1
|
Masel R, Roche ME, Martinez-Outschoorn U. Hodgkin Lymphoma: A disease shaped by the tumor micro- and macroenvironment. Best Pract Res Clin Haematol 2023; 36:101514. [PMID: 38092473 DOI: 10.1016/j.beha.2023.101514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 12/18/2023]
Abstract
The tumor microenvironment (TMicroE) and tumor macroenvironment (TMacroE) are defining features of classical Hodgkin lymphoma (cHL). They are of critical importance to clinicians since they explain the common signs and symptoms, allow us to classify these neoplasms, develop prognostic and predictive biomarkers, bioimaging and novel treatments. The TMicroE is defined by effects of cancer cells to their immediate surrounding and within the tumor. Effects of cancer cells at a distance or outside of the tumor define the TMacroE. Paraneoplastic syndromes are signs and symptoms due to effects of cancer at a distance or the TMacroE, which are not due to direct cancer cell infiltration. The most common paraneoplastic symptoms are B-symptoms, which manifest as fevers, chills, drenching night sweats, and/or weight loss. Less common paraneoplastic syndromes include those that affect the central nervous system, skin, kidney, and hematological autoimmune phenomena including hemophagocytic lymphohistiocytosis (HLH). Paraneoplastic signs such as leukocytosis, lymphopenia, anemia, and hypoalbuminemia are prognostic biomarkers. The neoplastic cells in cHL are the Hodgkin and Reed Sternberg (HRS) cells, which are preapoptotic germinal center B cells with a high mutational burden and almost universal genetic alterations at the 9p24.1 locus primarily through copy gain and amplification with strong activation of signaling via PD-L1, JAK-STAT, NFkB, and c-MYC. In the majority of cases of cHL over 95% of the tumor cells are non-neoplastic. In the TMicroE, HRS cells recruit and mold non-neoplastic cells vigorously via extracellular vesicles, chemokines, cytokines and growth factors such as CCL5, CCL17, IL6, and TGF-β to promote a feed-forward inflammatory loop, which drives cancer aggressiveness and anti-cancer immune evasion. Novel single cell profiling techniques provide critical information on the role in cHL of monocytes-macrophages, neutrophils, T helper, Tregs, cytotoxic CD8+ T cells, eosinophils, mast cells and fibroblasts. Here, we summarize the effects of EBV on the TMicroE and TMacroE. In addition, how the metabolism of the TMicroE of cHL affects bioimaging and contributes to cancer aggressiveness is reviewed. Finally, we discuss how the TMicroE is being leveraged for risk adapted treatment strategies based on bioimaging results and novel immune therapies. In sum, it is clear that we cannot effectively manage patients with cHL without understanding the TMicroE and TMacroE and its clinical importance is expected to continue to grow rapidly.
Collapse
Affiliation(s)
- Rebecca Masel
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University-Philadelphia, USA; Department of Medicine, Cardeza Foundation for Hematological Research, Thomas Jefferson University-Philadelphia, USA
| | - Megan E Roche
- Department of Medicine, Cardeza Foundation for Hematological Research, Thomas Jefferson University-Philadelphia, USA
| | - Ubaldo Martinez-Outschoorn
- Department of Medicine, Cardeza Foundation for Hematological Research, Thomas Jefferson University-Philadelphia, USA.
| |
Collapse
|
2
|
Zijtregtop EAM, Diez C, Zwaan CM, Veening MA, Beishuizen A, Meyer-Wentrup FAG. Thymus and activation-regulated chemokine (TARC) as treatment response marker for paediatric Hodgkin lymphoma: A pilot study. Br J Haematol 2023; 200:70-78. [PMID: 36128637 PMCID: PMC10087307 DOI: 10.1111/bjh.18473] [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: 07/07/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/28/2022]
Abstract
Classical Hodgkin lymphoma (cHL) is characterised by malignant Hodgkin Reed-Sternberg cells located in an inflammatory microenvironment. Blood biomarkers result from active cross-talk between malignant and non-malignant cells. One promising biomarker in adult patients with cHL is thymus and activation-regulated chemokine (TARC). We investigated TARC as marker for interim and end-of-treatment response in paediatric cHL. In this multicentre prospective study, TARC levels were measured among 99 paediatric patients with cHL before each cycle of chemotherapy and were linked with interim and end-of-treatment remission status. TARC levels were measured by enzyme-linked immunosorbent assay. At diagnosis, TARC levels were elevated in 96% of patients. Plasma TARC levels declined significantly after one cycle of chemotherapy (p < 0.01 vs. baseline) but did not differ at interim assessment by positron emission tomography (p = 0.31). In contrast, median plasma TARC at end of treatment was significantly higher in three patients with progressive disease compared to those in complete remission (1.226 vs. 90 pg/ml; p < 0.001). We demonstrate that, in paediatric patients, plasma TARC is a valuable response marker at end-of-treatment, but not at interim analysis after the first two chemotherapy cycles. Further research is necessary to investigate TARC as marker for long-term progression free survival.
