1
|
Wenzl K, Stokes ME, Novak JP, Bock AM, Khan S, Hopper MA, Krull JE, Dropik AR, Walker JS, Sarangi V, Mwangi R, Ortiz M, Stong N, Huang CC, Maurer MJ, Rimsza L, Link BK, Slager SL, Asmann Y, Mondello P, Morin R, Ansell SM, Habermann TM, Witzig TE, Feldman AL, King RL, Nowakowski G, Cerhan JR, Gandhi AK, Novak AJ. Multiomic analysis identifies a high-risk signature that predicts early clinical failure in DLBCL. Blood Cancer J 2024; 14:100. [PMID: 38902256 PMCID: PMC11189905 DOI: 10.1038/s41408-024-01080-0] [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: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/22/2024] Open
Abstract
Recent genetic and molecular classification of DLBCL has advanced our knowledge of disease biology, yet were not designed to predict early events and guide anticipatory selection of novel therapies. To address this unmet need, we used an integrative multiomic approach to identify a signature at diagnosis that will identify DLBCL at high risk of early clinical failure. Tumor biopsies from 444 newly diagnosed DLBCL were analyzed by WES and RNAseq. A combination of weighted gene correlation network analysis and differential gene expression analysis was used to identify a signature associated with high risk of early clinical failure independent of IPI and COO. Further analysis revealed the signature was associated with metabolic reprogramming and identified cases with a depleted immune microenvironment. Finally, WES data was integrated into the signature and we found that inclusion of ARID1A mutations resulted in identification of 45% of cases with an early clinical failure which was validated in external DLBCL cohorts. This novel and integrative approach is the first to identify a signature at diagnosis, in a real-world cohort of DLBCL, that identifies patients at high risk for early clinical failure and may have significant implications for design of therapeutic options.
Collapse
Affiliation(s)
- Kerstin Wenzl
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Matthew E Stokes
- Informatics and Predictive Sciences, , Bristol Myers Squibb, Summit, NJ, USA
| | | | | | - Sana Khan
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Vivekananda Sarangi
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Raphael Mwangi
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Maria Ortiz
- Informatics and Predictive Sciences, Celgene Institute for Translational Research Europe (CITRE), Seville, Spain
| | - Nicholas Stong
- Informatics and Predictive Sciences, , Bristol Myers Squibb, Summit, NJ, USA
| | - C Chris Huang
- Translational Medicine Hematology, Bristol Myers Squibb, Summit, NJ, USA
| | - Matthew J Maurer
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Lisa Rimsza
- Division of Hematopathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Brian K Link
- Division of Hematology, University of Iowa, Iowa, USA
| | - Susan L Slager
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Yan Asmann
- Department of Quantitative Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | - Ryan Morin
- Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | | | | | | | - Andrew L Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Rebecca L King
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - James R Cerhan
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Anita K Gandhi
- Translational Medicine Hematology, Bristol Myers Squibb, Summit, NJ, USA
| | - Anne J Novak
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Bantilan KS, Smith AN, Maurer MJ, Teruya-Feldstein J, Matasar MJ, Moskowitz AJ, Straus DJ, Noy A, Palomba ML, Horwitz SM, Hamlin PA, Portlock CS, Cerhan JR, Habermann TM, Salles GA, Nowakowski GS, Moskowitz CH, Zelenetz AD. Matched control analysis suggests that R-CHOP followed by (R)-ICE may improve outcome in non-GCB DLBCL compared with R-CHOP. Blood Adv 2024; 8:2172-2181. [PMID: 38271621 PMCID: PMC11068505 DOI: 10.1182/bloodadvances.2023011408] [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: 08/16/2023] [Revised: 11/17/2023] [Accepted: 12/06/2023] [Indexed: 01/27/2024] Open
Abstract
ABSTRACT Rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) is considered the standard-of-care for patients with advanced-stage diffuse large B-cell lymphoma (DLBCL), despite findings that patients with nongerminal center B-cell like (non-GCB) have significantly worse outcome with this regimen. We evaluated the prognostic significance of baseline risk factors, including cell of origin (COO) classified by the Hans algorithm, within an alternative chemoimmunotherapy program. At Memorial Sloan Kettering Cancer Center (MSK), 151 patients with DLBCL received sequential R-CHOP induction and (R)-ICE (rituximab, ifosfamide, carboplatin, and etoposide) consolidation. Outcome analysis based on COO was validated with a propensity score-matched cohort treated with R-CHOP from the Mayo Clinic component of the Molecular Epidemiology Resource (MER). Among the patients with GCB (n = 69) and non-GCB (n = 69) at MSK, event-free survival (EFS) of non-GCB was superior to that of GCB (hazard ratio [HR], 0.53; 95% confidence interval [CI], 0.29-0.98). Overall survival (OS) demonstrated an association in the same direction but was not statistically significant (HR, 0.68; 95% CI, 0.33-1.42). Propensity score-matched patients from MSK (n = 108) demonstrated a small attenuation in the HRs for EFS (HR, 0.57; 95% CI, 0.27-1.18) and OS (HR, 0.76; 95% CI, 0.33-1.79) and were no longer statistically significant. In contrast, the matched MER cohort (n = 108) demonstrated an EFS association (HR, 1.17; 95% CI, 0.70-1.95) and OS association (HR, 1.13; 95% CI, 0.64-2.00) in the opposite direction, but were also not statistically significant. R-CHOP induction and (R)-ICE consolidation may overcome the negative prognostic impact of the non-GCB phenotype, per the Hans algorithm, and can be preferentially selected for this population. This trial was registered at www.ClinicalTrials.gov as #NCT00039195 and #NCT00712582.
Collapse
Affiliation(s)
- Kurt S. Bantilan
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | - Matthew J. Matasar
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Alison J. Moskowitz
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - David J. Straus
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ariela Noy
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - M. Lia Palomba
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Steven M. Horwitz
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Paul A. Hamlin
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carol S. Portlock
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - James R. Cerhan
- Department of Quantitative Sciences, Mayo Clinic, Rochester, MN
| | | | - Gilles A. Salles
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | | | - Craig H. Moskowitz
- Department of Medicine, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL
| | - Andrew D. Zelenetz
- Department of Medicine, Division of Hematologic Malignancies, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
3
|
Kehl A, Aupperle-Lellbach H, de Brot S, van der Weyden L. Review of Molecular Technologies for Investigating Canine Cancer. Animals (Basel) 2024; 14:769. [PMID: 38473154 PMCID: PMC10930838 DOI: 10.3390/ani14050769] [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: 12/21/2023] [Revised: 02/09/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Genetic molecular testing is starting to gain traction as part of standard clinical practice for dogs with cancer due to its multi-faceted benefits, such as potentially being able to provide diagnostic, prognostic and/or therapeutic information. However, the benefits and ultimate success of genomic analysis in the clinical setting are reliant on the robustness of the tools used to generate the results, which continually expand as new technologies are developed. To this end, we review the different materials from which tumour cells, DNA, RNA and the relevant proteins can be isolated and what methods are available for interrogating their molecular profile, including analysis of the genetic alterations (both somatic and germline), transcriptional changes and epigenetic modifications (including DNA methylation/acetylation and microRNAs). We also look to the future and the tools that are currently being developed, such as using artificial intelligence (AI) to identify genetic mutations from histomorphological criteria. In summary, we find that the molecular genetic characterisation of canine neoplasms has made a promising start. As we understand more of the genetics underlying these tumours and more targeted therapies become available, it will no doubt become a mainstay in the delivery of precision veterinary care to dogs with cancer.
Collapse
Affiliation(s)
- Alexandra Kehl
- Laboklin GmbH & Co. KG, Steubenstr. 4, 97688 Bad Kissingen, Germany; (A.K.); (H.A.-L.)
- School of Medicine, Institute of Pathology, Technical University of Munich, Trogerstr. 18, 81675 München, Germany
| | - Heike Aupperle-Lellbach
- Laboklin GmbH & Co. KG, Steubenstr. 4, 97688 Bad Kissingen, Germany; (A.K.); (H.A.-L.)
- School of Medicine, Institute of Pathology, Technical University of Munich, Trogerstr. 18, 81675 München, Germany
| | - Simone de Brot
- Institute of Animal Pathology, COMPATH, University of Bern, 3012 Bern, Switzerland;
| | | |
Collapse
|
4
|
Wenzl K, Stokes M, Novak JP, Bock AM, Khan S, Hopper MA, Krull JE, Dropik AR, Walker JS, Sarangi V, Mwangi R, Ortiz M, Stong N, Huang CC, Maurer MJ, Rimsza L, Link BK, Slager SL, Asmann Y, Mondello P, Morin R, Ansell SM, Habermann TM, Feldman AL, King RL, Nowakowski G, Cerhan JR, Gandhi AK, Novak AJ. Multiomic Analysis Identifies a High-Risk Metabolic and TME Depleted Signature that Predicts Early Clinical Failure in DLBCL. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.07.23290748. [PMID: 37333387 PMCID: PMC10274962 DOI: 10.1101/2023.06.07.23290748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
PURPOSE 60-70% of newly diagnosed diffuse large B-cell lymphoma (DLBCL) patients avoid events within 24 months of diagnosis (EFS24) and the remainder have poor outcomes. Recent genetic and molecular classification of DLBCL has advanced our knowledge of disease biology, yet were not designed to predict early events and guide anticipatory selection of novel therapies. To address this unmet need, we used an integrative multiomic approach to identify a signature at diagnosis that will identify DLBCL at high risk of early clinical failure. PATIENTS AND METHODS Tumor biopsies from 444 newly diagnosed DLBCL were analyzed by WES and RNAseq. A combination of weighted gene correlation network analysis and differential gene expression analysis followed by integration with clinical and genomic data was used to identify a multiomic signature associated with high risk of early clinical failure. RESULTS Current DLBCL classifiers are unable to discriminate cases who fail EFS24. We identified a high risk RNA signature that had a hazard ratio (HR, 18.46 [95% CI 6.51-52.31] P < .001) in a univariate model, which did not attenuate after adjustment for age, IPI and COO (HR, 20.8 [95% CI, 7.14-61.09] P < .001). Further analysis revealed the signature was associated with metabolic reprogramming and a depleted immune microenvironment. Finally, WES data was integrated into the signature and we found that inclusion of ARID1A mutations resulted in identification of 45% of cases with an early clinical failure which was validated in external DLBCL cohorts. CONCLUSION This novel and integrative approach is the first to identify a signature at diagnosis that will identify DLBCL at high risk for early clinical failure and may have significant implications for design of therapeutic options.
Collapse
|
5
|
Rodríguez M, Alonso‐Alonso R, Fernández‐Miranda I, Mondéjar R, Cereceda L, Tráscasa Á, Antonio‐Da Conceiçao A, Borregón J, Gato L, Tomás‐Roca L, Bárcena C, Iglesias B, Climent F, González‐Barca E, Camacho FI, Mayordomo É, Olmedilla G, Gómez‐Prieto P, Castro Y, Serrano‐López J, Sánchez‐García J, Montes‐Moreno S, García‐Cosío M, Martín‐Acosta P, García JF, Planelles M, Quero C, Provencio M, Mahíllo‐Fernández I, Rodríguez‐Pinilla SM, Derenzini E, Pileri S, Sánchez‐Beato M, Córdoba R, Piris MA. An integrated prognostic model for diffuse large B-cell lymphoma treated with immunochemotherapy. EJHAEM 2022; 3:722-733. [PMID: 36051055 PMCID: PMC9422037 DOI: 10.1002/jha2.457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL), the most frequent non-Hodgkin's lymphoma subtype, is characterized by strong biological, morphological, and clinical heterogeneity, but patients are treated with immunochemotherapy in a relatively homogeneous way. Here, we have used a customized NanoString platform to analyze a series of 197 homogeneously treated DLBCL cases. The platform includes the most relevant genes or signatures known to be useful for predicting response to R-CHOP (Rituximab, Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone) in DLBCL cases. We generated a risk score that combines the International Prognostic Index with cell of origin and double expression of MYC/BCL2, and stratified the series into three groups, yielding hazard ratios from 0.15 to 5.49 for overall survival, and from 0.17 to 5.04 for progression-free survival. Group differences were highly significant (p < 0.0001), and the scoring system was applicable to younger patients (<60 years of age) and patients with advanced or localized stages of the disease. Results were validated in an independent dataset from 166 DLBCL patients treated in two distinct clinical trials. This risk score combines clinical and biological data in a model that can be used to integrate biological variables into the prognostic models for DLBCL cases.
Collapse
|
6
|
Dlouhy I, Karube K, Enjuanes A, Salaverria I, Nadeu F, Ramis-Zaldivar JE, Valero JG, Rivas-Delgado A, Magnano L, Martin-García D, Pérez-Galán P, Clot G, Rovira J, Jares P, Balagué O, Giné E, Mozas P, Briones J, Sancho JM, Salar A, Mercadal S, Alcoceba M, Valera A, Campo E, López-Guillermo A. Revised International Prognostic Index and genetic alterations are associated with early failure to R-CHOP in patients with diffuse large B-cell lymphoma. Br J Haematol 2021; 196:589-598. [PMID: 34632572 DOI: 10.1111/bjh.17858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/26/2021] [Accepted: 09/12/2021] [Indexed: 11/28/2022]
Abstract
Relapsed or refractory diffuse large B-cell lymphoma (DLBCL) cases have a poor outcome. Here we analysed clinico-biological features in 373 DLBCL patients homogeneously treated with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R-CHOP), in order to identify variables associated with early failure to treatment (EF), defined as primary refractoriness or relapse within 12 months from diagnosis. In addition to clinical features, mutational status of 106 genes was studied by targeted next-generation sequencing in 111 cases, copy number alterations in 87, and gene expression profile (GEP) in 39. Ninety-seven cases (26%) were identified as EF and showed significantly shorter overall survival (OS). Patients with B symptoms, advanced stage, high levels of serum lactate dehydrogenase (LDH) or β2-microglobulin, low lymphocyte/monocyte ratio and higher Revised International Prognostic Index (R-IPI) scores, as well as those with BCL2 rearrangements more frequently showed EF, with R-IPI being the most important in logistic regression. Mutations in NOTCH2, gains in 5p15·33 (TERT), 12q13 (CDK2), 12q14·1 (CDK4) and 12q15 (MDM2) showed predictive importance for EF independently from R-IPI. GEP studies showed that EF cases were significantly enriched in sets related to cell cycle regulation and inflammatory response, while cases in response showed over-representation of gene sets related to extra-cellular matrix and tumour microenvironment.
