162 (74%) specimens were successfully tested for TILs, Immunoscore, Immunoscore-IC and PD-L1 expression, respectively, and 69 (38%), 45 (58%), 50 (32%) and 21 (13%) tumors were classified as TILs-high, Immunoscore-high, Immunoscore-IC-high and PD-L1-high, respectively. A poor agreement was observed between TILs and Immunoscore or Immunoscore-IC (K of Cohen <0.20). In the pMMR population, longer progression-free survival (PFS) was reported for Immunoscore-high and Immunoscore-IC-high groups compared with Immunoscore-low (16.4 vs 12.2 months; HR: 0.55, 95% CI: 0.30 to 0.99; p=0.049) and Immunoscore-IC-low (14.8 vs 11.5 months; HR: 0.55, 95% CI: 0.35 to 0.85; p=0.007), respectively, with a significant interaction effect between treatment arms and Immunoscore-IC (p for interaction: 0.006) and a trend for Immunoscore (p for interaction: 0.13). No PFS difference was shown according to TILs and PD-L1 expression. Consistent results were reported in the overall population. CONCLUSIONS The digital evaluation of tumor immune cell infiltrate by means of Immunoscore-IC or Immunoscore identifies the subset of patients with pMMR mCRC achieving more benefit from the addition of the anti-PD-L1 to the upfront treatment. Immunoscore-IC stands as the most promising predictor of benefit from ICIs.
Collapse
Affiliation(s)
- Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Daniele Rossini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Carlotta Antoniotti
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Mirella Giordano
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandra Boccaccino
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Clara Ugolini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Agnese Proietti
- Unit of Pathological Anatomy 3, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Veronica Conca
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lisa Salvatore
- Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Oncologia Medica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sara Lonardi
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Stefano Tamberi
- Oncology Unit, Ravenna Hospital, AUSL Romagna, Ravenna, Italy
| | - Emiliano Tamburini
- Department of Oncology and Palliative Care, Cardinale G Panico, Tricase City Hospital, Tricase, Italy
| | - Anello Marcello Poma
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | | | - Gabriella Fontanini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Gianluca Masi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, F-75006, France
- Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
37
|
Iliadi C, Verset L, Bouchart C, Martinive P, Van Gestel D, Krayem M. The current understanding of the immune landscape relative to radiotherapy across tumor types. Front Immunol 2023; 14:1148692. [PMID: 37006319 PMCID: PMC10060828 DOI: 10.3389/fimmu.2023.1148692] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Radiotherapy is part of the standard of care treatment for a great majority of cancer patients. As a result of radiation, both tumor cells and the environment around them are affected directly by radiation, which mainly primes but also might limit the immune response. Multiple immune factors play a role in cancer progression and response to radiotherapy, including the immune tumor microenvironment and systemic immunity referred to as the immune landscape. A heterogeneous tumor microenvironment and the varying patient characteristics complicate the dynamic relationship between radiotherapy and this immune landscape. In this review, we will present the current overview of the immunological landscape in relation to radiotherapy in order to provide insight and encourage research to further improve cancer treatment. An investigation into the impact of radiation therapy on the immune landscape showed in several cancers a common pattern of immunological responses after radiation. Radiation leads to an upsurge in infiltrating T lymphocytes and the expression of programmed death ligand 1 (PD-L1) which can hint at a benefit for the patient when combined with immunotherapy. In spite of this, lymphopenia in the tumor microenvironment of 'cold' tumors or caused by radiation is considered to be an important obstacle to the patient's survival. In several cancers, a rise in the immunosuppressive populations is seen after radiation, mainly pro-tumoral M2 macrophages and myeloid-derived suppressor cells (MDSCs). As a final point, we will highlight how the radiation parameters themselves can influence the immune system and, therefore, be exploited to the advantage of the patient.