Collapse
Affiliation(s)
- Eline A M Zijtregtop
- Department of Paediatric Haematology and Oncology, Erasmus Medical Centre - Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Paediatric Haemato-Oncology, Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| | - Claudius Diez
- Department of Paediatric Haemato-Oncology, Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| | - C Michel Zwaan
- Department of Paediatric Haematology and Oncology, Erasmus Medical Centre - Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Paediatric Haemato-Oncology, Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| | - Margreet A Veening
- Department of Paediatric Haemato-Oncology, Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands.,Department of Paediatric Haemato-Oncology, Amsterdam University Medical Centre, Location VUmc, Amsterdam, The Netherlands
| | - Auke Beishuizen
- Department of Paediatric Haematology and Oncology, Erasmus Medical Centre - Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Paediatric Haemato-Oncology, Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| | - Friederike A G Meyer-Wentrup
- Department of Paediatric Haemato-Oncology, Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
3
|
Immune Biomarkers in the Peripheral Blood and Tumor Microenvironment of Classical Hodgkin Lymphoma Patients in Relation to Tumor Burden and Response to Treatment. Hemasphere 2022; 6:e794. [PMID: 36325271 PMCID: PMC9619233 DOI: 10.1097/hs9.0000000000000794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/25/2022] [Indexed: 12/13/2022] Open
Abstract
In classical Hodgkin lymphoma (cHL), the malignant cells represent only a small fraction of the tumor. Yet, they orchestrate a lymphocyte-dominated tumor microenvironment (TME) that supports their survival and growth. The systemic effects of this local immunomodulation are not fully elucidated. Here, we aimed at characterizing circulating lymphocytes and plasma proteins in relation to clinical parameters and treatment effect. Peripheral blood (PB) samples were obtained from 48 consecutive patients at diagnosis and at 2 time points after successful primary treatment. Single-cell suspensions were prepared from lymph node (LN) biopsies obtained for routine diagnostic purposes. Twenty healthy individuals were included as controls. Cells from PB and LN were analyzed by flow cytometry, and plasma proteins by Proximity Extension Assay. We found that the frequencies of T and B cells positively correlated between the LN and the PB compartments. Compared to controls, cHL patients had higher frequencies of proliferating T cells as well as higher expression of programmed death (PD)-1 and cytotoxic T lymphocyte antigen (CTLA)-4 in circulating T cells, and lower naive T-cell frequencies. Advanced-stage patients had fewer NK cells with a functionally impaired phenotype. Differences in the immune profile were observed in patients with a high tumor burden and with high inflammation, respectively. Most of these deviations disappeared after standard first-line treatment. Patients who received radiotherapy involving the mediastinum had low T-cell counts for a prolonged period. Our findings suggest that the immunomodulation of lymphocytes in the TME of cHL might affect immune biomarkers in the PB.
Collapse
|
4
|
Immune-Proteome Profiling in Classical Hodgkin Lymphoma Tumor Diagnostic Tissue. Cancers (Basel) 2021; 14:cancers14010009. [PMID: 35008176 PMCID: PMC8750205 DOI: 10.3390/cancers14010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
In classical Hodgkin Lymphoma (cHL), immunoediting via protein signaling is key to evading tumor surveillance. We aimed to identify immune-related proteins that distinguish diagnostic cHL tissues (=diagnostic tumor lysates, n = 27) from control tissues (reactive lymph node lysates, n = 30). Further, we correlated our findings with the proteome plasma profile between cHL patients (n = 26) and healthy controls (n = 27). We used the proximity extension assay (PEA) with the OlinkTM multiplex Immuno-Oncology panel, consisting of 92 proteins. Univariate, multivariate-adjusted analysis and Benjamini–Hochberg’s false discovery testing (=Padj) were performed to detect significant discrepancies. Proteins distinguishing cHL cases from controls were more numerous in plasma (30 proteins) than tissue (17 proteins), all Padj < 0.05. Eight of the identified proteins in cHL tissue (PD-L1, IL-6, CCL17, CCL3, IL-13, MMP12, TNFRS4, and LAG3) were elevated in both cHL tissues and cHL plasma compared with control samples. Six proteins distinguishing cHL tissues from controls tissues were significantly correlated to PD-L1 expression in cHL tissue (IL-6, MCP-2, CCL3, CCL4, GZMB, and IFN-gamma, all p ≤0.05). In conclusion, this study introduces a distinguishing proteomic profile in cHL tissue and potential immune-related markers of pathophysiological relevance.