Collapse
Affiliation(s)
- Ivan Dlouhy
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain
| | - Kennosuke Karube
- Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Cell Biology & Pathology Department, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | - Anna Enjuanes
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Itziar Salaverria
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ferran Nadeu
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Juan Enric Ramis-Zaldivar
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Juan G Valero
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alfredo Rivas-Delgado
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain
| | - Laura Magnano
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain
| | - David Martin-García
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Patricia Pérez-Galán
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Guillem Clot
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordina Rovira
- Department of Hematology, Hospital Clínic, Barcelona, Spain
| | - Pedro Jares
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Olga Balagué
- Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Eva Giné
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain
| | - Pablo Mozas
- Department of Hematology, Hospital Clínic, Barcelona, Spain
| | | | | | | | | | - Miguel Alcoceba
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Hospital Clínico Universitario, Salamanca, Spain
| | - Alexandra Valera
- Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elías Campo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,University of Barcelona, Barcelona, Spain
| | - Armando López-Guillermo
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,University of Barcelona, Barcelona, Spain
| |
Collapse
|
7
|
Huang Y, Tu WL, Yao YQ, Cai YL, Ma LP. Construction of a Novel Gene-Based Model for Survival Prediction of Hepatitis B Virus Carriers With HCC Development. Front Genet 2021; 12:720888. [PMID: 34531900 PMCID: PMC8439286 DOI: 10.3389/fgene.2021.720888] [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: 06/09/2021] [Accepted: 07/30/2021] [Indexed: 11/14/2022] Open
Abstract
Despite the effectiveness of hepatitis B virus (HBV) vaccination in reducing the prevalence of chronic HBV infection as well as the incidence of acute hepatitis B, fulminant hepatitis, liver cirrhosis and hepatocellular carcinoma (HCC), there was still a large crowd of chronically infected populations at risk of developing cirrhosis or HCC. In this study, we established a comprehensive prognostic system covering multiple signatures to elevate the predictive accuracy for overall survival (OS) of hepatitis B virus carriers with HCC development. Weighted Gene Co-Expression Network Analysis (WGCNA), Least Absolute Shrinkage and Selection Operator (LASSO), Support Vector Machine Recursive Feature Elimination (SVM-RFE), and multivariate COX analysis, along with a suite of other online analyses were successfully applied to filtrate a three-gene signature model (TP53, CFL1, and UBA1). Afterward, the gene-based risk score was calculated based on the Cox coefficient of the individual gene, and the prognostic power was assessed by time-dependent receiver operating characteristic (tROC) and Kaplan–Meier (KM) survival analysis. Furthermore, the predictive power of the nomogram, integrated with the risk score and clinical parameters (age at diagnosis and TNM stage), was revealed by the calibration plot and tROC curves, which was verified in the validation set. Taken together, our study may be more effective in guiding the clinical decision-making of personalized treatment for HBV carriers.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Biochemistry and Molecular Biology, School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Wen-Ling Tu
- Department of Genetics, School of Bioscience and Technology, Chengdu Medical College, Chengdu, China.,The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Yan-Qiu Yao
- Department of Biochemistry and Molecular Biology, School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Ye-Ling Cai
- Department of Biochemistry and Molecular Biology, School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Li-Ping Ma
- Department of Biochemistry and Molecular Biology, School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
8
|
Psaras AM, Chang KT, Hao T, Brooks TA. Targeted Downregulation of MYC through G-quadruplex Stabilization by DNAi. Molecules 2021; 26:5542. [PMID: 34577013 PMCID: PMC8464964 DOI: 10.3390/molecules26185542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/12/2022] Open
Abstract
Modulating the expression or function of the enigmatic MYC protein has demonstrated efficacy in an array of cancer types and a marked potential therapeutic index and safety profile. Despite its high therapeutic value, specific and selective inhibitors or downregulating therapeutics have proven difficult to develop. In the current study, we expanded our work on a MYC promoter G-quadruplex (G4) stabilizing DNA clamp to develop an oligonucleotide interfering DNA (DNAi) therapeutic. We explored six DNAi for G4-stabilization through EMSA, DMS footprinting, and thermal stability studies, focusing on the DNAi 5T as the lead therapeutic. 5T, but not its scramble control 5Tscr, was then shown to enter the nucleus, modulate cell viability, and decrease MYC expression through G4-stabilization. DNAi 5T is thus described to be our lead DNAi, targeting MYC regulation through stabilization of the higher-order DNA G4 structure in the proximal promoter, and it is poised for further preclinical development as an anticancer therapeutic.
Collapse
Affiliation(s)
- Alexandra Maria Psaras
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA; (A.M.P.); (K.T.C.)
| | - Katarina T. Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA; (A.M.P.); (K.T.C.)
| | - Taisen Hao
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| | - Tracy A. Brooks
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA; (A.M.P.); (K.T.C.)
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| |
Collapse
|
9
|
Ennishi D. The biology of the tumor microenvironment in DLBCL: Targeting the "don't eat me" signal. J Clin Exp Hematop 2021; 61:210-215. [PMID: 34511583 PMCID: PMC8808113 DOI: 10.3960/jslrt.21015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common type of malignant lymphoma with biologically and clinically heterogeneous features. Recently, the tumor microenvironment of this disease has been recognized as an important biological aspect of tumor development and therapeutic targets. Recurrent genetic alterations play significant roles in immune recognition of lymphoma cells. In particular, novel genetic alterations promoting phagocytosis were identified, suggesting a potential therapeutic strategy targeting the “don’t eat me” signal.
Collapse
Affiliation(s)
- Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
10
|
Iqbal M, Castano YG, Paludo J, Rosenthal A, Li Z, Beltran M, Moustafa MA, Inwards D, Porrata L, Micallef I, Bisneto JCV, Johnston P, Ansell SM, Reeder C, Murthy H, Roy V, Foran J, Tun HW, Kharfan-Dabaja MA, Ayala E. Impact of Cell of Origin on Outcomes After Autologous Hematopoietic Cell Transplant in Diffuse Large B-Cell Lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:e89-e95. [PMID: 34593359 DOI: 10.1016/j.clml.2021.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 11/30/2022]
Abstract
Germinal center B-cell-like diffuse large B cell lymphoma (GCB-DLBCL) at diagnosis is associated with superior long-term outcomes compared to non-GCB-DLBCL in patients treated with conventional chemo-immunotherapy. Whether cell of origin (COO) by Hans algorithm retains its prognostic significance in patients with (R/R) relapsed/refractory DLBCL undergoing autologous hematopoietic cell transplant (auto-HCT) is not well established. Three hundred and fifty-seven patients underwent auto-HCT between 2005 and 2018. The COO status was determined in 284 patients and these were included in the analysis. One hundred ninety-four patients had GCB-DLBCL while 90 had non-GCB-DLBCL. Median follow up was 1.7 (0-13) years. The GCB-DLBCL was associated with inferior 5-year overall survival at 44% (95%CI, 36-52) versus 64% (95%CI, 54-77) (P = .004) and a higher relapse incidence at 67% (95%CI, 58-74) versus 49% (95%CI, 35-60) (P = .01) in the non-GCB-DLBCL. The difference between GCB and non-GCB-DLBCL remained statistically significant in multivariate analysis. Additionally, response at the time of transplant was an independent prognostic factor. GCB-DLBCL was enriched in double-hit and triple hit phenotype based on available fluorescence in situ hybridization data. These results suggest an enrichment of high-risk genetic rearrangements in R/R GCB-DLBCL resulting in limited efficacy of auto-HCT.
Collapse
Affiliation(s)
- Madiha Iqbal
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL.
| | | | - Jonas Paludo
- Division of Hematology-Oncology, Mayo Clinic, Rochester, MN
| | | | - Zhuo Li
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, FL
| | | | - Muhamad Alhaj Moustafa
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | - David Inwards
- Division of Hematology-Oncology, Mayo Clinic, Rochester, MN
| | - Luis Porrata
- Division of Hematology-Oncology, Mayo Clinic, Rochester, MN
| | - Ivana Micallef
- Division of Hematology-Oncology, Mayo Clinic, Rochester, MN
| | | | | | | | - Craig Reeder
- Division of Hematology-Oncology, Mayo Clinic, Phoenix, AZ
| | - Hemant Murthy
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | - Vivek Roy
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | - James Foran
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | - Han W Tun
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| | - Ernesto Ayala
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL
| |
Collapse
|
11
|
Papageorgiou SG, Thomopoulos TP, Katagas I, Bouchla A, Pappa V. Prognostic molecular biomarkers in diffuse large B-cell lymphoma in the rituximab era and their therapeutic implications. Ther Adv Hematol 2021; 12:20406207211013987. [PMID: 34104369 PMCID: PMC8150462 DOI: 10.1177/20406207211013987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) represents a group of tumors characterized by substantial heterogeneity in terms of their pathological and biological features, a causal factor of their varied clinical outcome. This variation has persisted despite the implementation of rituximab in treatment regimens over the last 20 years. In this context, prognostic biomarkers are of great importance in order to identify high-risk patients that might benefit from treatment intensification or the introduction of novel therapeutic agents. Herein, we review current knowledge on specific immunohistochemical or genetic biomarkers that might be useful in clinical practice. Gene-expression profiling is a tool of special consideration in this effort, as it has enriched our understanding of DLBCL biology and has allowed for the classification of DLBCL by cell-of-origin as well as by more elaborate molecular signatures based on distinct gene-expression profiles. These subgroups might outperform individual biomarkers in terms of prognostication; however, their use in clinical practice is still limited. Moreover, the underappreciated role of the tumor microenvironment in DLBCL prognosis is discussed in terms of prognostic gene-expression signatures, as well as in terms of individual biomarkers of prognostic significance. Finally, the efficacy of novel therapeutic agents for the treatment of DLBCL patients are discussed and an evidence-based therapeutic approach by specific genetic subgroup is suggested.
Collapse
Affiliation(s)
- Sotirios G. Papageorgiou
- Second Department of Internal Medicine and Research Unit, University General Hospital ‘Attikon’, 1 Rimini Street, Haidari, Athens 12462, Greece
| | - Thomas P. Thomopoulos
- Second Department of Internal Medicine and Research Unit, Hematology Unit, University General Hospital, ‘Attikon’, Haidari, Athens, Greece
| | - Ioannis Katagas
- Second Department of Internal Medicine and Research Unit, Hematology Unit, University General Hospital, ‘Attikon’, Haidari, Athens, Greece
| | - Anthi Bouchla
- Second Department of Internal Medicine and Research Unit, Hematology Unit, University General Hospital, ‘Attikon’, Haidari, Athens, Greece
| | - Vassiliki Pappa
- Second Department of Internal Medicine and Research Unit, Hematology Unit, University General Hospital, ‘Attikon’, Haidari, Athens, Greece
| |
Collapse
|
12
|
Cuadros M, García DJ, Andrades A, Arenas AM, Coira IF, Baliñas-Gavira C, Peinado P, Rodríguez MI, Álvarez-Pérez JC, Ruiz-Cabello F, Camós M, Jiménez-Velasco A, Medina PP. LncRNA-mRNA Co-Expression Analysis Identifies AL133346.1/CCN2 as Biomarkers in Pediatric B-Cell Acute Lymphoblastic Leukemia. Cancers (Basel) 2020; 12:cancers12123803. [PMID: 33348573 PMCID: PMC7765782 DOI: 10.3390/cancers12123803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Dysregulation of noncoding RNAs has been described in numerous types of cancers and it has been associated with oncogenic or tumor suppressor activities. However, the signature of clinically relevant noncoding RNAs in pediatric B-cell acute lymphoblastic leukemia is still poorly understood. In a search for long non-coding RNAs that characterize pediatric B-cell acute lymphoblastic leukemia, we found that the long non-coding RNA AL133346.1 and a neighbouring protein-coding mRNA (CCN2) were significantly over-expressed in leukemia samples compared to healthy bone marrow. Survival analysis showed that patients with high CCN2 expression had a significantly better prognosis. These data suggest that AL133346.1/CCN2 could be useful for discriminating subtypes of leukemia and that CCN2 expression could predict the prognosis of pediatric patients with B-cell acute lymphoblastic leukemia. Abstract Pediatric acute B-cell lymphoblastic leukemia (B-ALL) constitutes a heterogeneous and aggressive neoplasia in which new targeted therapies are required. Long non-coding RNAs have recently emerged as promising disease-specific biomarkers for the clinic. Here, we identified pediatric B-ALL-specific lncRNAs and associated mRNAs by comparing the transcriptomic signatures of tumoral and non-tumoral samples. We identified 48 lncRNAs that were differentially expressed between pediatric B-ALL and healthy bone marrow samples. The most relevant lncRNA/mRNA pair was AL133346.1/CCN2 (previously known as RP11-69I8.3/CTGF), whose expression was positively correlated and increased in B-ALL samples. Their differential expression pattern and their strong correlation were validated in external B-ALL datasets (Therapeutically Applicable Research to Generate Effective Treatments, Cancer Cell Line Encyclopedia). Survival curve analysis demonstrated that patients with “high” expression levels of CCN2 had higher overall survival than those with “low” levels (p = 0.042), and this gene might be an independent prognostic biomarker in pediatric B-ALL. These findings provide one of the first detailed descriptions of lncRNA expression profiles in pediatric B-ALL and indicate that these potential biomarkers could help in the classification of leukemia subtypes and that CCN2 expression could predict the survival outcome of pediatric B-cell acute lymphoblastic leukemia patients.