Collapse
Affiliation(s)
- Chrysanthi Iliadi
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
| | - Laurine Verset
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
| | - Christelle Bouchart
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
| | - Philippe Martinive
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
| | - Dirk Van Gestel
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
| | - Mohammad Krayem
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
| |
Collapse
|
38
|
Foersch S, Glasner C, Woerl AC, Eckstein M, Wagner DC, Schulz S, Kellers F, Fernandez A, Tserea K, Kloth M, Hartmann A, Heintz A, Weichert W, Roth W, Geppert C, Kather JN, Jesinghaus M. Multistain deep learning for prediction of prognosis and therapy response in colorectal cancer. Nat Med 2023; 29:430-439. [PMID: 36624314 DOI: 10.1038/s41591-022-02134-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/17/2022] [Indexed: 01/11/2023]
Abstract
Although it has long been known that the immune cell composition has a strong prognostic and predictive value in colorectal cancer (CRC), scoring systems such as the immunoscore (IS) or quantification of intraepithelial lymphocytes are only slowly being adopted into clinical routine use and have their limitations. To address this we established and evaluated a multistain deep learning model (MSDLM) utilizing artificial intelligence (AI) to determine the AImmunoscore (AIS) in more than 1,000 patients with CRC. Our model had high prognostic capabilities and outperformed other clinical, molecular and immune cell-based parameters. It could also be used to predict the response to neoadjuvant therapy in patients with rectal cancer. Using an explainable AI approach, we confirmed that the MSDLM's decisions were based on established cellular patterns of anti-tumor immunity. Hence, the AIS could provide clinicians with a valuable decision-making tool based on the tumor immune microenvironment.
Collapse
Affiliation(s)
- Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany.
| | - Christina Glasner
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Ann-Christin Woerl
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
- Institute of Computer Science, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Eckstein
- Institute of Pathology and Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Stefan Schulz
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Franziska Kellers
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
- Department of Pathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Aurélie Fernandez
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | | | - Michael Kloth
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Arndt Hartmann
- Institute of Pathology and Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Achim Heintz
- Department of General Visceral and Vascular Surgery, Marien Hospital Mainz, Mainz, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Carol Geppert
- Institute of Pathology and Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, Technical University Munich, Munich, Germany
- Institute of Pathology, University Hospital Marburg, Marburg, Germany
| |
Collapse
|
39
|
Machado Carvalho JV, Dutoit V, Corrò C, Koessler T. Promises and Challenges of Predictive Blood Biomarkers for Locally Advanced Rectal Cancer Treated with Neoadjuvant Chemoradiotherapy. Cells 2023; 12:413. [PMID: 36766755 PMCID: PMC9913546 DOI: 10.3390/cells12030413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
The treatment of locally advanced rectal cancer (LARC) requires a multimodal approach combining neoadjuvant radiotherapy or chemoradiotherapy (CRT) and surgery. Predicting tumor response to CRT can guide clinical decision making and improve patient care while avoiding unnecessary toxicity and morbidity. Circulating biomarkers offer both the advantage to be easily accessed and followed over time. In recent years, biomarkers such as proteins, blood cells, or nucleic acids have been investigated for their predictive value in oncology. We conducted a comprehensive literature review with the aim to summarize the status of circulating biomarkers predicting response to CRT in LARC. Forty-nine publications, of which forty-seven full-text articles, one review and one systematic review, were retrieved. These studies evaluated circulating markers (CEA and CA 19-9), inflammatory biomarkers (CRP, albumin, and lymphocytes), hematologic markers (hemoglobin and thrombocytes), lipids and circulating nucleic acids (cell-free DNA [cfDNA], circulating tumor DNA [ctDNA], and microRNA [miRNA]). Post-CRT CEA levels had the most consistent association with tumor response, while cfDNA integrity index, MGMT promoter methylation, ERCC-1, miRNAs, and miRNA-related SNPs were identified as potential predictive markers. Although circulating biomarkers hold great promise, inconsistent results, low statistical power, and low specificity and sensibility prevent them from reliably predicting tumor response following CRT. Validation and standardization of methods and technologies are further required to confirm results.