Collapse
|
5
|
Jachimowicz RD, Klapper W, Glehr G, Müller H, Haverkamp H, Thorns C, Hansmann ML, Möller P, Stein H, Rehberg T, von Tresckow B, Reinhardt HC, Borchmann P, Chan FC, Spang R, Scott DW, Engert A, Steidl C, Altenbuchinger M, Rosenwald A. Gene expression-based outcome prediction in advanced stage classical Hodgkin lymphoma treated with BEACOPP. Leukemia 2021; 35:3589-3593. [PMID: 34112956 PMCID: PMC8632672 DOI: 10.1038/s41375-021-01314-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/23/2021] [Accepted: 05/27/2021] [Indexed: 12/26/2022]
Affiliation(s)
- Ron D Jachimowicz
- Department I of Internal Medicine; German Hodgkin Study Group, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
- Max Planck Research Group Mechanisms of DNA Repair, Max Planck Institute for Biology of Ageing, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section, University Hospital Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Gunther Glehr
- Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Horst Müller
- Department I of Internal Medicine; German Hodgkin Study Group, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Heinz Haverkamp
- Department I of Internal Medicine; German Hodgkin Study Group, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christoph Thorns
- Department of Pathology, Marien-Hospital Hamburg, Hamburg, Germany
| | | | - Peter Möller
- Department of Pathology, University Hospital Ulm, Ulm, Germany
| | | | - Thorsten Rehberg
- Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Bastian von Tresckow
- Department I of Internal Medicine; German Hodgkin Study Group, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, German Cancer Consortium (DKTK partner site Essen), Essen, Germany
| | - H C Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, German Cancer Consortium (DKTK partner site Essen), Essen, Germany
| | - Peter Borchmann
- Department I of Internal Medicine; German Hodgkin Study Group, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | | | - Rainer Spang
- Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - David W Scott
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Andreas Engert
- Department I of Internal Medicine; German Hodgkin Study Group, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | | | - Michael Altenbuchinger
- Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
- Research Group Computational Biology, University of Hohenheim, Stuttgart, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, and Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| |
Collapse
|
6
|
Kopińska A, Koclęga A, Francuz T, Helbig G. Serum thymus and activation-regulated chemokine (TARC) levels in newly diagnosed patients with Hodgkin lymphoma: a new promising and predictive tool? Preliminary report. J Hematop 2021. [DOI: 10.1007/s12308-021-00470-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
AbstractThymus and activation-regulated chemokine (TARC) is expressed on Reed-Sternberg cells of patients with classical Hodgkin lymphoma (HL) and may serve as a marker in response assessment. In our study, we correlated serum TARC levels with early response to treatment measured by PET/CT in 19 newly diagnosed patients with HL who received ABVD (Adriblastin, Bleomycin, Vinblastine, Dacarbazine) regimen. Finally, 17 patients were analyzed and six of them (35%) achieved PET/CT negativity defined as Deauville (D) 1 or 2 after 2 cycles of ABVD; 11 pts (65%) had D3 on PET/CT. None of the patients presented D 4/5. Median serum TARC levels at diagnosis were significantly higher when compared with healthy controls: 5718 pg/ml vs 76.1 pg/ml (p < 0.001). All study patients were treated with ABVD regimen and there was a significant decrease of baseline serum TARC levels after 2 cycles of therapy. No significant difference of baseline serum TARC levels was demonstrated between patients with D1/2 and D3 whereas levels were significantly decreased after 2 cycles of ABVD in patients D1/2 vs D3; p = 0.049. There was a tendency to higher baseline serum TARC levels in patients with an increased LDH (lactate dehydrogenase) activity (p = 0.08) and in those who progressed when compared with those who maintained response (p = 0.09). Serum TARC levels decrease after chemotherapy and may serve as a marker of response assessment.