Collapse
Affiliation(s)
- Marta Cuadros
- Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, Av. de la Investigación 11, 18016 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (ibs. Granada), Av. Fuerzas Armadas 2, 18014 Granada, Spain
| | - Daniel J García
- Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, Av. de la Investigación 11, 18016 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
| | - Alvaro Andrades
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (ibs. Granada), Av. Fuerzas Armadas 2, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Av. de Fuente Nueva S/N, 18071 Granada, Spain
| | - Alberto M Arenas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Av. de Fuente Nueva S/N, 18071 Granada, Spain
| | - Isabel F Coira
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Av. de Fuente Nueva S/N, 18071 Granada, Spain
| | - Carlos Baliñas-Gavira
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (ibs. Granada), Av. Fuerzas Armadas 2, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Av. de Fuente Nueva S/N, 18071 Granada, Spain
| | - Paola Peinado
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (ibs. Granada), Av. Fuerzas Armadas 2, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Av. de Fuente Nueva S/N, 18071 Granada, Spain
| | - María I Rodríguez
- Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, Av. de la Investigación 11, 18016 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (ibs. Granada), Av. Fuerzas Armadas 2, 18014 Granada, Spain
| | - Juan Carlos Álvarez-Pérez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (ibs. Granada), Av. Fuerzas Armadas 2, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Av. de Fuente Nueva S/N, 18071 Granada, Spain
| | - Francisco Ruiz-Cabello
- Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, Av. de la Investigación 11, 18016 Granada, Spain
- Department of Clinical Analysis and Immunology, UGC Laboratorio Clínico, University Hospital Virgen de las Nieves, 18014 Granada, Spain
| | - Mireia Camós
- Hematology Laboratory, Institut de Recerca Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Leukemia and Other Pediatric Hemopathies, Developmental Tumors Biology Group, Institut de Recerca Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Antonio Jiménez-Velasco
- Hematology Laboratory, Universitary Regional Hospital, Av. de Carlos Haya, 29010 Málaga, Spain
| | - Pedro P Medina
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración 114, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria (ibs. Granada), Av. Fuerzas Armadas 2, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, Av. de Fuente Nueva S/N, 18071 Granada, Spain
| |
Collapse
|
13
|
Carr M, Mamand S, Chapman KL, Perrior T, Wagner SD. IKKε and TBK1 in diffuse large B-cell lymphoma: A possible mechanism of action of an IKKε/TBK1 inhibitor to repress NF-κB and IL-10 signalling. J Cell Mol Med 2020; 24:11573-11582. [PMID: 32858764 PMCID: PMC7576278 DOI: 10.1111/jcmm.15774] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/29/2020] [Accepted: 08/02/2020] [Indexed: 12/14/2022] Open
Abstract
The IKK‐related kinases, IKKε and TBK1, have essential roles in innate immunity in part through modifying MYD88 signalling from the Toll‐like receptors to regulate NF‐κB signalling. We investigated the expression and function of IKKε and TBK1, in diffuse large B‐cell lymphoma (DLBCL). DLBCL cell lines and patient‐derived xenografts were used to determine their sensitivity to IKKε and TBK1 inhibitors. To understand the function of IKKε and TBK1 secreted factors were determined following administration of inhibitors. Gene expression microarrays were used to determine the transcriptional effects of inhibitors. Higher TBK1 mRNA levels associated with poorer clinical outcome but IKKε and TBK1 were expressed in both germinal centre and non‐germinal centre types of DLBCL. Survival of cell lines Ly10, Ly03 and Pfeiffer, and of some primary human lymphoma cells, was suppressed by a small molecule IKKε/TBK1 inhibitor, DMX3433. DMX3433 reduced IL‐10 production from Ly10 and repressed NF‐κB mediated transcription. Inhibition of IKKε and TBK1 warrants further investigation as a potential therapeutic route to suppress NF‐κB signalling in lymphoma.
Collapse
Affiliation(s)
- Matthew Carr
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematological Research Unit, University of Leicester, Leicester, UK
| | - Sami Mamand
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematological Research Unit, University of Leicester, Leicester, UK
| | | | - Trevor Perrior
- Domainex Ltd.,, Chesterford Research Park, Saffron Walden, UK
| | - Simon D Wagner
- Leicester Cancer Research Centre and Ernest and Helen Scott Haematological Research Unit, University of Leicester, Leicester, UK
| |
Collapse
|
14
|
Wang X, Waschke BC, Woolaver RA, Chen SMY, Chen Z, Wang JH. HDAC inhibitors overcome immunotherapy resistance in B-cell lymphoma. Protein Cell 2020; 11:472-482. [PMID: 32162275 PMCID: PMC7305292 DOI: 10.1007/s13238-020-00694-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has been applied successfully to treat B-cell lymphomas in preclinical models or clinical settings. However, immunotherapy resistance is a major challenge for B-cell lymphoma treatment. To overcome this issue, combinatorial therapeutic strategies have been pursued to achieve a better efficacy for treating B-cell lymphomas. One of such strategies is to combine immunotherapy with histone deacetylase (HDAC) inhibitors. HDAC inhibitors can potentially increase tumor immunogenicity, promote anti-tumor immune responses, or reverse immunosuppressive tumor environments. Thus, the combination of HDAC inhibitors and immunotherapy has drawn much attention in current cancer treatment. However, not all HDAC inhibitors are created equal and their net effects are highly dependent on the specific inhibitors used and the HDACs they target. Hence, we suggest that optimal treatment efficacy requires personalized design and rational combination based on prognostic biomarkers and unique profiles of HDAC inhibitors. Here, we discuss the possible mechanisms by which B-cell lymphomas acquire immunotherapy resistance and the effects of HDAC inhibitors on tumor cells and immune cells that could help overcome immunotherapy resistance.
Collapse
Affiliation(s)
- Xiaoguang Wang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Brittany C Waschke
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Rachel A Woolaver
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Samantha M Y Chen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Zhangguo Chen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA
| | - Jing H Wang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Mail Stop 8333, Aurora, CO, 80045, USA.
| |
Collapse
|
15
|
MHC class I-independent activation of virtual memory CD8 T cells induced by chemotherapeutic agent-treated cancer cells. Cell Mol Immunol 2020; 18:723-734. [PMID: 32427883 DOI: 10.1038/s41423-020-0463-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/31/2020] [Accepted: 04/24/2020] [Indexed: 11/09/2022] Open
Abstract
Cancer cells can evade immune recognition by losing major histocompatibility complex (MHC) class I. Hence, MHC class I-negative cancers represent the most challenging cancers to treat. Chemotherapeutic drugs not only directly kill tumors but also modulate the tumor immune microenvironment. However, it remains unknown whether chemotherapy-treated cancer cells can activate CD8 T cells independent of tumor-derived MHC class I and whether such MHC class I-independent CD8 T-cell activation can be exploited for cancer immunotherapy. Here, we showed that chemotherapy-treated cancer cells directly activated CD8 T cells in an MHC class I-independent manner and that these activated CD8 T cells exhibit virtual memory (VM) phenotypes. Consistently, in vivo chemotherapeutic treatment preferentially increased tumor-infiltrating VM CD8 T cells. Mechanistically, MHC class I-independent activation of CD8 T cells requires cell-cell contact and activation of the PI3K pathway. VM CD8 T cells contribute to a superior therapeutic effect on MHC class I-deficient tumors. Using humanized mouse models or primary human CD8 T cells, we also demonstrated that chemotherapy-treated human lymphomas activated VM CD8 T cells independent of tumor-derived MHC class I. In conclusion, CD8 T cells can be directly activated in an MHC class I-independent manner by chemotherapy-treated cancers, and these activated CD8 T cells may be exploited for developing new strategies to treat MHC class I-deficient cancers.
Collapse
|
16
|
Inhibition of Aurora A Kinase in Combination with Chemotherapy Induces Synthetic Lethality and Overcomes Chemoresistance in Myc-Overexpressing Lymphoma. Target Oncol 2020; 14:563-575. [PMID: 31429028 DOI: 10.1007/s11523-019-00662-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Aberrant Myc expression plays a critical role in various tumors, including non-Hodgkin lymphoma (NHL). Myc-positive lymphoma is clinically aggressive, more resistant to chemotherapy, and associated with high mortality. OBJECTIVE The current study aimed to show inhibition of aurora A kinase (AURKA) may overcome resistance to chemotherapy and improve outcomes in Myc-overexpressing lymphoma. METHODS Myc-overexpressing lymphoma cell lines were evaluated by trypan blue, annexin V/propidium iodide staining, and western blotting for cytotoxicity, cell cycle, apoptosis, and Myc-associated protein expression, respectively, in the presence of cyclophosphamide with or without MLN8237, an AURKA inhibitor. Immunofluorescence for apoptosis-inducing factor (AIF) and acridine orange staining were used to analyze levels of autophagy. EµMyc genetically modified mouse model and xenograft models bearing Myc-overexpressing lymphoma cells were used to determine the efficacy of cyclophosphamide, MLN8237, or the combination in chemosensitive and chemoresistant tumors. RESULTS In our in vitro experiments using chemoresistant lymphoma cells, MLN8237 and cyclophosphamide showed synergistic effects. Mice bearing lymphoma xenograft had rapid disease progression with median survival of ~ 35 days when treated with cyclophosphamide alone. In contrast, the combination of cyclophosphamide and MLN8237 induced complete tumor regression in all mice, which led to improvement in survival compared with the single agent control (p = 0.022). Kinome analysis of tumors treated with MLN8237 showed global suppression of various kinases. CONCLUSION Our data demonstrate that AURKA inhibition induces synthetic lethality and overcomes chemoresistance in Myc-overexpressing lymphoma. The combination of MLN8237 and conventional chemotherapy showed promising safety and anti-tumor activities in preclinical models of Myc-positive NHL.
Collapse
|
17
|
Phuoc V, Sandoval-Sus J, Chavez JC. Drug therapy for double-hit lymphoma. Drugs Context 2019; 8:dic-8-2019-8-1. [PMID: 31844420 PMCID: PMC6905641 DOI: 10.7573/dic.2019-8-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 12/19/2022] Open
Abstract
Double-hit lymphoma (DHL) is a rare type of aggressive B-cell lymphoma defined as a high-grade B-cell lymphoma (HGBCL) with the presence of MYC, BCL2 and/or BCL6 rearrangements. Patients usually present with rapidly progressive and advanced stage of disease and, commonly, with extranodal involvement. Typically, patients become refractory to standard R-CHOP, and more aggressive regimens such as DA-EPOCH-R, R-hyperCVAD or CODOX-R regimens are typically needed. MYC is considered an “undruggable” mutation. Recent evidence suggests that pathogenic mechanisms associated with MYC could be potential targets. In this review, we also discuss the role of hematopoietic stem cell transplantation (HCT) and chimeric antigen receptor (CAR) T-cell therapy in DHL. We also discuss the role of potential novel agents such as BCL2 inhibitors, checkpoint inhibitors, bromodomain and extraterminal (BET) family inhibitors, Pi3K inhibitors, and others.
Collapse
Affiliation(s)
- Vania Phuoc
- Division Hematology/Oncology, University of South Florida, Tampa, FL, USA
| | - Jose Sandoval-Sus
- Department of Malignant Hematology and Cellular Therapy at Memorial Healthcare System, Pembroke Pines, FL, USA
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa FL, USA
| |
Collapse
|
18
|
Wang X, Waschke BC, Woolaver RA, Chen Z, Zhang G, Piscopio AD, Liu X, Wang JH. Histone Deacetylase Inhibition Sensitizes PD1 Blockade-Resistant B-cell Lymphomas. Cancer Immunol Res 2019; 7:1318-1331. [PMID: 31235619 PMCID: PMC6679731 DOI: 10.1158/2326-6066.cir-18-0875] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/22/2019] [Accepted: 06/13/2019] [Indexed: 01/05/2023]
Abstract
PD1 blockade is effective in a subset of patients with B-cell lymphoma (e.g., classical-Hodgkin lymphomas); however, most patients do not respond to anti-PD1 therapy. To study PD1 resistance, we used an isoform-selective histone deacetylase inhibitor (HDACi; OKI-179), and a mouse mature B-cell lymphoma, G1XP lymphoma, immunosuppressive features of which resemble those of human B-cell lymphomas, including downregulation of MHC class I and II, exhaustion of CD8+ and CD4+ tumor-infiltrating lymphocytes (TIL), and PD1-blockade resistance. Using two lymphoma models, we show that treatment of B-cell lymphomas refractory to PD1 blockade with both OKI-179 and anti-PD1 inhibited growth; furthermore, sensitivity to single or combined treatment required tumor-derived MHC class I, and positively correlated with MHC class II expression level. We conclude that OKI-179 sensitizes lymphomas to PD1-blockade by enhancing tumor immunogenicity. In addition, we found that different HDACis exhibited distinct effects on tumors and T cells, yet the same HDACi could differentially affect HLA expression on different human B-cell lymphomas. Our study highlights the immunologic effects of HDACis on antitumor responses and suggests that optimal treatment efficacy requires personalized design and rational combination based on prognostic biomarkers (e.g., MHCs) and the individual profiles of HDACi.