Collapse
Affiliation(s)
- Joao Victor Machado Carvalho
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Swiss Cancer Center Léman, 1005 Lausanne, Switzerland
- Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Valérie Dutoit
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Swiss Cancer Center Léman, 1005 Lausanne, Switzerland
| | - Claudia Corrò
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Swiss Cancer Center Léman, 1005 Lausanne, Switzerland
- Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Thibaud Koessler
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Swiss Cancer Center Léman, 1005 Lausanne, Switzerland
- Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
| |
Collapse
|
40
|
Jeon SH, Chie EK. Characterization of the gene signature correlated with favorable response to chemoradiotherapy in rectal cancer: A hypothesis-generating study. Cancer Med 2023; 12:8981-8990. [PMID: 36621808 PMCID: PMC10134325 DOI: 10.1002/cam4.5586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/17/2022] [Accepted: 12/21/2022] [Indexed: 01/10/2023] Open
Abstract
PURPOSE This study aimed to define the gene signature associated with response to neoadjuvant chemoradiotherapy (nCRT), or chemoradiosensitivity (CRS) signature, in rectal cancer, and investigate the correlation between the CRS signature and characteristics of tumor. MATERIALS AND METHODS Three public microarray datasets of pre-nCRT rectal cancer were used to discover and validate the CRS signature, and the pathway analysis of the CRS signature was performed. Patients in The Cancer Genome Atlas (TCGA) dataset were stratified according to the CRS signature enrichment score, and mutational profile and proportions of infiltrated immune cells were compared. RESULTS In the discovery dataset (GSE53781), 95 genes were upregulated in complete responders compared to non-complete responders and defined as the CRS signature. Pathways regarding DNA replication and repair processes as well as inflammatory response were enriched in the CRS signature. In the validation datasets (GSE35452 and GSE45404), patients with favorable response to nCRT exhibited higher enrichment score of the CRS. In TCGA-READ cohort, patients with high CRS signature harbored KRAS mutation in lower frequency than those with low CRS signature. In addition, proportions of proinflammatory immune cells were higher, but proportion of immunosuppressive M2 macrophages was lower in patients with high CRS signature than those with low CRS signature. CONCLUSIONS The current integrative bioinformatic analysis suggests the CRS signature and showed that the CRS signature is associated with dissimilar mutational profile and increased immune response. The discovered CRS signature and related characteristics may serve as candidate of stratification factor in upcoming studies for rectal cancer.
Collapse
Affiliation(s)
- Seung Hyuck Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Eui Kyu Chie
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
41
|
Akiyoshi T, Wang Z, Kaneyasu T, Gotoh O, Tanaka N, Amino S, Yamamoto N, Kawachi H, Mukai T, Hiyoshi Y, Nagasaki T, Yamaguchi T, Konishi T, Fukunaga Y, Noda T, Mori S. Transcriptomic Analyses of Pretreatment Tumor Biopsy Samples, Response to Neoadjuvant Chemoradiotherapy, and Survival in Patients With Advanced Rectal Cancer. JAMA Netw Open 2023; 6:e2252140. [PMID: 36662520 PMCID: PMC9860531 DOI: 10.1001/jamanetworkopen.2022.52140] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/03/2022] [Indexed: 01/21/2023] Open
Abstract
Importance Neoadjuvant chemoradiotherapy (CRT) is the standard of care for advanced rectal cancer. Yet, estimating response to CRT remains an unmet clinical challenge. Objective To investigate and better understand the transcriptomic factors associated with response to neoadjuvant CRT and survival in patients with advanced rectal cancer. Design, Setting, and Participants A single-center, retrospective, case series was conducted at a comprehensive cancer center. Pretreatment biopsies from 298 patients with rectal cancer who were later treated with neoadjuvant CRT between April 1, 2004, and September 30, 2020, were analyzed by RNA sequencing. Data analysis was performed from July 1, 2021, to May 31, 2022. Exposures Chemoradiotherapy followed by total mesorectal excision or watch-and-wait management. Main Outcomes and Measures Transcriptional subtyping was performed by consensus molecular subtype (CMS) classification. Immune cell infiltration was assessed using microenvironment cell populations-counter (MCP-counter) scores and single-sample gene set enrichment analysis (ssGSEA). Patients with surgical specimens of tumor regression grade 3 to 4 or whose care was managed by the watch-and-wait approach for more than 3 years were defined as good responders. Results Of the 298 patients in the study, 205 patients (68.8%) were men, and the median age was 61 (IQR, 52-67) years. Patients classified as CMS1 (6.4%) had a significantly higher rate of good response, albeit survival was comparable among the 4 subtypes. Good responders exhibited an enrichment in various immune-related pathways, as determined by ssGSEA. Microenvironment