Collapse
|
7
|
Lymphoma-Associated Biomarkers Are Increased in Current Smokers in Twin Pairs Discordant for Smoking. Cancers (Basel) 2021; 13:cancers13215395. [PMID: 34771561 PMCID: PMC8582438 DOI: 10.3390/cancers13215395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Smoking is associated with a moderate increased risk of Hodgkin and follicular lymphoma. To help understand why, we examined lymphoma-related biomarker levels among 134 smoking and non-smoking twins (67 pairs) ascertained from the Finnish Twin Cohort. We validated self-reported smoking history by measuring serum cotinine, a metabolite of nicotine, from previously collected frozen serum samples. In total, 27 immune biomarkers were assayed using the Luminex Multiplex platform (R & D Systems). We found that four immune response biomarkers were higher and one was lower among smoking compared to non-smoking twins. The strongest association was observed for CCL17/TARC, a biomarker elevated in Hodgkin lymphoma patients. Immune biomarker levels were similar in former smokers and non-smokers. Current smoking may increase levels of immune proteins that could partially explain the association between smoking and risk of certain lymphomas. Abstract Smoking is associated with a moderate increased risk of Hodgkin and follicular lymphoma. To understand why, we examined lymphoma-related biomarker levels among 134 smoking and non-smoking twins (67 pairs) ascertained from the Finnish Twin Cohort. Previously collected frozen serum samples were tested for cotinine to validate self-reported smoking history. In total, 27 immune biomarkers were assayed using the Luminex Multiplex platform (R & D Systems). Current and non-current smokers were defined by a serum cotinine concentration of >3.08 ng/mL and ≤3.08 ng/mL, respectively. Associations between biomarkers and smoking were assessed using linear mixed models to estimate beta coefficients and standard errors, adjusting for age, sex and twin pair as a random effect. There were 55 never smokers, 43 current smokers and 36 former smokers. CCL17/TARC, sgp130, haptoglobin, B-cell activating factor (BAFF) and monocyte chemoattractant protein-1 (MCP1) were significantly (p < 0.05) associated with current smoking and correlated with increasing cotinine concentrations (Ptrend < 0.05). The strongest association was observed for CCL17/TARC (Ptrend = 0.0001). Immune biomarker levels were similar in former and never smokers. Current smoking is associated with increased levels of lymphoma-associated biomarkers, suggesting a possible mechanism for the link between smoking and risk of these two B-cell lymphomas.
Collapse
|
8
|
In-depth cell-free DNA sequencing reveals genomic landscape of Hodgkin’s lymphoma and facilitates ultrasensitive residual disease detection. MED 2021; 2:1171-1193.e11. [DOI: 10.1016/j.medj.2021.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/12/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
|
9
|
Rinaldi C, Corrigan DK, Dennany L, Jarrett RF, Lake A, Baker MJ. Development of an Electrochemical CCL17/TARC Biosensor toward Rapid Triage and Monitoring of Classic Hodgkin Lymphoma. ACS Sens 2021; 6:3262-3272. [PMID: 34478275 DOI: 10.1021/acssensors.1c00972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A point-of-care blood test for the detection of an emerging biomarker, CCL17/TARC, could prove transformative for the clinical management of classic Hodgkin lymphoma (cHL). Primary care diagnosis is challenging due to nonspecific clinical presentation and lack of a diagnostic test, leading to significant diagnostic delays. Treatment monitoring encounters false-positive and negative results, leading to avoidable chemotherapy toxicity, or undertreatment, impacting patient morbidity and mortality. Here, we present an amperometric CCL17/TARC immunosensor, based on the utilization of a thiolated heterobifunctional cross-linker and sandwich antibody assay, to facilitate novel primary care triage and chemotherapy monitoring strategies for cHL. The immunosensor shows excellent analytical performance for clinical testing; linearity (R2 = 0.986), detection limit (194 pg/mL), and lower and upper limits of quantitation (387-50 000 pg/mL). The biosensor differentiated all 42 newly diagnosed cHL patients from healthy volunteers, based on serum CCL17/TARC concentration, using blood samples collected prior to treatment intervention. The immunosensor also discriminated between paired blood samples of all seven cHL patients, respectively, collected prior to treatment and during chemotherapy, attributed to the decrease in serum CCL17/TARC concentration following chemotherapy response. Overall, we have shown, for the first time, the potential of an electrochemical CCL17/TARC biosensor for primary care triage and chemotherapy monitoring for cHL, which would have positive clinical and psychosocial implications for patients, while streamlining current healthcare pathways.