Collapse
Affiliation(s)
- Xiaoguang Wang
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Brittany C Waschke
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Rachel A Woolaver
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Zhangguo Chen
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Gan Zhang
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado
| | | | - Xuedong Liu
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado
- OnKure Inc., Boulder, Colorado
| | - Jing H Wang
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
19
|
Wang H, Zhou X, Zhu JW, Ye JN, Guo HF, Sun C. Association of CD117 and HLA-DR expression with shorter overall survival and/or progression-free survival in patients with multiple myeloma treated with bortezomib and thalidomide combination treatment without transplantation. Oncol Lett 2018; 16:5655-5666. [PMID: 30344721 PMCID: PMC6176261 DOI: 10.3892/ol.2018.9365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 06/27/2018] [Indexed: 12/14/2022] Open
Abstract
Certain immunophenotypes in multiple myeloma (MM), including CD56 and CD117, have been reported to be associated with overall survival (OS). However, previous reports have ignored the impact of different treatment regimens and the long-term prognostic value of immunophenotyping in MM when treated with novel agents, including thalidomide and bortezomib, in the absence of transplantation for autologous stem cell transplantation and allo-hematopoietic stem cell transplantation. To further understand the long-term prognostic value of immunophenotyping in MM, when treated with bortezomib combined with thalidomide-based regimens without transplantation, 80 patients who were newly diagnosed between January 2007 and December 2015, were analyzed retrospectively. In contrast to previous studies, no significant survival time difference was observed between CD56+/CD117+ and CD56-/CD117- groups. Multivariate analysis suggested that human leukocyte antigen-antigen D-related (HLA-DR)+ was independently associated with shorter OS and progression-free survival (PFS), while CD117+ was an independent prognostic factor for decreased PFS. In addition, the myeloma prognostic index (MPI), defined by HLA-DR+, age ≥65 years and international staging system stage III, was suitable for risk stratification of patients treated with novel agents for OS and PFS. The results of the current study suggested that HLA-DR+ patients had a shorter OS and PFS and CD117+ patients had shorter PFS. HLA-DR+ or CD117+ was sufficient to affect survival. Evaluating these markers may reveal valuable prognostic factors for MM in patients receiving bortezomib combined with thalidomide-based regimens without autologous stem cell transplantation and allo-hematopoietic stem cell transplantation). MPI may describe an accessible tool to predict the prognosis of patients with MM.
Collapse
Affiliation(s)
- Huan Wang
- Department of Hematology, Affiliated Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Xin Zhou
- Department of Hematology, Affiliated Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jian-Wei Zhu
- Department of Hematology, Affiliated Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jian-Nan Ye
- Department of Hematology, Affiliated Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Hong-Feng Guo
- Department of Hematology, Affiliated Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Chao Sun
- Department of Hematology, Affiliated Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| |
Collapse
|
20
|
Prognostic significances of overexpression MYC and/or BCL2 in R-CHOP-treated diffuse large B-cell lymphoma: A Systematic review and meta-analysis. Sci Rep 2018; 8:6267. [PMID: 29674626 PMCID: PMC5908914 DOI: 10.1038/s41598-018-24631-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 04/05/2018] [Indexed: 12/31/2022] Open
Abstract
Numerous studies have investigated the prognostic values of MYC and/or BCL2 protein overexpression in diffuse large B-cell lymphoma (DLBCL). However, the results still demonstrate discrepancies among different studies. We aimed to do a systematic review and meta-analysis on the relationships between overexpression MYC and/or BCL2 and DLBCLs treated with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP). This study followed the guidelines of PRISMA and Cochrane handbook. The hazard ratios (HRs) for overall survival (OS) were pooled to estimate the main effect size. Twenty studies recruited a total of 5576 patients were available for this meta-analysis. The results showed that MYC (HR = 1.96, 95%CI (confidence interval) = 1.69–2.27)without heterogeneity(I2 = 17.2%, P = 0.280), BCL2 (HR = 1.65, 95%CI = 1.43–1.89, I2 = 20.7%, P = 0.234) protein overexpression, and co-overexpression (HR = 2.58, 95%CI = 2.19–3.04, I2 = 17.2%, P = 0.275) had a poor prognosis in R-CHOP treated DLBCL patients, respectively. The current analysis indicated that MYC and/or BCL2 protein overexpression, and particularly co-overexpression was related to short overall survival in R-CHOP treated DLBCL patients, showing that application of the two new biomarkers can help to better stratify DLBCL patients and guide targeted treatment.
Collapse
|
21
|
Wang J, Zhou M, Zhang QG, Xu J, Lin T, Zhou RF, Li J, Yang YG, Chen B, Ouyang J. Prognostic value of expression of nuclear factor kappa-B/p65 in non-GCB DLBCL patients. Oncotarget 2018; 8:9708-9716. [PMID: 28039454 PMCID: PMC5354765 DOI: 10.18632/oncotarget.14182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/22/2016] [Indexed: 12/12/2022] Open
Abstract
Purpose We estimated the expression of nuclear factor kappa B/p65 in non-germinal center B-cell-like subtype diffuse large B-cell lymphoma, to investigate its relationship to clinicopathological features, and to further evaluate its prognostic value and clarify its impact on survival. Results Among the 49 patients enrolled in this study, 14 (28.6%) had positive p65 expression. The negative p65 group had significantly better survival compared to the positive p65 group in terms of both the 3-year estimated OS (91.2% vs. 39.3%, p = 0.003) and PFS (75.6% vs. 26.5%, p = 0.002). In patients with 4 or more risk factors, p65 was an independent prognostic factor of OS (HR 5.99, 95%CI=1.39-25.75, p=0.016) and PFS (HR 4.01, 95%CI=1.15-14.00, p=0.029). Materials and Methods The expression of the NF-κB/p65 protein was deteremined by immunohistochemistry in 49 non-GCB DLBCL. Survival was assessed by the Kaplan–Meier method and Cox multivariate analysis. The median patient follow-up period was 24 months. Conclusions The expression of NF-κB/p65 has prognostic value in high risk non-GCB DLBCL, and it is a suitable target for the development of new therapies.
Collapse
Affiliation(s)
- Jing Wang
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| | - Min Zhou
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| | - Qi-Guo Zhang
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| | - Jingyan Xu
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| | - Tong Lin
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| | - Rong-Fu Zhou
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| | - Juan Li
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| | - Yong-Gong Yang
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| | - Bing Chen
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| | - Jian Ouyang
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, PR China
| |
Collapse
|
22
|
Chadly DM, Best J, Ran C, Bruska M, Woźniak W, Kempisty B, Schwartz M, LaFleur B, Kerns BJ, Kessler JA, Matsuoka AJ. Developmental profiling of microRNAs in the human embryonic inner ear. PLoS One 2018; 13:e0191452. [PMID: 29373586 PMCID: PMC5786302 DOI: 10.1371/journal.pone.0191452] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/04/2018] [Indexed: 12/12/2022] Open
Abstract
Due to the extreme inaccessibility of fetal human inner ear tissue, defining of the microRNAs (miRNAs) that regulate development of the inner ear has relied on animal tissue. In the present study, we performed the first miRNA sequencing of otic precursors in human specimens. Using HTG miRNA Whole Transcriptome assays, we examined miRNA expression in the cochleovestibular ganglion (CVG), neural crest (NC), and otic vesicle (OV) from paraffin embedded (FFPE) human specimens in the Carnegie developmental stages 13-15. We found that in human embryonic tissues, there are different patterns of miRNA expression in the CVG, NC and OV. In particular, members of the miR-183 family (miR-96, miR-182, and miR-183) are differentially expressed in the CVG compared to NC and OV at Carnegie developmental stage 13. We further identified transcription factors that are differentially targeted in the CVG compared to the other tissues from stages 13-15, and we performed gene set enrichment analyses to determine differentially regulated pathways that are relevant to CVG development in humans. These findings not only provide insight into the mechanisms governing the development of the human inner ear, but also identify potential signaling pathways for promoting regeneration of the spiral ganglion and other components of the inner ear.
Collapse
Affiliation(s)
- Duncan M. Chadly
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Jennifer Best
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Cong Ran
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | | | - Witold Woźniak
- Department of Anatomy, Poznań University, Poznań, Poland
| | | | - Mark Schwartz
- HTG Molecular Diagnostics, Inc., Tucson, Arizona, United States of America
| | - Bonnie LaFleur
- HTG Molecular Diagnostics, Inc., Tucson, Arizona, United States of America
| | - B. J. Kerns
- HTG Molecular Diagnostics, Inc., Tucson, Arizona, United States of America
| | - John A. Kessler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Akihiro J. Matsuoka
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Department of Communication Sciences and Disorders, Northwestern University, Evanston, Illinois, United States of America
- Hugh Knowles Center for Hearing Research, Northwestern University, Evanston, Illinois, United States of America
| |
Collapse
|
23
|
Wong KK, Gascoyne DM, Soilleux EJ, Lyne L, Spearman H, Roncador G, Pedersen LM, Møller MB, Green TM, Banham AH. FOXP2-positive diffuse large B-cell lymphomas exhibit a poor response to R-CHOP therapy and distinct biological signatures. Oncotarget 2018; 7:52940-52956. [PMID: 27224915 PMCID: PMC5288160 DOI: 10.18632/oncotarget.9507] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 05/04/2016] [Indexed: 01/24/2023] Open
Abstract
FOXP2 shares partially overlapping normal tissue expression and functionality with FOXP1; an established diffuse large B-cell lymphoma (DLBCL) oncogene and marker of poor prognosis. FOXP2 is expressed in the plasma cell malignancy multiple myeloma but has not been studied in DLBCL, where a poor prognosis activated B-cell (ABC)-like subtype display partially blocked plasma cell differentiation. FOXP2 protein expression was detected in ABC-DLBCL cell lines, and in primary DLBCL samples tumoral FOXP2 protein expression was detected in both germinal center B-cell-like (GCB) and non-GCB DLBCL. In biopsies from DLBCL patients treated with immunochemotherapy (R-CHOP), ≥ 20% nuclear tumoral FOXP2-positivity (n = 24/158) correlated with significantly inferior overall survival (OS: P = 0.0017) and progression-free survival (PFS: P = 0.0096). This remained significant in multivariate analysis against either the international prognostic index score or the non-GCB DLBCL phenotype (P < 0.05 for both OS and PFS). Expression of BLIMP1, a marker of plasmacytic differentiation that is commonly inactivated in ABC-DLBCL, did not correlate with patient outcome or FOXP2 expression in this series. Increased frequency of FOXP2 expression significantly correlated with FOXP1-positivity (P = 0.0187), and FOXP1 co-immunoprecipitated FOXP2 from ABC-DLBCL cells indicating that these proteins can co-localize in a multi-protein complex. FOXP2-positive DLBCL had reduced expression of HIP1R (P = 0.0348), which is directly repressed by FOXP1, and exhibited distinct patterns of gene expression. Specifically in ABC-DLBCL these were associated with lower expression of immune response and T-cell receptor signaling pathways. Further studies are warranted to investigate the potential functional cooperativity between FOXP1 and FOXP2 in repressing immune responses during the pathogenesis of high-risk DLBCL.
Collapse
Affiliation(s)
- Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Duncan M Gascoyne
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Elizabeth J Soilleux
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Linden Lyne
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Hayley Spearman
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Giovanna Roncador
- Monoclonal Antibody Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Lars M Pedersen
- Department of Haematology, Roskilde Hospital, Roskilde, Denmark
| | - Michael B Møller
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Tina M Green
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Alison H Banham
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
24
|
Toledano MN, Desbordes P, Banjar A, Gardin I, Vera P, Ruminy P, Jardin F, Tilly H, Becker S. Combination of baseline FDG PET/CT total metabolic tumour volume and gene expression profile have a robust predictive value in patients with diffuse large B-cell lymphoma. Eur J Nucl Med Mol Imaging 2018; 45:680-688. [PMID: 29344718 DOI: 10.1007/s00259-017-3907-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 12/03/2017] [Indexed: 01/12/2023]
Abstract
PURPOSE This study evaluated the predictive significance of total metabolic tumour volume (TMTV) measured on baseline FDG PET/CT and its value in addition to gene expression profiling using a new method of gene analysis (rapid reverse transcriptase multiplex ligation-dependent probe amplification assay, RT-MLPA) in patients with diffuse large B-cell lymphoma treated with R-CHOP or R-CHOP-like chemotherapies. METHODS The analysis included 114 patients. TMTV was measured using a 41% SUVmax threshold and tumours were classified into GCB or ABC subtypes according to the RT-MLPA assay. RESULTS The median follow-up was 40 months. the 5-year progression-free survival (PFS) was 54% and the 5-year overall survival (OS) was 62%. The optimal TMTV cut-off value was 261 cm3. In 59 patients with a high TMTV the 5-year PFS and OS were 37% and 39%, respectively, in comparison with 72% and 83%, respectively, in 55 patients with a low TMTV (p = 0.0002 for PFS, p < 0.0001 for OS). ABC status was significantly associated with a worse prognosis. TMTV combined with molecular data identified three groups with very different outcomes: (1) patients with a low TMTV whatever their phenotype (n = 55), (2) patients with a high TMTV and GCB phenotype (n = 33), and (3) patients with a high TMTV and ABC phenotype (n = 26). In the three groups, 5-year PFS rates were 72%, 51% and 17% (p < 0.0001), and 5-year OS rates were 83%, 55% and 17% (p < 0.0001), respectively. In multivariate analysis, TMTV, ABC/GCB phenotype and International Prognostic Index were independent predictive factors for both PFS and OS (p < 0.05 for both). CONCLUSIONS This integrated risk model could lead to more accurate selection of patients that would allow better individualization of therapy.