cell populations-counter scores for cytotoxic lymphocytes were significantly higher for good responders than nonresponders (median, 0.76 [IQR, 0.53-1.01] vs 0.58 [IQR, 0.43-0.83]; P < .001). Cytotoxic lymphocyte MCP-counter score was independently associated with response to CRT, as determined in the multivariable analysis (odds ratio, 3.81; 95% CI, 1.82-7.97; P < .001). Multivariable Cox proportional hazards regression analysis, including postoperative pathologic factors, revealed the cytotoxic lymphocyte MCP-counter score to be independently associated with recurrence-free survival (hazard ratio [HR], 0.38; 95% CI, 0.16-0.92; P = .03) and overall survival (HR, 0.16; 95% CI, 0.03-0.83; P = .03). Conclusions and Relevance In this case series of patients with rectal cancer treated with neoadjuvant CRT, the cytotoxic lymphocyte score in pretreatment biopsy samples, as computed by RNA sequencing, was associated with response to CRT and survival. This finding suggests that the cytotoxic lymphocyte score might serve as a biomarker in personalized multimodal rectal cancer treatment.
Collapse
Affiliation(s)
- Takashi Akiyoshi
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Zhe Wang
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomoko Kaneyasu
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Osamu Gotoh
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Norio Tanaka
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Sayuri Amino
- Project for Development of Genomics-Based Cancer Medicine, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Noriko Yamamoto
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroshi Kawachi
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiki Mukai
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yukiharu Hiyoshi
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiya Nagasaki
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomohiro Yamaguchi
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tsuyoshi Konishi
- Department of Colon and Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston
| | - Yosuke Fukunaga
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tetsuo Noda
- Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Seiichi Mori
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
42
|
Cerdán-Santacruz C, Vailati BB, São Julião GP, Habr-Gama A, Perez RO. Local tumor regrowth after clinical complete response following neoadjuvant therapy for rectal cancer: what happens when organ preservation falls short. Tech Coloproctol 2023; 27:1-9. [PMID: 35986804 DOI: 10.1007/s10151-022-02654-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/09/2022] [Indexed: 01/12/2023]
Abstract
Organ preservation strategies, especially watch and wait, after neoadjuvant treatment in locally advanced rectal cancer, have become topics that generate significant interest, for both patients and clinicians. The obvious advantage of these strategies is the avoidance of surgery with its associated risks and functional consequences. Over time, it has become evident that these strategies offer acceptable safety in oncological terms and, in most patients, allows preservation of the rectum without harming patients in terms of distant metastasis or survival. However, there is a small group of patients in whom the tumor returns after an initially diagnosed clinical complete response; patients with local tumor regrowth. The main threat in these patients is not simply local disease, which can be successfully managed in most cases, but the possible effects it may have on distant metastases. The pathophysiology of the phenomenon of local tumor regrowth is not well known and, therefore, strategies to minimize possible impact on survival are not well defined. Our aim is to review key issues in this subgroup that pose a substantial threat to the safety and viability of organ-preserving and watch-and-wait strategies. We also explore possible pathophysiologic explanations and future directions and perspectives that may improve both local and systemic disease control.
Collapse
Affiliation(s)
- C Cerdán-Santacruz
- Colorectal Surgery, Hospital Universitario de la Princesa, Madrid, Spain
| | - B B Vailati
- Department of Surgical Oncology, Hospital Beneficencia Portuguesa, São Paulo, Brazil
- Division of Colorectal Surgery, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
- Angelita and Joaquim Gama Institute, Praça Amadeu Amaral 47, con. 111, São Paulo, 01327-904, Brazil
| | - G P São Julião
- Department of Surgical Oncology, Hospital Beneficencia Portuguesa, São Paulo, Brazil
- Division of Colorectal Surgery, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
- Angelita and Joaquim Gama Institute, Praça Amadeu Amaral 47, con. 111, São Paulo, 01327-904, Brazil
| | - A Habr-Gama
- Department of Surgical Oncology, Hospital Beneficencia Portuguesa, São Paulo, Brazil
- Division of Colorectal Surgery, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil
- Angelita and Joaquim Gama Institute, Praça Amadeu Amaral 47, con. 111, São Paulo, 01327-904, Brazil
| | - R O Perez
- Department of Surgical Oncology, Hospital Beneficencia Portuguesa, São Paulo, Brazil.