Collapse
Affiliation(s)
- Christopher Rinaldi
- WestCHEM, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, 99 George Street, Glasgow G1 1RD, U.K
| | - Damion K. Corrigan
- Department of Biomedical Engineering, University of Strathclyde, 40 George Street, Glasgow G1 1QE, U.K
| | - Lynn Dennany
- WestCHEM, Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, 99 George Street, Glasgow G1 1RD, U.K
| | - Ruth F. Jarrett
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Bearsden, Glasgow G61 1QH, U.K
| | - Annette Lake
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Bearsden, Glasgow G61 1QH, U.K
| | - Matthew J. Baker
- DXCOVER Ltd., University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow G1 1RD, U.K
| |
Collapse
|
10
|
Drees EEE, Roemer MGM, Groenewegen NJ, Perez‐Boza J, van Eijndhoven MAJ, Prins LI, Verkuijlen SAWM, Tran X, Driessen J, Zwezerijnen GJC, Stathi P, Mol K, Karregat JJJP, Kalantidou A, Vallés‐Martí A, Molenaar TJ, Aparicio‐Puerta E, van Dijk E, Ylstra B, Groothuis‐Oudshoorn CGM, Hackenberg M, de Jong D, Zijlstra JM, Pegtel DM. Extracellular vesicle miRNA predict FDG-PET status in patients with classical Hodgkin Lymphoma. J Extracell Vesicles 2021; 10:e12121. [PMID: 34295456 PMCID: PMC8282992 DOI: 10.1002/jev2.12121] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
Abstract
Minimally-invasive tools to assess tumour presence and burden may improve clinical management. FDG-PET (metabolic) imaging is the current gold standard for interim response assessment in patients with classical Hodgkin Lymphoma (cHL), but this technique cannot be repeated frequently. Here we show that microRNAs (miRNA) associated with tumour-secreted extracellular vesicles (EVs) in the circulation of cHL patients may improve response assessment. Small RNA sequencing and qRT-PCR reveal that the relative abundance of cHL-expressed miRNAs, miR-127-3p, miR-155-5p, miR-21-5p, miR-24-3p and let-7a-5p is up to hundred-fold increased in plasma EVs of cHL patients pre-treatment when compared to complete metabolic responders (CMR). Notably, in partial responders (PR) or treatment-refractory cases (n = 10) the EV-miRNA levels remain elevated. In comparison, tumour specific copy number variations (CNV) were detected in cell-free DNA of 8 out of 10 newly diagnosed cHL patients but not in patients with PR. Combining EV-miR-127-3p and/or EV-let-7a-5p levels, with serum TARC (a validated protein cHL biomarker), increases the accuracy for predicting PET-status (n = 129) to an area under the curve of 0.93 (CI: 0.87-0.99), 93.5% sensitivity, 83.8/85.0% specificity and a negative predictive value of 96%. Thus the level of tumour-associated miRNAs in plasma EVs is predictive of metabolic tumour activity in cHL patients. Our findings suggest that plasma EV-miRNA are useful for detection of small residual lesions and may be applied as serial response prediction tool.
Collapse
Affiliation(s)
- Esther E. E. Drees
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Margaretha G. M. Roemer
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Nils J. Groenewegen
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
- ExBiome B.V.AmsterdamThe Netherlands
| | - Jennifer Perez‐Boza
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | | | - Leah I. Prins
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Sandra A. W. M. Verkuijlen
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Xuan‐Mai Tran
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Julia Driessen
- Department of HematologyAmsterdam UMCCancer Center Amsterdam, University of AmsterdamAmsterdamThe Netherlands
| | - G. J. C. Zwezerijnen
- Department of Radiology and Nuclear MedicineCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Phylicia Stathi
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Kevin Mol
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Joey J. J. P. Karregat
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Aikaterini Kalantidou
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Andrea Vallés‐Martí
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - T. J. Molenaar
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Ernesto Aparicio‐Puerta
- Department of GeneticsComputational Epigenomics and BioinformaticsUniversity of GranadaGranadaSpain
| | - Erik van Dijk
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Bauke Ylstra
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | | | - Michael Hackenberg
- ExBiome B.V.AmsterdamThe Netherlands
- Department of GeneticsComputational Epigenomics and BioinformaticsUniversity of GranadaGranadaSpain
| | - Daphne de Jong
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Josée M. Zijlstra
- Department of HematologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - D. Michiel Pegtel