Collapse
Affiliation(s)
- Mathieu Nessim Toledano
- Nuclear Medicine Department, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France. .,QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France.
| | - P Desbordes
- QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France
| | - A Banjar
- Nuclear Medicine Department, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France.,QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France
| | - I Gardin
- Nuclear Medicine Department, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France.,QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France
| | - P Vera
- Nuclear Medicine Department, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France.,QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France
| | - P Ruminy
- INSERM U918, Centre Henri Becquerel, Rouen, France
| | - F Jardin
- INSERM U918, Centre Henri Becquerel, Rouen, France.,Hematology Department, Centre Henri Becquerel, Rouen, France
| | - H Tilly
- INSERM U918, Centre Henri Becquerel, Rouen, France.,Hematology Department, Centre Henri Becquerel, Rouen, France
| | - S Becker
- Nuclear Medicine Department, Henri Becquerel Cancer Centre and Rouen University Hospital, Rouen, France.,QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France
| |
Collapse
|
25
|
Amin AD, Peters TL, Li L, Rajan SS, Choudhari R, Puvvada SD, Schatz JH. Diffuse large B-cell lymphoma: can genomics improve treatment options for a curable cancer? Cold Spring Harb Mol Case Stud 2017; 3:a001719. [PMID: 28487884 PMCID: PMC5411687 DOI: 10.1101/mcs.a001719] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gene-expression profiling and next-generation sequencing have defined diffuse large B-cell lymphoma (DLBCL), the most common lymphoma diagnosis, as a heterogeneous group of subentities. Despite ongoing explosions of data illuminating disparate pathogenic mechanisms, however, the five-drug chemoimmunotherapy combination R-CHOP remains the frontline standard treatment. This has not changed in 15 years, since the anti-CD20 monoclonal antibody rituximab was added to the CHOP backbone, which first entered use in the 1970s. At least a third of patients are not cured by R-CHOP, and relapsed or refractory DLBCL is fatal in ∼90%. Targeted small-molecule inhibitors against distinct molecular pathways activated in different subgroups of DLBCL have so far translated poorly into the clinic, justifying the ongoing reliance on R-CHOP and other long-established chemotherapy-driven combinations. New drugs and improved identification of biomarkers in real time, however, show potential to change the situation eventually, despite some recent setbacks. Here, we review established and putative molecular drivers of DLBCL identified through large-scale genomics, highlighting among other things the care that must be taken when differentiating drivers from passengers, which is influenced by the promiscuity of activation-induced cytidine deaminase. Furthermore, we discuss why, despite having so much genomic data available, it has been difficult to move toward personalized medicine for this umbrella disorder and some steps that may be taken to hasten the process.
Collapse
Affiliation(s)
- Amit Dipak Amin
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Tara L Peters
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Lingxiao Li
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Soumya Sundara Rajan
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ramesh Choudhari
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Soham D Puvvada
- Department of Medicine, Division of Hematology-Oncology, University of Arizona Comprehensive Cancer Center, Tucson, Arizona 85719, USA
| | - Jonathan H Schatz
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
26
|
Leonard JP, Kolibaba KS, Reeves JA, Tulpule A, Flinn IW, Kolevska T, Robles R, Flowers CR, Collins R, DiBella NJ, Papish SW, Venugopal P, Horodner A, Tabatabai A, Hajdenberg J, Park J, Neuwirth R, Mulligan G, Suryanarayan K, Esseltine DL, de Vos S. Randomized Phase II Study of R-CHOP With or Without Bortezomib in Previously Untreated Patients With Non-Germinal Center B-Cell-Like Diffuse Large B-Cell Lymphoma. J Clin Oncol 2017; 35:3538-3546. [PMID: 28862883 DOI: 10.1200/jco.2017.73.2784] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose To evaluate the impact of the addition of bortezomib to rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) on outcomes in previously untreated patients with non-germinal center B-cell-like (non-GCB) diffuse large B-cell lymphoma (DLBCL). Patients and Methods After real-time determination of non-GCB DLBCL using the Hans immunohistochemistry algorithm, 206 patients were randomly assigned (1:1; stratified by International Prognostic Index [IPI] score) to six 21-day cycles of standard R-CHOP alone or R-CHOP plus bortezomib 1.3 mg/m2 intravenously on days 1 and 4 (VR-CHOP). The primary end point, progression-free survival (PFS), was evaluated in 183 patients with centrally confirmed non-GCB DLBCL who received one or more doses of study drug (91 R-CHOP, 92 VR-CHOP). Results After a median follow-up of 34 months, with 25% (R-CHOP) and 18% (VR-CHOP) of patients having had PFS events, the hazard ratio (HR) for PFS was 0.73 (90% CI, 0.43 to 1.24) with VR-CHOP ( P = .611). Two-year PFS rates were 77.6% with R-CHOP and 82.0% with VR-CHOP; they were 65.1% versus 72.4% in patients with high-intermediate/high IPI (HR, 0.67; 90% CI, 0.34 to 1.29), and 90.0% versus 88.9% (HR, 0.85; 90% CI, 0.35 to 2.10) in patients with low/low-intermediate IPI. Overall response rate with R-CHOP and VR-CHOP was 98% and 96%, respectively. The overall survival HR was 0.75 (90% CI, 0.38 to 1.45); 2-year survival rates were 88.4% and 93.0%, respectively. In the safety population (100 R-CHOP and 101 VR-CHOP patients), grade ≥ 3 adverse events included neutropenia (53% v 49%), thrombocytopenia (13% v 29%), anemia (7% v 15%), leukopenia (26% v 25%), and neuropathy (1% v 5%). Conclusion Outcomes for newly diagnosed, prospectively enrolled patients with non-GCB DLBCL were more favorable than expected with R-CHOP and were not significantly improved by adding bortezomib.
Collapse
Affiliation(s)
- John P Leonard
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Kathryn S Kolibaba
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - James A Reeves
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Anil Tulpule
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Ian W Flinn
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Tatjana Kolevska
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Robert Robles
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Christopher R Flowers
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Robert Collins
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Nicholas J DiBella
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Steven W Papish
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Parameswaran Venugopal
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Andrew Horodner
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Amir Tabatabai
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Julio Hajdenberg
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Jaehong Park
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Rachel Neuwirth
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - George Mulligan
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Kaveri Suryanarayan
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Dixie-Lee Esseltine
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| | - Sven de Vos
- John P. Leonard, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY; Kathryn S. Kolibaba, Compass Oncology, Vancouver, WA; Kathryn S. Kolibaba and Nicholas J. DiBella, US Oncology Research, The Woodlands; Robert Collins, University of Texas Southwestern Medical Center, Dallas, TX; James A. Reeves, Florida Cancer Specialists, Fort Myers; Julio Hajdenberg, University of Florida Health Cancer Center at Orlando Health, Orlando, FL; Anil Tulpule, Keck Medicine of University of Southern California; Sven de Vos, University of California at Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles; Tatjana Kolevska, Kaiser Permanente Medical Center Northern California, Vallejo; Robert Robles, Bay Area Cancer Research Group (Diablo Valley Medical Group), Pleasant Hill; Andrew Horodner, Cancer Care Associates Medical Group, Redondo Beach, CA; Ian W. Flinn, Sarah Cannon Research Institute, Nashville, TN; Christopher R. Flowers, Winship Cancer Institute of Emory University, Atlanta, GA; Nicholas J. DiBella, Rocky Mountain Cancer Centers, Aurora, CO; Steven W. Papish, Summit Medical Group MD Anderson Cancer Center, Camden, NJ; Parameswaran Venugopal, Rush University Medical Center, Chicago, IL; Amir Tabatabai, York Cancer Center/Cancer Care Associates of York, York, PA; and Jaehong Park, Rachel Neuwirth, George Mulligan, Kaveri Suryanarayan, and Dixie-Lee Esseltine, Millennium Pharmaceuticals, Cambridge, MA
| |
Collapse
|
27
|
Davis B, Schwartz M, Duchemin D, Carl Barrett J, Post G. Validation of a Multiplexed Gene Signature Assay for Diagnosis of Canine Cancers from Formalin-Fixed Paraffin-Embedded Tissues. J Vet Intern Med 2017; 31:854-863. [PMID: 28370296 PMCID: PMC5435129 DOI: 10.1111/jvim.14686] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 01/05/2017] [Accepted: 02/07/2017] [Indexed: 01/09/2023] Open
Abstract
Background Use of molecular‐based diagnostics for companion animals is impeded by availability of technology platforms, tissue acquisition requirements, and species‐specific reagents. Hypothesis/Objectives To validate a quantitative nuclease protection assay (qNPA) to simultaneously measure RNA expression of multiple genes in archived formalin‐fixed paraffin‐embedded (FFPE) tumors from dogs. Animals All tumor biopsy samples were collected retrospectively from surgical biopsies and in the care of veterinarians. Methods Retrospective case series. A qNPA 96‐well ArrayPlate was built using 30 canine‐specific genes, 5 housekeeping genes, positive and negative controls with qualified gene‐specific oligonucleotides. Pearson's correlation, coefficient of variation (CV), and multivariate analysis were used to determine analytical performance using 40 FFPE dog tumors. Once validated, 70 FFPE dog tumors were analyzed for differences in gene expression using hierarchical clustering and analysis of variance of log transformed data. Immunohistochemistry (IHC) was performed to correlate gene expression and protein expression in a subset of tumors. Results The assay was linear with decreasing sample input (R2 = 0.978), reproducible within and between 96‐well plates (r = 0.988 and 0.95, respectively) and between different laboratories (CV = 0.96). Hierarchical cluster analysis showed grouping of tumors by histogenesis and oncogenes. Significant differences were found between BCl2, E2F transcription factor 1, MDM2, COX‐2, MET proto‐oncogene receptor kinase, and other biologically relevant gene expression in tumor subtypes. Immunohistochemistry confirmed protein expression. Conclusions and Clinical Implications Because this technology works reliably on FFPE specimens, it can help expedite the broad introduction of multiplexed genomic information for improved diagnostics and discovery of new targets for therapies in veterinary oncology.
Collapse
Affiliation(s)
| | | | - D Duchemin
- Innogenics Inc, Harvard, MA.,Veterinary Cancer Center, Norwalk, CT
| | | | - G Post
- Innogenics Inc, Harvard, MA.,Veterinary Cancer Center, Norwalk, CT
| |
Collapse
|
28
|
Kendrick S, Rimsza LM, Scott DW, Slack GW, Farinha P, Tan KL, Persky D, Puvvada S, Connors JM, Sehn L, Gascoyne RD, Schmelz M. Aberrant cytoplasmic expression of MHCII confers worse progression free survival in diffuse large B-cell lymphoma. Virchows Arch 2016; 470:113-117. [PMID: 27888357 DOI: 10.1007/s00428-016-2041-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/11/2016] [Accepted: 11/07/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Samantha Kendrick
- Department of Pathology, University of Arizona, P.O. Box 24-5043, 1501 N Campbell Ave, Tucson, AZ, 85724, USA
| | - Lisa M Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - David W Scott
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Graham W Slack
- Department of Pathology, British Columbia Cancer Agency, Vancouver, Canada
| | - Pedro Farinha
- Department of Pathology, British Columbia Cancer Agency, Vancouver, Canada
| | - King L Tan
- Department of Pathology, British Columbia Cancer Agency, Vancouver, Canada
| | - Daniel Persky
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Soham Puvvada
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Joseph M Connors
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Laurie Sehn
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Randy D Gascoyne
- Centre for Lymphoid Cancer, British Columbia Cancer Agency, Vancouver, BC, Canada.,Department of Pathology, British Columbia Cancer Agency, Vancouver, Canada
| | - Monika Schmelz
- Department of Pathology, University of Arizona, P.O. Box 24-5043, 1501 N Campbell Ave, Tucson, AZ, 85724, USA.
| |
Collapse
|
29
|
Tada K, Maeshima AM, Hiraoka N, Yamauchi N, Maruyama D, Kim SW, Watanabe T, Katayama N, Heike Y, Tobinai K, Kobayashi Y. Prognostic significance of HLA class I and II expression in patients with diffuse large B cell lymphoma treated with standard chemoimmunotherapy. Cancer Immunol Immunother 2016; 65:1213-22. [PMID: 27522583 PMCID: PMC11029644 DOI: 10.1007/s00262-016-1883-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 08/06/2016] [Indexed: 12/17/2022]
Abstract
Loss of tumor cell human leukocyte antigen (HLA) is an immune escape mechanism for malignancies. However, the effect of low HLA class I or class II expression in diffuse large B cell lymphoma (DLBCL) treated with chemoimmunotherapy with the monoclonal antibody rituximab is largely unknown. We retrospectively analyzed samples and other data from 144 patients with DLBCL who were newly diagnosed in our institution and treated with standard R-CHOP therapy. We used antibodies against pan-HLA class I and pan-HLA class II molecules to assess HLA expression and its effect on prognosis. In a multivariate analysis, loss of HLA class II expression was a significantly independent adverse factor for progression-free survival (PFS; hazard ratio 2.3; 95 % confidence interval 1.2-4.6; P = 0.01). Although HLA class I loss of expression did not correlate with prognosis, the combination of HLA class I(+) with either low peripheral lymphocyte count or CD3(+) lymphocyte count was an adverse prognostic factor for PFS. Loss of HLA class II is an International Prognostic Index (IPI)-independent adverse factor for PFS in patients with DLBCL treated with standard therapy. However, in contrast to other solid cancers, HLA class I loss was not solely a prognostic factor in DLBCL.