- Division of Colorectal Surgery, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil.
- Angelita and Joaquim Gama Institute, Praça Amadeu Amaral 47, con. 111, São Paulo, 01327-904, Brazil.
| |
Collapse
|
43
|
Wang Y, Shen L, Wan J, Zhang H, Wu R, Wang J, Wang Y, Xu Y, Cai S, Zhang Z, Xia F. Neoadjuvant chemoradiotherapy combined with immunotherapy for locally advanced rectal cancer: A new era for anal preservation. Front Immunol 2022; 13:1067036. [PMID: 36569918 PMCID: PMC9772444 DOI: 10.3389/fimmu.2022.1067036] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
For locally advanced (T3-4/N+M0) rectal cancer (LARC), neoadjuvant chemoradiotherapy (nCRT) followed by total mesorectal excision (TME) is the standard treatment. It was demonstrated to decrease the local recurrence rate and increase the tumor response grade. However, the distant metastasis remains an unresolved issue. And the demand for anus preservation and better quality of life increases in recent years. Radiotherapy and immunotherapy can be supplement to each other and the combination of the two treatments has a good theoretical basis. Recently, multiple clinical trials are ongoing in terms of the combination of nCRT and immunotherapy in LARC. It was reported that these trials achieved promising short-term efficacy in both MSI-H and MSS rectal cancers, which could further improve the rate of clinical complete response (cCR) and pathological complete response (pCR), so that increase the possibility of 'Watch and Wait (W&W)' approach. However, the cCR and pCR is not always consistent, which occurs more frequent when nCRT is combined with immunotherapy. Thus, the efficacy evaluation after neoadjuvant therapy is an important issue for patient selection of W&W approach. Evaluating the cCR accurately needs the combination of multiple traditional examinations, new detective methods, such as PET-CT, ctDNA-MRD and various omics studies. And finding accurate biomarkers can help guide the risk stratification and treatment decisions. And large-scale clinical trials need to be performed in the future to demonstrate the surprising efficacy and to explore the long-term prognosis.
Collapse
Affiliation(s)
- Yaqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China,Shanghai Key Laboratory of Radiation Oncology, 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,Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China,Shanghai Key Laboratory of Radiation Oncology, 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,Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China,Shanghai Key Laboratory of Radiation Oncology, 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,Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Ruiyan Wu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Jingwen Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China,Shanghai Key Laboratory of Radiation Oncology, 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,Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Ye Xu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Sanjun Cai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China,*Correspondence: Fan Xia, ; Zhen Zhang,
| | - Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China,*Correspondence: Fan Xia, ; Zhen Zhang,
| |
Collapse
|
44
|
Jameson MB, Stevenson ARL, Ngan SY. TNT: Raising more questions than answers? Asia Pac J Clin Oncol 2022; 18:489-492. [PMID: 35322550 DOI: 10.1111/ajco.13760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/16/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Michael B Jameson
- Waikato Clinical Campus, University of Auckland, Hamilton, New Zealand.,Oncology Department, Waikato District Health Board, Hamilton, New Zealand
| | - Andrew R L Stevenson
- Department of Surgery, University of Queensland, Brisbane, Queensland, Australia
| | - Samuel Y Ngan
- Division of Radiation Oncology, Sir Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
45
|
Cho H, Kim JE, Hong YS, Kim SY, Kim J, Ryu YM, Kim SY, Kim TW. Comprehensive evaluation of the tumor immune microenvironment and its dynamic changes in patients with locally advanced rectal cancer treated with preoperative chemoradiotherapy: From the phase II ADORE study. Oncoimmunology 2022; 11:2148374. [PMID: 36451674 PMCID: PMC9704411 DOI: 10.1080/2162402x.2022.2148374] [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] [Indexed: 11/27/2022] Open
Abstract
A better understanding of the effects of preoperative chemoradiotherapy (CRT) on tumor immune microenvironment (TIME) is essential to improve the treatment outcomes of patients with locally advanced rectal cancer (LARC). In this context, we performed a multiplex immunofluorescence staining to evaluate the TIME in 158 patients with LARC who underwent preoperative CRT followed by surgery and adjuvant chemotherapy in the ADORE trial. We found that higher levels of T-cell subsets (CD3+, CD4+, and CD8+) and dendritic cells in the tumor compartment of pretreatment biopsy samples were associated with good response to preoperative CRT. After CRT, there was a significant increase in the densities of CD3+ T cells, CD8+ T cells, and dendritic cells, while that of CD4+FoxP3+ regulatory T cells decreased, indicating that CRT changed the TIME into a more immune-active status. However, CRT also conferred an immunosuppressive effect by polarizing the tumor-associated macrophages from pro-inflammatory M1 macrophage to immune-suppressive M2 macrophages and decreasing the density of B cells. High delta values of CD3+ T cells and PD-L1+ lymphocytes after CRT were associated with good disease-free survival (DFS), while that of CD4+FoxP3+ regulatory T cells was associated with poor DFS. These findings provide a framework for future studies incorporating strategies to modulate the TIME in patients with LARC.