- Department of PathologyCancer Center AmsterdamAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
11
|
Şumnu Ş, Mehtap Ö, Mersin S, Toptaş T, Görür G, Gedük A, Ünal S, Polat MG, Aygün K, Yenihayat EM, Albayrak H, Uluköylü Mengüç M, Tarkun P, Hacıhanifioğlu A. Serum calprotectin (S100A8/A9) levels as a new potential biomarker of treatment response in Hodgkin lymphoma. Int J Lab Hematol 2021; 43:638-644. [PMID: 33904653 DOI: 10.1111/ijlh.13559] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 04/04/2021] [Accepted: 04/06/2021] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Hodgkin lymphoma (HL) is unusual among malignancies, with inflammation playing such a prominent role in its pathogenesis. S100A8/A9 (calprotectin) is a heterodimeric protein, which has a role in the inflammatory response and oncogenesis. In this study in HL patients, the correlation between serum S100A8/A9 levels and treatment responses was investigated along with whether this marker is correlated with other inflammatory markers. MATERIALS AND METHODS Thirty-three HL patients and 20 healthy volunteers were included. Demographic and clinical characteristics were recorded. Calprotectin levels were measured with Human S100A8/A9 Heterodimer Quantikine ELISA kit. Calprotectin levels were measured twice in patients, before and after treatment, and once in the control group. Treatment responses were evaluated with positron emission tomography-computed tomography (PET-CT). RESULTS The mean age of patients was 44.3 ± 18.1 (66.3% male). The median (IQR) values of S100A8/A9 before and after treatment in the patient group were 4.98 (2.6-7.8) and 1.87 (1.1-4.8)µg/mL. Median (IQR) S100A8/A9 concentration in the control group was 1.41 (0.98-2.73)µg/mL. In patients, pretreatment values were significantly higher than in controls (P < .001). However, median values of patients after treatment and controls were similar. Patient median S100A8/A9 levels were significantly lower post-treatment compared with pretreatment values (P = .001). When inflammatory markers were examined within groups, no relationship was found between markers. In ROC analysis, a S100A8/A9 cutoff value of ≥3.31µg/mL accurately discriminated end-of-treatment PET positivity (AUC = 0.78; 95% CI 0.58-0.98; accuracy = 76.2%). CONCLUSION S100A8/A9 may be a potential biomarker for treatment response in HL independent of inflammation. This is the first study to investigate and show this finding. However, further large-scale studies are still required.
Collapse
Affiliation(s)
- Şeyma Şumnu
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Özgür Mehtap
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Sinan Mersin
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Tayfur Toptaş
- Department of Hematology, Marmara University Hospital, Istanbul, Turkey
| | - Gözde Görür
- Department of Nuclear Medicine, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Ayfer Gedük
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Serkan Ünal
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Merve Gökçen Polat
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Kemal Aygün
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Emel Merve Yenihayat
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Hayrunnisa Albayrak
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | - Meral Uluköylü Mengüç
- Department of Hematology, Medical Faculty, Bolu İzzet Baysal University, Bolu, Turkey
| | - Pınar Tarkun
- Department of Hematology, Medical Faculty, University of Kocaeli, Kocaeli, Turkey
| | | |
Collapse
|
12
|
Easier and more explanatory indices by integrating leukocyte lymphocyte ratio (LLR) and prognostic nutritional index (PNI) to IPS systems in cases with classical Hodgkin lymphoma. Leuk Res 2021; 107:106586. [PMID: 34082249 DOI: 10.1016/j.leukres.2021.106586] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 01/04/2023]
Abstract
The aim of this study is to determine the power of he international prognostic scoring systems (IPS-7 and IPS-3) and to obtain indices by integrating leukocyte lymphocyte ratio (LLR) and prognostic nutritional index (PNI) factors as prognostic indicators in cases with classical Hodgkin lymphoma (cHL). 1012 patients with cHL were evaluated with 2 different IPS-4 scores with four parameters: stage, age, hemoglobin level, and either LLR or PNI. Statistical package SPSS v 22.0 was used. Two different Cox regression models were obtained for OS and PFS. Model 1 showed LLR ≥ 5,8 as the highest risk for OS and anemia as the highest risk for PFS. Model 2 showed PNI ≤ 45,2 as the highest risk for OS and anemia as the highest risk for PFS. IPS-4 scores obtained by integrating either LLR or PNI to IPS-3 integration of a biologic parameter either LLR or PNI need to be determined with clinical risk scoring parameters.