Collapse
Affiliation(s)
- Kohei Tada
- Department of Hematology and Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
- Immunotherapy and Cell Therapy Service, St. Luke's International Hospital, Tokyo, Japan
| | - Akiko Miyagi Maeshima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Nobuyoshi Hiraoka
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Nobuhiko Yamauchi
- Department of Hematology and Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Dai Maruyama
- Department of Hematology and Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Sung-Won Kim
- Department of Hematology and Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takashi Watanabe
- Department of Hematology and Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Naoyuki Katayama
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yuji Heike
- Immunotherapy and Cell Therapy Service, St. Luke's International Hospital, Tokyo, Japan
- Immunotherapy Research Field, Translational Research Group, and Translational Medicine Department, Phase 1 Group, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Kensei Tobinai
- Department of Hematology and Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yukio Kobayashi
- Department of Hematology and Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
30
|
Cottereau AS, Lanic H, Mareschal S, Meignan M, Vera P, Tilly H, Jardin F, Becker S. Molecular Profile and FDG-PET/CT Total Metabolic Tumor Volume Improve Risk Classification at Diagnosis for Patients with Diffuse Large B-Cell Lymphoma. Clin Cancer Res 2016; 22:3801-9. [PMID: 26936916 DOI: 10.1158/1078-0432.ccr-15-2825] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/21/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE The prognostic impact of total metabolic tumor volume (TMTV) measured on pretreatment (18)F-FDG PET/CT and its added value to molecular characteristics was investigated in patients with diffuse large B-cell lymphoma (DLBCL). EXPERIMENTAL DESIGN For 81 newly diagnosed patients with DLBCL treated with rituximab and CHOP/CHOP-like regimen, TMTV was computed using the 41% SUVmax thresholding method. According to the gene expression profile, determined using DASL (cDNA-mediated Annealing, Selection, Ligation and extension) technology, a subset of 57 patients was classified in germinal center B (GCB) or activated B-cell (ABC) subtypes and MYC or BCL2 overexpressed. RESULTS Median follow-up was 64 months. Five-year progression-free survival (PFS) and overall survival (OS) were 60% and 63% in the whole population. Median pretherapy TMTV was 320 cm(3) (25th-75th percentiles 106-668 cm(3)). With a 300 cm(3) cutoff, patients with high TMTV (n = 43) had a 5-year PFS and OS of 43% and 46% compared with 76% and 78% for patients with a low TMTV (P = 0.0023, P = 0.0047). ABC status, MYC, or BCL2 overexpression and both overexpression ("dual expressor," DE) were significantly associated with a worse PFS and OS. TMTV combined with molecular data allowed a significant better risk substratification of ABC/GCB patients, on PFS and OS. High TMTV individualized in molecular-low-risk patients a group with a poor outcome (MYC, PFS=51%, OS=55% BCL2, PFS=49%, OS=49% or DE PFS=50%, OS=50%) and a group with a good outcome (MYC, PFS=93%, OS=93% BCL2, PFS=86%, OS=86%, or DE PFS=81%, OS=81%). CONCLUSIONS The combination of molecular and imaging characteristics at diagnosis could lead to a more accurate selection of patients, to increase tailor therapy. Clin Cancer Res; 22(15); 3801-9. ©2016 AACR.
Collapse
Affiliation(s)
- Anne-Ségolène Cottereau
- Nuclear Medicine Department, Henri Becquerel Cancer Center and Rouen University Hospital, Rouen, France. QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France.
| | - Hélène Lanic
- Hematology Department, Centre Henri Becquerel, Rouen, France. UMR INSERM U918, Centre Henri Becquerel, Rouen, France
| | - Sylvain Mareschal
- UMR INSERM U918, Centre Henri Becquerel, Rouen, France. Bioinformatics, University of Rouen, Mont Saint-Aignan, France
| | - Michel Meignan
- Nuclear Medicine Department, Hôpital Henri Mondor, Créteil, France
| | - Pierre Vera
- Nuclear Medicine Department, Henri Becquerel Cancer Center and Rouen University Hospital, Rouen, France. QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France
| | - Hervé Tilly
- Hematology Department, Centre Henri Becquerel, Rouen, France. UMR INSERM U918, Centre Henri Becquerel, Rouen, France
| | - Fabrice Jardin
- Hematology Department, Centre Henri Becquerel, Rouen, France. UMR INSERM U918, Centre Henri Becquerel, Rouen, France
| | - Stéphanie Becker
- Nuclear Medicine Department, Henri Becquerel Cancer Center and Rouen University Hospital, Rouen, France. QuantIF-LITIS (EA 4108-FR CNRS 3638), Faculty of Medicine, University of Rouen, Rouen, France
| |
Collapse
|
31
|
Mondello P, Steiner N, Willenbacher W, Ferrero S, Ghione P, Marabese A, Pitini V, Cuzzocrea S, Mian M. Lenalidomide in Relapsed or Refractory Diffuse Large B-Cell Lymphoma: Is It a Valid Treatment Option? Oncologist 2016; 21:1107-12. [PMID: 27382029 DOI: 10.1634/theoncologist.2016-0103] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 04/05/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Despite the advent of new treatment strategies, many patients with diffuse large B-cell lymphoma (DLBCL) relapse or die of the disease. Prospective clinical trials have demonstrated that lenalidomide is an effective and safe treatment option, especially for non-germinal center B-cell (non-GCB) DLBCL. However, routine clinical data are lacking, which is why we provide the results of the so-far largest relapsed/refractory (R/R) DLBCL real-life analysis. METHODS We retrospectively assessed 123 R/R DLBCL patients who received either 15 or 25 mg/day of lenalidomide from January 2006 to January 2015. RESULTS During a median follow-up period of 4.5 years, complete remission was achieved in 32% and a partial remission in 33% non-GCB patients compared with 0% and 3% in the GCB group (p < .001 and .001, respectively), with median response durations of 15 and 5 months, respectively (p < .001). Lenalidomide at 25 mg was superior to 15 mg in terms of response (complete remission 21% and partial remission 23% vs. 0% and 8%; p = .007 and .05) and median response duration (10 vs. 4 months; p = .03). Toxicity was limited and reversible. Median progression-free survival differed between non-GCB and GCB patients (37 vs. 30 months; p < .001) and between the two dosages (24 vs. 34 months; p = .002). However, overall survival was similar between the subgroups (38-42 months). CONCLUSION We provide evidence that lenalidomide is a valid treatment option for R/R DLBCL, with limited and reversible toxicity, and is more efficient in non-GCB DLBCL and at higher doses. IMPLICATIONS FOR PRACTICE Despite the advent of new treatment strategies, many patients with diffuse large B-cell lymphoma (DLBCL) relapse or die of the disease; hence, novel therapeutic approaches are urgently needed. This study confirms that lenalidomide is a valid and well-tolerated treatment option for relapsed/refractory (R/R) DLBCL. Superior outcomes were observed in non-germinal center B-cell (GCB) DLBCL, probably because of inhibition of the nuclear factor-κB pathway. Similarly, high drug doses resulted in greater clinical benefits. Overall, lenalidomide is a suitable therapeutic option for R/R DLBCL, especially in non-GCB DLBCL, and 25 mg/day dosing should be preferred.
Collapse
Affiliation(s)
- Patrizia Mondello
- Department of Human Pathology, University of Messina, Messina, Italy Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Normann Steiner
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wolfgang Willenbacher
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Simone Ferrero
- Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Paola Ghione
- Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Alessandra Marabese
- Department of Hematology and Center for Bone Marrow Transplantation, Ospedale di Bolzano, Bolzano, Italy
| | - Vincenzo Pitini
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| | - Michael Mian
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria Department of Hematology and Center for Bone Marrow Transplantation, Ospedale di Bolzano, Bolzano, Italy
| |
Collapse
|
32
|
Green TM, Jensen AK, Holst R, Falgreen S, Bøgsted M, de Stricker K, Plesner T, Mourits-Andersen T, Frederiksen M, Johnsen HE, Pedersen LM, Møller MB. Multiplex polymerase chain reaction-based prognostic models in diffuse large B-cell lymphoma patients treated with R-CHOP. Br J Haematol 2016; 174:876-86. [PMID: 27196819 DOI: 10.1111/bjh.14138] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 03/02/2016] [Indexed: 11/30/2022]
Abstract
We present a multiplex analysis for genes known to have prognostic value in an attempt to design a clinically useful classification model in patients with diffuse large B-cell lymphoma (DLBCL). Real-time polymerase chain reaction was used to measure transcript levels of 28 relevant genes in 194 de novo DLBCL patients treated with R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone). Including International Prognostic Index (IPI) as a variable in a penalized Cox regression, we investigated the association with disease progression for single genes or gene combinations in four models. The best model was validated in data from an online available R-CHOP treated cohort. With progression-free survival (PFS) as primary endpoint, the best performing IPI independent model incorporated the LMO2 and HLADQA1 as well as gene interactions for GCSAMxMIB1, GCSAMxCTGF and FOXP1xPDE4B. This model assigned 33% of patients (n = 60) to poor outcome with an estimated 3-year PFS of 40% vs. 87% for low risk (n = 61) and intermediate (n = 60) risk groups (P < 0·001). However, a simpler, IPI independent model incorporated LMO2 and BCL2 and assigned 33% of the patients with a 3-year PFS of 35% vs. 82% for low risk group (P < 0·001). We have documented the impact of a few single genes added to IPI for assignment in new drug trials.
Collapse
Affiliation(s)
- Tina M Green
- Department of Pathology, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Andreas K Jensen
- Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - René Holst
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Steffen Falgreen
- Department of Haematology, Clinical Cancer Research Centre, Aalborg University Hospital, Aalborg, Denmark
| | - Martin Bøgsted
- Department of Haematology, Clinical Cancer Research Centre, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Karin de Stricker
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Torben Plesner
- Department of Haematology, Vejle Hospital, Vejle, Denmark
| | | | - Mikael Frederiksen
- Department of Haematology, Hospital of Southern Denmark, Aabenraa, Denmark
| | - Hans E Johnsen
- Department of Haematology, Clinical Cancer Research Centre, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Lars M Pedersen
- Department of Haematology, Roskilde Hospital, Roskilde, Denmark
| | - Michael B Møller
- Department of Pathology, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
33
|
Chen J, Xu-Monette ZY, Deng L, Shen Q, Manyam GC, Martinez-Lopez A, Zhang L, Montes-Moreno S, Visco C, Tzankov A, Yin L, Dybkaer K, Chiu A, Orazi A, Zu Y, Bhagat G, Richards KL, Hsi ED, Choi WWL, van Krieken JH, Huh J, Ponzoni M, Ferreri AJM, Zhao X, Møller MB, Farnen JP, Winter JN, Piris MA, Pham L, Young KH. Dysregulated CXCR4 expression promotes lymphoma cell survival and independently predicts disease progression in germinal center B-cell-like diffuse large B-cell lymphoma. Oncotarget 2016; 6:5597-614. [PMID: 25704881 PMCID: PMC4467389 DOI: 10.18632/oncotarget.3343] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 01/04/2015] [Indexed: 12/13/2022] Open
Abstract
Abnormal expression of the chemokine receptor CXCR4 plays an essential role in tumor cell dissemination and disease progression. However, the significance of CXCR4 overexpression in de novo diffuse large B cell lymphoma (DLBCL) is unknown. In 743 patients with de novo diffuse large B cell lymphoma (DLBCL) who received standard Rituximab-CHOP immunochemotherapy, we assessed the expression of CXCR4 and dissected its prognostic significance in various DLBCL subsets. Our results showed that CXCR4+ patients was associated with male, bulky tumor, high Ki-67 index, activated B-cell-like (ABC) subtype, and Myc, Bcl-2 or p53 overexpression. Moreover, CXCR4+ was an independent factor predicting poorer progression-free survival in germinal-center B-cell-like (GCB)-DLBCL, but not in ABC-DLBCL; and in patients with an IPI of ≤2, but not in those with an IPI>2. The lack of prognostic significance of CXCR4 in ABC-DLBCL was likely due to the activation of p53 tumor suppressor attenuating CXCR4 signaling. Furthermore, concurrent CXCR4+ and BCL2 translocation showed dismal outcomes resembling but independent of MYC/BCL2 double-hit DLBCL. Gene expression profiling suggested that alterations in the tumor microenvironment and immune responses, increased tumor proliferation and survival, and the dissemination of CXCR4+ tumor cells to distant organs or tissues were underlying molecular mechanisms responsible for the CXCR4+ associated poor prognosis.