Collapse
Affiliation(s)
- Hyungwoo Cho
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Eun Kim
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yong Sang Hong
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sun Young Kim
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jihun Kim
- Departments of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yeon-Mi Ryu
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae Won Kim
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea,CONTACT Tae Won Kim Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, Korea
| |
Collapse
|
46
|
Chatila WK, Kim JK, Walch H, Marco MR, Chen CT, Wu F, Omer DM, Khalil DN, Ganesh K, Qu X, Luthra A, Choi SH, Ho YJ, Kundra R, Groves KI, Chow OS, Cercek A, Weiser MR, Widmar M, Wei IH, Pappou EP, Nash GM, Paty PB, Shi Q, Vakiani E, Duygu Selcuklu S, Donoghue MTA, Solit DB, Berger MF, Shia J, Pelossof R, Romesser PB, Yaeger R, Smith JJ, Schultz N, Sanchez-Vega F, Garcia-Aguilar J. Genomic and transcriptomic determinants of response to neoadjuvant therapy in rectal cancer. Nat Med 2022; 28:1646-1655. [PMID: 35970919 PMCID: PMC9801308 DOI: 10.1038/s41591-022-01930-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 06/29/2022] [Indexed: 01/03/2023]
Abstract
The incidence of rectal cancer is increasing in patients younger than 50 years. Locally advanced rectal cancer is still treated with neoadjuvant radiation, chemotherapy and surgery, but recent evidence suggests that patients with a complete response can avoid surgery permanently. To define correlates of response to neoadjuvant therapy, we analyzed genomic and transcriptomic profiles of 738 untreated rectal cancers. APC mutations were less frequent in the lower than in the middle and upper rectum, which could explain the more aggressive behavior of distal tumors. No somatic alterations had significant associations with response to neoadjuvant therapy in a treatment-agnostic manner, but KRAS mutations were associated with faster relapse in patients treated with neoadjuvant chemoradiation followed by consolidative chemotherapy. Overexpression of IGF2 and L1CAM was associated with decreased response to neoadjuvant therapy. RNA-sequencing estimates of immune infiltration identified a subset of microsatellite-stable immune hot tumors with increased response and prolonged disease-free survival.