Collapse
|
13
|
Gibb A, Pirrie SJ, Linton K, Warbey V, Paterson K, Davies AJ, Collins GP, Menne T, McKay P, Fields PA, Miall FM, Nagy E, Wheatley K, Reed R, Baricevic-Jones I, Barrington S, Radford J. Results of a UK National Cancer Research Institute Phase II study of brentuximab vedotin using a response-adapted design in the first-line treatment of patients with classical Hodgkin lymphoma unsuitable for chemotherapy due to age, frailty or comorbidity (BREVITY). Br J Haematol 2021; 193:63-71. [PMID: 32926420 DOI: 10.1111/bjh.17073] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/04/2020] [Indexed: 11/25/2022]
Abstract
Standard treatment for classical Hodgkin lymphoma (cHL) is poorly tolerated in older patients and results disappointing. We assessed safety and efficacy of brentuximab vedotin (BV), in previously untreated patients with cHL unfit for standard treatment due to age, frailty or comorbidity. The primary outcome was complete metabolic response (CMR) by positron emission tomography/computed tomography after four BV cycles (PET4). The secondary outcomes included progression-free survival (PFS), overall survival (OS), and toxicity. In all, 35 patients with a median age of 77 years and median total Cumulative Illness Rating Scale for Geriatrics (CIRS-G) score of 6 were evaluable for toxicity and 31 for response. A median of four cycles were given (range one-16). In all, 14 patients required dose reduction due to toxicity and 11 patients stopped treatment due to adverse events (AEs). A total of 716 AEs were reported, of which 626 (88%) were Grade 1/2 and 27 (77%) patients had at least one AE Grade ≥3. At PET4, CMR was 25·8% [95% confidence interval (CI) 13·7-42.2%] and objective response rate 83·9% (95% CI 63·7-90·8%). Median PFS was 7·3 months (95% CI 5·2-9·0), and OS 19·5 months. Our results suggest that BV monotherapy is tolerable but suboptimal in the front-line therapy of elderly or comorbid patients with cHL. Combining BV with other agents may be more effective. Trial Registration: Clinicaltrials.gov identifier: NCT02567851.
Collapse
Affiliation(s)
- Adam Gibb
- National Institute for Health Research (NIHR) Manchester Clinical Research Facility, The Christie NHS Foundation Trust, Manchester, UK
| | - Sarah J Pirrie
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, UK
| | - Kim Linton
- National Institute for Health Research (NIHR) Manchester Clinical Research Facility, The Christie NHS Foundation Trust, Manchester, UK
- University of Manchester, Manchester, UK
| | - Victoria Warbey
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, London, UK
| | - Kathryn Paterson
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, UK
| | - Andrew J Davies
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Graham P Collins
- Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, UK
| | | | - Pamela McKay
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | | | | | - Eszter Nagy
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, UK
| | - Keith Wheatley
- Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, UK
| | - Rachel Reed
- Stoller Biomarker Discovery Centre, University of Manchester, Manchester, UK
| | | | - Sally Barrington
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, London, UK
| | - John Radford
- National Institute for Health Research (NIHR) Manchester Clinical Research Facility, The Christie NHS Foundation Trust, Manchester, UK
- University of Manchester, Manchester, UK
| |
Collapse
|
14
|
Veldman J, Alsada ZND, van den Berg A, Plattel WJ, Diepstra A, Visser L. Soluble PD-L1 is a promising disease biomarker but does not reflect tissue expression in classic Hodgkin lymphoma. Br J Haematol 2021; 193:506-514. [PMID: 33620088 PMCID: PMC8247981 DOI: 10.1111/bjh.17362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/13/2021] [Indexed: 12/12/2022]
Abstract
Individually, tissue and soluble markers involved in the programmed cell death protein 1/programmed death-ligand (PD-1/PD-L) axis have been described as biomarkers with clinical value in classical Hodgkin lymphoma (cHL). In the context of the success of immune checkpoint blockade therapy in cHL, it is interesting to discover whether plasma levels of proteins in the PD-1/PD-L axis are a reflection of expression by the corresponding tissue. Paired tissue and plasma samples of cHL patients were collected and analysed for PD-1, PD-L1 and PD-L2 levels. In addition, vascular endothelial growth factor (VEGF) and CD83, molecules regarded to influence the expression of PD-1, PD-L1 and/or PD-L2, were included. PD-L1 was upregulated in the plasma of cHL patients compared to healthy controls and correlated well with several clinical parameters. Strong PD-L1 expression in the tumour microenvironment contributed to high soluble (s)PD-L1 levels, although there was no direct correlation between plasma PD-L1 levels and total expression of PD-L1 in corresponding cHL tissue. Interestingly, we observed a positive correlation between VEGF and PD-1 levels in both tissue and plasma. In conclusion, although PD-L1 is a promising soluble biomarker in cHL, its levels do not reflect the total tissue expression. Future studies focusing on PD-L1 as a predictor for immune checkpoint treatment response, should include both biopsy and plasma samples.