Collapse
Affiliation(s)
- Jiayu Chen
- Medical School of Taizhou University, Taizhou, Zhejiang, China.,Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zijun Y Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lijuan Deng
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Shen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganiraju C Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Li Zhang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Lihui Yin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - April Chiu
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Attilio Orazi
- Weill Medical College of Cornell University, New York, NY, USA
| | - Youli Zu
- The Methodist Hospital, Houston, TX, USA
| | - Govind Bhagat
- Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | - Kristy L Richards
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | - William W L Choi
- University of Hong Kong Li Ka Shing Faculty of Medicine, Hong Kong, China
| | | | - Jooryung Huh
- Asan Medical Center, Ulsan University College of Medicine, Seoul, Korea
| | | | | | - Xiaoying Zhao
- Zhejiang University School of Medicine, Second University Hospital, Hangzhou, China
| | | | - John P Farnen
- Gundersen Lutheran Health System, La Crosse, WI, USA
| | - Jane N Winter
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Miguel A Piris
- Hospital Universitario Marques de Valdecilla, Santander, Spain
| | - Lan Pham
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas School of Medicine, Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
34
|
Gifford GK, Gill AJ, Stevenson WS. Molecular subtyping of diffuse large B-cell lymphoma: update on biology, diagnosis and emerging platforms for practising pathologists. Pathology 2016; 48:5-16. [DOI: 10.1016/j.pathol.2015.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/02/2015] [Accepted: 10/12/2015] [Indexed: 01/04/2023]
|
35
|
Brown PJ, Wong KK, Felce SL, Lyne L, Spearman H, Soilleux EJ, Pedersen LM, Møller MB, Green TM, Gascoyne DM, Banham AH. FOXP1 suppresses immune response signatures and MHC class II expression in activated B-cell-like diffuse large B-cell lymphomas. Leukemia 2015; 30:605-16. [PMID: 26500140 PMCID: PMC4777777 DOI: 10.1038/leu.2015.299] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/07/2015] [Accepted: 10/13/2015] [Indexed: 12/25/2022]
Abstract
The FOXP1 (forkhead box P1) transcription factor is a marker of poor prognosis in diffuse large B-cell lymphoma (DLBCL). Here microarray analysis of FOXP1-silenced DLBCL cell lines identified differential regulation of immune response signatures and major histocompatibility complex class II (MHC II) genes as some of the most significant differences between germinal center B-cell (GCB)-like DLBCL with full-length FOXP1 protein expression versus activated B-cell (ABC)-like DLBCL expressing predominantly short FOXP1 isoforms. In an independent primary DLBCL microarray data set, multiple MHC II genes, including human leukocyte antigen DR alpha chain (HLA-DRA), were inversely correlated with FOXP1 transcript expression (P<0.05). FOXP1 knockdown in ABC-DLBCL cells led to increased cell-surface expression of HLA-DRA and CD74. In R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone)-treated DLBCL patients (n=150), reduced HLA-DRA (<90% frequency) expression correlated with inferior overall survival (P=0.0003) and progression-free survival (P=0.0012) and with non-GCB subtype stratified by the Hans, Choi or Visco-Young algorithms (all P<0.01). In non-GCB DLBCL cases with <90% HLA-DRA, there was an inverse correlation with the frequency (P=0.0456) and intensity (P=0.0349) of FOXP1 expression. We propose that FOXP1 represents a novel regulator of genes targeted by the class II MHC transactivator CIITA (MHC II and CD74) and therapeutically targeting the FOXP1 pathway may improve antigen presentation and immune surveillance in high-risk DLBCL patients.
Collapse
Affiliation(s)
- P J Brown
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - K K Wong
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
| | - S L Felce
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - L Lyne
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - H Spearman
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - E J Soilleux
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - L M Pedersen
- Department of Haematology, Roskilde Hospital, Roskilde, Denmark
| | - M B Møller
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - T M Green
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - D M Gascoyne
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - A H Banham
- NDCLS, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
36
|
Wang XJ, Seegmiller AC, Reddy NM, Li S. CD30 expression and its correlation withMYCrearrangement inde novodiffuse large B-cell lymphoma. Eur J Haematol 2015; 97:39-47. [DOI: 10.1111/ejh.12680] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Xuan J. Wang
- Department of Pathology, Microbiology and Immunology; Vanderbilt University School of Medicine; Nashville TN USA
| | - Adam C. Seegmiller
- Department of Pathology, Microbiology and Immunology; Vanderbilt University School of Medicine; Nashville TN USA
| | - Nishitha M. Reddy
- Department of Medicine; Division of Hematology/Oncology; Vanderbilt University School of Medicine; Nashville TN USA
| | - Shaoying Li
- Department of Pathology, Microbiology and Immunology; Vanderbilt University School of Medicine; Nashville TN USA
- Now at Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston TX USA
| |
Collapse
|
37
|
Novak AJ, Asmann YW, Maurer MJ, Wang C, Slager SL, Hodge LS, Manske M, Price-Troska T, Yang ZZ, Zimmermann MT, Nowakowski GS, Ansell SM, Witzig TE, McPhail E, Ketterling R, Feldman AL, Dogan A, Link BK, Habermann TM, Cerhan JR. Whole-exome analysis reveals novel somatic genomic alterations associated with outcome in immunochemotherapy-treated diffuse large B-cell lymphoma. Blood Cancer J 2015; 5:e346. [PMID: 26314988 PMCID: PMC4558593 DOI: 10.1038/bcj.2015.69] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/21/2015] [Indexed: 01/14/2023] Open
Abstract
Lack of remission or early relapse remains a major clinical issue in diffuse large B-cell lymphoma (DLBCL), with 30% of patients failing standard of care. Although clinical factors and molecular signatures can partially predict DLBCL outcome, additional information is needed to identify high-risk patients, particularly biologic factors that might ultimately be amenable to intervention. Using whole-exome sequencing data from 51 newly diagnosed and immunochemotherapy-treated DLBCL patients, we evaluated the association of somatic genomic alterations with patient outcome, defined as failure to achieve event-free survival at 24 months after diagnosis (EFS24). We identified 16 genes with mutations, 374 with copy number gains and 151 with copy number losses that were associated with failure to achieve EFS24 (P<0.05). Except for FOXO1 and CIITA, known driver mutations did not correlate with EFS24. Gene losses were localized to 6q21-6q24.2, and gains to 3q13.12-3q29, 11q23.1-11q23.3 and 19q13.12-19q13.43. Globally, the number of gains was highly associated with poor outcome (P=7.4 × 10−12) and when combined with FOXO1 mutations identified 77% of cases that failed to achieve EFS24. One gene (SLC22A16) at 6q21, a doxorubicin transporter, was lost in 54% of EFS24 failures and our findings suggest it functions as a doxorubicin transporter in DLBCL cells.
Collapse
Affiliation(s)
- A J Novak
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Y W Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - M J Maurer
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - C Wang
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - S L Slager
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - L S Hodge
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - M Manske
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Z-Z Yang
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - M T Zimmermann
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | - S M Ansell
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - T E Witzig
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - E McPhail
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - R Ketterling
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - A L Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - A Dogan
- Departments of Pathology and Laboratory Medicine, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - B K Link
- Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - T M Habermann
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - J R Cerhan
- Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
38
|
Staton AD, Koff JL, Chen Q, Ayer T, Flowers CR. Next-generation prognostic assessment for diffuse large B-cell lymphoma. Future Oncol 2015; 11:2443-57. [PMID: 26289217 DOI: 10.2217/fon.15.144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Current standard of care therapy for diffuse large B-cell lymphoma (DLBCL) cures a majority of patients with additional benefit in salvage therapy and autologous stem cell transplant for patients who relapse. The next generation of prognostic models for DLBCL aims to more accurately stratify patients for novel therapies and risk-adapted treatment strategies. This review discusses the significance of host genetic and tumor genomic alterations seen in DLBCL, clinical and epidemiologic factors, and how each can be integrated into risk stratification algorithms. In the future, treatment prediction and prognostic model development and subsequent validation will require data from a large number of DLBCL patients to establish sufficient statistical power to correctly predict outcome. Novel modeling approaches can augment these efforts.
Collapse
Affiliation(s)
- Ashley D Staton
- Department of Hematology & Medical Oncology, Emory University, Atlanta, GA 30322, USA
| | - Jean L Koff
- Department of Hematology & Medical Oncology, Emory University, Atlanta, GA 30322, USA
| | - Qiushi Chen
- H Milton Stewart School of Industrial & Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30318, USA
| | - Turgay Ayer
- H Milton Stewart School of Industrial & Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30318, USA
| | - Christopher R Flowers
- Department of Hematology & Medical Oncology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
39
|
Mendes F, Domingues C, Teixo R, Abrantes AM, Gonçalves AC, Nobre-Gois I, Jacobetty M, Sarmento AB, Botelho MF, Rosa MS. The importance of radiotherapy on diffuse large B cell lymphoma treatment: a current review. Cancer Metastasis Rev 2015; 34:511-25. [PMID: 26267803 DOI: 10.1007/s10555-015-9581-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Diffuse large B cell lymphoma is recognized as a heterogeneous group of hematological malignancies; two main subtypes germinal center B and activated B cells are well defined although 15% of patients remain with unclassifiable disease. R-CHOP treatment has proven to provide very effective results in limited or advanced stage of the disease. However, treatment solely involving R-CHOP submits the patient to possible chemotherapy-induced toxicities, which may be avoided with the use of radiotherapy. Patients with early stage localized disease or who are particularly unresponsive to chemotherapy may be more suitable for mixed modality treatment with R-CHOP and consolidative radiotherapy. Although radiotherapy is being slowly phased out by other treatment strategies including chemotherapy and therapeutic drugs, it is still a highly important method of treatment. The different forms of radiotherapy can be used alongside these "new-age" treatment strategies to further improve prognostic outcomes and overall survival rates. The establishment of radiotherapy as a treatment strategy provides a highly beneficial prognostic advantage in early stage, localized disease.
Collapse
Affiliation(s)
- Fernando Mendes
- Biophysics Unit-IBILI.CNC, Faculty of Medicine of University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548, Coimbra, Portugal. .,ESTESC-Coimbra Health School, Department Biomedical Laboratory Sciences, Polytechnic Institute of Coimbra, Coimbra, Portugal. .,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal.
| | - Cátia Domingues
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal.,Applied Molecular Biology and Clinical University of Hematology, Faculty of Medicine of University of Coimbra, Coimbra, Portugal
| | - Ricardo Teixo
- Biophysics Unit-IBILI.CNC, Faculty of Medicine of University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548, Coimbra, Portugal
| | - Ana Margarida Abrantes
- Biophysics Unit-IBILI.CNC, Faculty of Medicine of University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548, Coimbra, Portugal.,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal.,Applied Molecular Biology and Clinical University of Hematology, Faculty of Medicine of University of Coimbra, Coimbra, Portugal
| | - Inês Nobre-Gois
- Radiation Oncology Department, Hospital and University Center of Coimbra, Coimbra, Portugal
| | - Miguel Jacobetty
- Radiation Oncology Department, Hospital and University Center of Coimbra, Coimbra, Portugal
| | - Ana Bela Sarmento
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal.,Applied Molecular Biology and Clinical University of Hematology, Faculty of Medicine of University of Coimbra, Coimbra, Portugal
| | - Maria Filomena Botelho
- Biophysics Unit-IBILI.CNC, Faculty of Medicine of University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548, Coimbra, Portugal.,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal
| | - Manuel Santos Rosa
- Immunology Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
40
|
Frontline rituximab, cyclophosphamide, doxorubicin, and prednisone with bortezomib (VR-CAP) or vincristine (R-CHOP) for non-GCB DLBCL. Blood 2015; 126:1893-901. [PMID: 26232170 DOI: 10.1182/blood-2015-03-632430] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/02/2015] [Indexed: 12/14/2022] Open
Abstract
This phase 2 study evaluated whether substituting bortezomib for vincristine in frontline rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) therapy could improve efficacy in non-germinal center B-cell-like diffuse large B-cell lymphoma (non-GCB DLBCL), centrally confirmed by immunohistochemistry (Hans method). In total, 164 patients were randomized 1:1 to receive six 21-day cycles of rituximab 375 mg/m(2), cyclophosphamide 750 mg/m(2), and doxorubicin 50 mg/m(2), all IV day 1, prednisone 100 mg/m(2) orally days 1-5, plus either bortezomib 1.3 mg/m(2) IV days 1, 4, 8, 11 (rituximab, cyclophosphamide, doxorubicin, and prednisone with bortezomib [VR-CAP]; n = 84) or vincristine 1.4 mg/m(2) (maximum 2 mg) IV day 1 (R-CHOP; n = 80). There were no significant differences between VR-CAP and R-CHOP in complete response rate (64.5%, 66.2%; odds ratio [OR], 0.91; P = .80), overall response rate (93.4%, 98.6%; OR, 0.21; P = .11), progression-free survival (hazard ratio [HR], 1.12; P = .76), or overall survival (HR, 0.89; P = .75). Rates of grade ≥3 adverse events (AEs; 88%, 89%), serious AEs (38%, 34%), discontinuations due to AEs (7%, 3%), and deaths due to AEs (2%, 5%) were similar with VR-CAP and R-CHOP. Grade ≥3 peripheral neuropathy rates were 6% and 3%, respectively. VR-CAP did not improve efficacy vs R-CHOP in non-GCB DLBCL. This trial was registered at www.clinicaltrials.gov as #NCT01040871.
Collapse
|
41
|
Wells JE, Howlett M, Cheung LC, Kees UR. The role of CCN family genes in haematological malignancies. J Cell Commun Signal 2015; 9:267-78. [PMID: 26026820 DOI: 10.1007/s12079-015-0296-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/19/2015] [Indexed: 12/12/2022] Open
Abstract
Haematological malignancies, although a broad range of specific disease types, continue to show considerable overlap in classification, and patients are treated using similar chemotherapy regimes. In this review we look at the role of the CCN family of matricellular proteins and indicate their role in nine haematological malignancies including both myeloid and lymphoid neoplasms. The potential for further haematological neoplasms with CCN family associations is argued by summarising the demonstrated role of CCN family genes in the differentiation of haematopoietic stem cells (HSC) and mesenchymal stem cells. The expanding field of knowledge encompassing CCN family genes and cancers of the HSC-lineage highlights the importance of extracellular matrix-interactions in both normal physiology and tumorigenesis of the blood, bone marrow and lymph nodes.