Collapse
Affiliation(s)
- Walid K Chatila
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Tri-Institutional Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Jin K Kim
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Henry Walch
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael R Marco
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chin-Tung Chen
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fan Wu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana M Omer
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Danny N Khalil
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Karuna Ganesh
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xuan Qu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anisha Luthra
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Seo-Hyun Choi
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu-Jui Ho
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ritika Kundra
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katharine I Groves
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Oliver S Chow
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York-Presbyterian, New York, NY, USA
| | - Andrea Cercek
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin R Weiser
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Widmar
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Iris H Wei
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emmanouil P Pappou
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Garrett M Nash
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Philip B Paty
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Efsevia Vakiani
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Duygu Selcuklu
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark T A Donoghue
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael F Berger
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jinru Shia
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raphael Pelossof
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul B Romesser
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rona Yaeger
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - J Joshua Smith
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francisco Sanchez-Vega
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Julio Garcia-Aguilar
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
47
|
Fiehn AMK, Reiss B, Gögenur M, Bzorek M, Gögenur I. Development of a Fully Automated Method to Obtain Reproducible Lymphocyte Counts in Patients With Colorectal Cancer. Appl Immunohistochem Mol Morphol 2022; 30:493-500. [PMID: 35703148 DOI: 10.1097/pai.0000000000001041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/27/2022] [Indexed: 11/26/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. Although clinical outcome varies among patients diagnosed within the same TNM stage it is the cornerstone in treatment decisions as well as follow-up programmes. Tumor-infiltrating lymphocytes have added value when evaluating survival outcomes. The aim of this study was to develop a fully automated method for quantification of subsets of T lymphocytes in the invasive margin and central tumor in patients with CRC based on Deep Learning powered artificial intelligence. The study cohort consisted of 163 consecutive patients with a primary diagnosis of CRC followed by a surgical resection. Double-labeling immunohistochemical staining with cytokeratin in combination with CD3 or CD8, respectively, was performed on 1 representative slide from each patient. Visiopharm Quantitative Digital Pathology software was used to develop Application Protocol Packages for visualization of architectural details (background, normal epithelium, cancer epithelium, surrounding tissue), identification of central tumor and invasive margin as well as subsequent quantitative analysis of immune cells. Fully automated counts for CD3 and CD8 positive T cells were obtained in 93% and 92% of the cases, respectively. In the remaining cases, manual editing was required. In conclusion, the development of a fully automated method for counting CD3 + and CD8 + lymphocytes in a cohort of patients with CRC provided excellent results eliminating not only observer variability in lymphocyte counts but also in identifying the regions of interest for the quantitative analysis. Validation of the performance of the Application Protocol Packages including clinical correlation is needed.
Collapse
Affiliation(s)
- Anne-Marie K Fiehn
- Department of Pathology
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Roskilde
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Mikail Gögenur
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Roskilde
| | | | - Ismail Gögenur
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Roskilde
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
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: 9] [Impact Index Per Article: 3.0] [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
|
49
|
Ascierto PA, Avallone A, Bhardwaj N, Bifulco C, Bracarda S, Brody JD, Buonaguro L, Demaria S, Emens LA, Ferris RL, Galon J, Khleif SN, Klebanoff CA, Laskowski T, Melero I, Paulos CM, Pignata S, Ruella M, Svane IM, Taube JM, Fox BA, Hwu P, Puzanov I. Perspectives in Immunotherapy: meeting report from the Immunotherapy Bridge, December 1st-2nd, 2021. J Transl Med 2022; 20:257. [PMID: 35672823 PMCID: PMC9172186 DOI: 10.1186/s12967-022-03471-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/30/2022] [Indexed: 01/16/2023] Open
Abstract
Over the past decade, immunotherapy has become an increasingly fundamental modality in the treatment of cancer. The positive impact of immune checkpoint inhibition, especially anti-programmed death (PD)-1/PD-ligand (L)1 blockade, in patients with different cancers has focused attention on the potential for other immunotherapeutic approaches. These include inhibitors of additional immune checkpoints, adoptive cell transfer (ACT), and therapeutic vaccines. Patients with advanced cancers who previously had limited treatment options available may now benefit from immunotherapies that can offer durable responses and improved survival outcomes. However, despite this, a significant proportion of patients fail to respond to immunotherapy, especially those with less immunoresponsive cancer types, and there remains a need for new treatment strategies.The virtual Immunotherapy Bridge (December 1st-2nd, 2021), organized by the Fondazione Melanoma Onlus, Naples, Italy in collaboration with the Society for Immunotherapy of Cancer addressed several areas of current research in immunotherapy, including lessons learned from cell therapies, drivers of immune response, and trends in immunotherapy across different cancers, and these are summarised here.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy.
| | - Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carlo Bifulco
- Providence Genomics and Earle A. Chiles Research Institute, Portland, OR, USA
| | - Sergio Bracarda
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, Terni, Italy
| | - Joshua D Brody
- Department of Medicine, Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luigi Buonaguro
- Department of Experimental Oncology, Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College; Sandra and Edward Meyer Cancer Center; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Leisha A Emens
- Magee Women's Hospital/UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | | | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology/Equipe Labellisée Ligue Contre Le Cancer/Centre de Recherche Des Cordeliers, Sorbonne Université, Université Paris Cité, Marseille, France
| | - Samir N Khleif
- The Loop Immuno Oncology Laboratory, Georgetown University Medical School, Washington, DC, USA
| | - Christopher A Klebanoff
- Human Oncology and Pathogenesis Program, Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center (MSKCC)/Center for Cell Engineering, MSKCC/Parker Institute for Cancer Immunotherapy/Weill Cornell Medical College, New York, NY, USA
| | - Tamara Laskowski
- Head of New Therapeutic Products - Personalized Medicine, Lonza Global, Houston, TX, USA
| | - Ignacio Melero
- Department of Immunology and Immunotherapy, Clinica Universidad de Navarra and CIBERONC, Pamplona, Spain
| | | | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Marco Ruella
- Center for Cellular Immunotherapies and Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Janis M Taube
- Department of Dermatology, Johns Hopkins University SOM, Baltimore, MD, USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Research Center, Providence Cancer Institute, Portland, OR, USA
| | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
50
|
Tumor-Infiltrating Lymphocytes (TILs) in Early Breast Cancer Patients: High CD3+, CD8+, and Immunoscore Are Associated with a Pathological Complete Response. Cancers (Basel) 2022; 14:cancers14102525. [PMID: 35626126 PMCID: PMC9139282 DOI: 10.3390/cancers14102525] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary In 2021, the World Health Organization announced that breast cancer had overtaken lung cancer to become the most common cancer globally, accounting for 12% of all new cancer cases, with younger women resident in low-income countries having the lowest 5-year survival rates. The main aim of the current study was to evaluate the prognostic utility of an innovative, objective, computer-assisted, digital imaging procedure known as the Immunoscore for clinical research (ISCR) as a strategy to reveal the efficiency of the anti-tumor cellular immune landscape of the tumor microenvironment (TME) in biopsies taken from women diagnosed with early breast cancer prior to administration of neoadjuvant chemotherapy followed by surgical resection. Our results demonstrated the ability of the ISCR to enumerate tumor-infiltrating lymphocytes in the TME and, in particular, to illustrate the spatial arrangement of these cells, which, importantly, correlated with clinical outcome, measured as the pathological complete response. Abstract Background: Tumor-infiltrating lymphocytes are associated with a better prognosis in early triple-negative breast cancer (TNBC). These cells can be enumerated in situ by the “Immunoscore Clinical Research” (ISCR). The original Immunoscore® is a prognostic tool that categorizes the densities of CD3+ and CD8+ cells in both the invasive margin (IM) and center of the tumor (CT) in localized colon cancer, yielding a five-tiered classification (0–4). We evaluated the prognostic potential of ISCR and pathological complete response (pCR) following neoadjuvant chemotherapy (NACT). Methods: The cohort included 53 TNBC, 32 luminal BC, and 18 HER2-positive BC patients undergoing NACT. Pre-treatment tumor biopsies were immune-stained for CD3+ and CD8+ T-cell markers. Quantitative analysis of these cells in different tumor locations was performed using computer-assisted image analysis. Results: The pCR rate was 44%. Univariate analysis showed that primary tumor size, estrogen-receptor negative, progesterone-receptor negative, luminal vs. HER2-positive vs. TNBC, high Ki-67, high densities (cells/mm2) of CD3 CT, CD8+ CT, CD3+ IM, and CD8+ IM cells were associated with a high pCR. ISCR was associated with pCR following NACT. A multivariate model consisting of ISCR and the significant variables from the univariate analysis showed a significant trend for ISCR; however, the low sample size did not provide enough power for the model to be included in this study. Conclusions: These results revealed a significant prognostic role for the spatial distributions of the CD3+, and CD8+ lymphocytes, as well as the ISCR in relation to pCR following NACT.
Collapse
|