Collapse
Affiliation(s)
- J E Wells
- Telethon Kids Institute, The University of Western Australia, PO Box 855 West Perth, Perth, Western Australia, 6872, Australia
| | - M Howlett
- Telethon Kids Institute, The University of Western Australia, PO Box 855 West Perth, Perth, Western Australia, 6872, Australia
| | - L C Cheung
- Telethon Kids Institute, The University of Western Australia, PO Box 855 West Perth, Perth, Western Australia, 6872, Australia
| | - Ursula R Kees
- Telethon Kids Institute, The University of Western Australia, PO Box 855 West Perth, Perth, Western Australia, 6872, Australia.
| |
Collapse
|
42
|
Chang C, Lin CH, Cheng AL, Medeiros LJ, Chang KC. Primary central nervous system diffuse large B-cell lymphoma has poorer immune cell infiltration and prognosis than its peripheral counterpart. Histopathology 2015; 67:625-35. [PMID: 25829022 DOI: 10.1111/his.12706] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/27/2015] [Indexed: 01/21/2023]
Abstract
AIMS Primary central nervous system (CNS) diffuse large B-cell lymphoma (PCNSL) is an ominous disease with a poor prognosis. The brain is an immune-privileged sanctuary, and this may contribute to an ineffective host immune response and thus a poorer outcome. The aim of this study was therefore to study the difference in the immune composition in PCNSL and non-CNS diffuse large B-cell lymphoma (DLBCL), and the role of the immune response in PCNSL prognosis. METHODS AND RESULTS Thirty-two biopsy specimens of PCNSL and 30 specimens of low-stage non-CNS DLBCL from immunocompetent patients formed the study group. The density and distribution of immune cells, including dendritic cells (dendritic cell-specific lysosomal-associated membrane protein-positive and S100-positive), effector/memory T cells (CD45RO-positive), and cytotoxic T cells (granzyme B-positive), and the expression of HLA-DR by lymphoma cells, were evaluated immunohistochemically. PCNSL patients showed poorer overall survival (P = 0.032). On comparison of the PCNSL and DLBCL biopsy specimens, the PCNSL cells showed less HLA-DR expression (P = 0.003), and there were fewer S100-positive cells (P < 0.01), and effector T cells (P = 0.026) infiltrating PCNSL than infiltrating DLBCL. For PCNSL patients, fewer cytotoxic T cells in the background constituted a poor prognostic factor (P = 0.004). Intratumoral S100-positive cell infiltration was positively correlated with T-cell infiltration, and a T-cell rimming pattern. CONCLUSIONS In PCNSL, the baseline antitumour immune response is less as compared with non-CNS DLBCL, and this response may play a role in the poorer prognosis. Adjuvant dendritic cell and T-cell immunotherapy may further boost treatment responses in PCNSL patients.
Collapse
Affiliation(s)
- Chen Chang
- Department of Pathology, College of Medicine, National Cheng Kung University and Hospital, Tainan, Taiwan
| | - Ching-Hung Lin
- Departments of Oncology and Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ann-Lii Cheng
- Departments of Oncology and Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kung-Chao Chang
- Department of Pathology, College of Medicine, National Cheng Kung University and Hospital, Tainan, Taiwan
| |
Collapse
|
43
|
Higashi M, Tokuhira M, Fujino S, Yamashita T, Abe K, Arai E, Kizaki M, Tamaru JI. Loss of HLA-DR expression is related to tumor microenvironment and predicts adverse outcome in diffuse large B-cell lymphoma. Leuk Lymphoma 2015; 57:161-6. [DOI: 10.3109/10428194.2015.1038708] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
44
|
Abstract
Non-Hodgkin lymphoma (NHL) consists of many histologically and biologically distinct lymphoid malignancies with poorly understood, but possibly distinct, etiologies. The patterns of incidence and time trend vary not only by age, sex, and race/ethnicity in the USA, but also show significant geographic differences, suggesting the potential role of infectious agents, environmental factors, and lifestyle factors in addition to host genetic status in the development of NHL. Important pathogenetic mechanisms include immune modulation and chronic antigen stimulation. Epidemiologic studies in the past two decades have provided intriguing new insights on the possible causes of lymphoma and support the idea that there is some mechanistic commonality of lymphomagenesis, but significant etiologic heterogeneity clearly exists. This review presents a summary of the current understanding of the descriptive epidemiology and etiology of NHL and suggests areas of focus for future epidemiologic research.
Collapse
|
45
|
Diffuse large B-cell lymphoma: optimizing outcome in the context of clinical and biologic heterogeneity. Blood 2015; 125:22-32. [DOI: 10.1182/blood-2014-05-577189] [Citation(s) in RCA: 379] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Abstract
Although the majority of patients with diffuse large B-cell lymphoma (DLBCL) can be cured with standard rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), patients who fail R-CHOP have a dismal outcome. Thus, optimization of front-line therapy, as well as the development of more effective salvage strategies, remains an important objective. Advances in molecular genetics have vastly improved our understanding of the biological diversity of DLBCL and have led to the discovery of key oncogenic pathways. In addition to the major molecular designations of germinal center B-cell and activated B-cell subtypes, next-generation sequencing technologies have unveiled the remarkable complexity of DLBCL and identified unique molecular targets that may be differentially exploited for therapeutic benefit. These findings have translated into a growing list of promising novel agents. Moving forward, it is of paramount importance to recognize the heterogeneity of DLBCL and to investigate these targeted agents within patient populations who are most likely to benefit. It will be necessary to prioritize drugs that affect key driver pathways and to combine them rationally to optimize their benefit. Improved prognostication and the availability of predictive biomarkers will be crucial to allow for the possibility of individualized risk-adapted therapy.
Collapse
|
46
|
Extranodal and nodal diffuse large B cell lymphoma of the head and neck: two different entities? Ann Hematol 2014; 94:609-16. [DOI: 10.1007/s00277-014-2256-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 11/08/2014] [Indexed: 10/24/2022]
|
47
|
Veldman-Jones MH, Lai Z, Wappett M, Harbron CG, Barrett JC, Harrington EA, Thress KS. Reproducible, Quantitative, and Flexible Molecular Subtyping of Clinical DLBCL Samples Using the NanoString nCounter System. Clin Cancer Res 2014; 21:2367-78. [PMID: 25301847 DOI: 10.1158/1078-0432.ccr-14-0357] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 10/03/2014] [Indexed: 11/16/2022]
Abstract
PURPOSE Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous disease with distinct molecular subtypes. The most established subtyping approach, the "Cell of Origin" (COO) algorithm, categorizes DLBCL into activated B-cell (ABC) and germinal center B-cell (GCB)-like subgroups through gene expression profiling. Recently developed immunohistochemical (IHC) techniques and other established methodologies can deliver discordant results and have various technical limitations. We evaluated the NanoString nCounter gene expression system to address issues with current platforms. EXPERIMENTAL DESIGN We devised a scoring system using 145 genes from published datasets to categorize DLBCL samples. After cell line validation, clinical tissue segmentation was tested using commercially available diagnostic DLBCL samples. Finally, we profiled biopsies from patients with relapsed/refractory DLBCL enrolled in the fostamatinib phase IIb clinical trial using three independent RNA expression platforms: NanoString, Affymetrix, and qNPA. RESULTS Diagnostic samples showed a typical spread of subtypes with consistent gene expression profiles across matched fresh, frozen, and formalin-fixed paraffin-embedded tissues. Results from biopsy samples across platforms were remarkably consistent, in contrast to published IHC data. Interestingly, COO segmentation of longitudinal fostamatinib biopsies taken at initial diagnosis and then again at primary relapse showed 88% concordance (15/17), suggesting that COO designation remains stable over the course of disease progression. CONCLUSIONS DLBCL segmentation of patient tumor samples is possible using a number of expression platforms. However, we found that NanoString offers the most flexibility and fewest limitations in regards to robust clinical tissue subtype characterization. These subtype distinctions should help guide disease prognosis and treatment options within DLBCL clinical practice.
Collapse
Affiliation(s)
| | - Zhongwu Lai
- AstraZeneca Oncology Innovative Medicines, Waltham, Massachusetts
| | - Mark Wappett
- AstraZeneca Oncology Innovative Medicines, Macclesfield, United Kingdom
| | - Chris G Harbron
- AstraZeneca Discovery Sciences, Macclesfield, United Kingdom
| | - J Carl Barrett
- AstraZeneca Oncology Innovative Medicines, Waltham, Massachusetts
| | | | - Kenneth S Thress
- AstraZeneca Oncology Innovative Medicines, Waltham, Massachusetts.
| |
Collapse
|
48
|
Slack GW, Steidl C, Sehn LH, Gascoyne RD. CD30 expression inde novodiffuse large B-cell lymphoma: a population-based study from British Columbia. Br J Haematol 2014; 167:608-17. [DOI: 10.1111/bjh.13085] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/10/2014] [Indexed: 01/20/2023]
Affiliation(s)
- Graham W. Slack
- Centre for Lymphoid Cancers; British Columbia Cancer Agency; Vancouver BC Canada
- Department of Pathology and Laboratory Medicine; British Columbia Cancer Agency; Vancouver BC Canada
| | - Christian Steidl
- Centre for Lymphoid Cancers; British Columbia Cancer Agency; Vancouver BC Canada
| | - Laurie H. Sehn
- Centre for Lymphoid Cancers; British Columbia Cancer Agency; Vancouver BC Canada
- Department of Medical Oncology; British Columbia Cancer Agency; Vancouver BC Canada
| | - Randy D. Gascoyne
- Centre for Lymphoid Cancers; British Columbia Cancer Agency; Vancouver BC Canada
- Department of Pathology and Laboratory Medicine; British Columbia Cancer Agency; Vancouver BC Canada
| |
Collapse
|
49
|
C-MYC aberrations as prognostic factors in diffuse large B-cell lymphoma: a meta-analysis of epidemiological studies. PLoS One 2014; 9:e95020. [PMID: 24740248 PMCID: PMC3989276 DOI: 10.1371/journal.pone.0095020] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 03/21/2014] [Indexed: 12/19/2022] Open
Abstract
Objectives Various studies have investigated the prognostic value of C-MYC aberrations in diffuse large B-cell lymphoma (DLBCL). However, the role of C-MYC as an independent prognostic factor in clinical practice remains controversial. A systematic review and meta-analysis were performed to clarify the clinical significance of C-MYC aberrations in DLBCL patients. Methods The pooled hazard ratios (HRs) for overall survival (OS) and event-free survival (EFS) were calculated as the main effect size estimates. The procedure was conducted according to the Cochrane handbook and PRISMA guidelines, including the use of a heterogeneity test, publication bias assessment, and meta-regression, as well as subgroup analyses. Results Twenty-four eligible studies enrolling 4662 patients were included in this meta-analysis. According to the nature of C-MYC aberrations (gene, protein, and mRNA), studies were divided into several subgroups. For DLBCL patients with C-MYC gene abnormalities, the combined HR was 2.22 (95% confidence interval, 1.89 to 2.61) for OS and 2.29 (95% confidence interval, 1.81 to 2.90) for EFS, compared to patients without C-MYC gene abnormalities. For DLBCL patients with overexpression of C-MYC protein and C-MYC mRNA, pooled HRs for OS were 2.13 and 1.62, respectively. C-MYC aberrations appeared to play an independent role among other well-known prognostic factors in DLBCL. Addition of rituximab could not overcome the inferior prognosis conferred by C-MYC. Conclusion The present systematic review and meta-analysis confirm the prognostic value of C-MYC aberrations. Screening of C-MYC should have definite prognostic meaning for DLBCL stratification, thus guaranteeing a more tailored therapy.
Collapse
|
50
|
Yamamoto W, Nakamura N, Tomita N, Takeuchi K, Ishii Y, Takahashi H, Watanabe R, Takasaki H, Motomura S, Kobayashi S, Yokose T, Ishigatsubo Y, Sakai R. Human leukocyte antigen-DR expression on flow cytometry and tumor-associated macrophages in diffuse large B-cell lymphoma treated by rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone therapy: retrospective cohort study. Leuk Lymphoma 2014; 55:2721-7. [PMID: 24528218 DOI: 10.3109/10428194.2014.893311] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Loss of human leukocyte antigen (HLA)-DR expression may be related to a poor prognosis of diffuse large B-cell lymphoma (DLBCL), and tumor-associated macrophages (TAMs) may influence tumor progression. We retrospectively reviewed 36 patients with newly diagnosed DLBCL who received R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone) therapy at Kanagawa Cancer Center in Japan from 2004 to 2010. HLA-DR expression by lymphoma cells was evaluated using flow cytometry, and TAMs in lymphoma tissue were detected by immunohistochemistry for CD68 as a marker of macrophages and CD163 as a marker of M2 TAMs. Three-year overall survival was, respectively, 100% versus 69.6% in the HLA-DR "bright" and "not bright" groups (p = 0.012). Patients from the HLA-DR "not bright" group with strong CD163 expression had a much worse prognosis than other patients. The HLA-DR status shown by flow cytometry can be used to predict the prognosis of patients with DLBCL receiving R-CHOP therapy and prognostic accuracy can be increased by also assessing TAMs.
Collapse
Affiliation(s)
- Wataru Yamamoto
- Department of Medical Oncology, Kanagawa Cancer Center , Yokohama , Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|