Collapse
Affiliation(s)
- Johanna Veldman
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Zainab N D Alsada
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wouter J Plattel
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Lydia Visser
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
15
|
Zijtregtop EAM, van der Strate I, Beishuizen A, Zwaan CM, Scheijde-Vermeulen MA, Brandsma AM, Meyer-Wentrup F. Biology and Clinical Applicability of Plasma Thymus and Activation-Regulated Chemokine (TARC) in Classical Hodgkin Lymphoma. Cancers (Basel) 2021; 13:884. [PMID: 33672548 PMCID: PMC7923750 DOI: 10.3390/cancers13040884] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 01/05/2023] Open
Abstract
Thymus and activation-regulated chemokine (TARC) is produced by different cell types and is highly expressed in the thymus. It plays an important role in T cell development, trafficking and activation of mature T cells after binding to its receptor C-C chemokine receptor type 4 (CCR4) and consecutive signal transducer and activator of transcription 6 (STAT6) activation. Importantly, TARC is also produced by malignant Hodgkin and Reed-Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL). In cHL, HRS cells survive and proliferate due to the micro-environment consisting primarily of type 2 T helper (Th2) cells. TARC-mediated signaling initiates a positive feedback loop that is crucial for the interaction between HRS and T cells. The clinical applicability of TARC is diverse. It is useful as diagnostic biomarker in both children and adults with cHL and in other Th2-driven diseases. In adult cHL patients, TARC is also a biomarker for treatment response and prognosis. Finally, blocking TARC signaling and thus inhibiting pathological Th2 cell recruitment could be a therapeutic strategy in cHL. In this review, we summarize the biological functions of TARC and focus on its role in cHL pathogenesis and as a biomarker for cHL and other diseases. We conclude by giving an outlook on putative therapeutic applications of antagonists and inhibitors of TARC-mediated signaling.
Collapse
Affiliation(s)
- Eline A. M. Zijtregtop
- Department of Pediatric Hematology and Oncology, Erasmus Medical Center-Sophia Children’s Hospital, 3015 GD Rotterdam, The Netherlands; (E.A.M.Z.); (A.B.); (C.M.Z.)
- Department of Pediatric Hemato-oncology, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (I.v.d.S.); (A.M.B.)
| | - Iris van der Strate
- Department of Pediatric Hemato-oncology, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (I.v.d.S.); (A.M.B.)
| | - Auke Beishuizen
- Department of Pediatric Hematology and Oncology, Erasmus Medical Center-Sophia Children’s Hospital, 3015 GD Rotterdam, The Netherlands; (E.A.M.Z.); (A.B.); (C.M.Z.)
- Department of Pediatric Hemato-oncology, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (I.v.d.S.); (A.M.B.)
| | - Christian M. Zwaan
- Department of Pediatric Hematology and Oncology, Erasmus Medical Center-Sophia Children’s Hospital, 3015 GD Rotterdam, The Netherlands; (E.A.M.Z.); (A.B.); (C.M.Z.)
- Department of Pediatric Hemato-oncology, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (I.v.d.S.); (A.M.B.)
| | | | - Arianne M. Brandsma
- Department of Pediatric Hemato-oncology, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (I.v.d.S.); (A.M.B.)
| | - Friederike Meyer-Wentrup
- Department of Pediatric Hemato-oncology, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (I.v.d.S.); (A.M.B.)
| |
Collapse
|
16
|
Defining the Inflammatory Plasma Proteome in Pediatric Hodgkin Lymphoma. Cancers (Basel) 2020; 12:cancers12123603. [PMID: 33276546 PMCID: PMC7761312 DOI: 10.3390/cancers12123603] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 01/08/2023] Open
Abstract
Simple Summary Hodgkin lymphoma (HL) is a common type of cancer that is characterized by rare, malignant cells among an inflammatory microenvironment. Specific systemic, inflammatory plasma proteins have demonstrated prognostic significance in adult HL; however, systemic inflammation has not been well-characterized in childhood HL. The aim of our study was to better define the inflammatory pre-therapy plasma proteome and identify plasma proteins associated with clinical features of childhood HL. We measured plasma concentrations of 135 proteins in 56 pediatric subjects with newly diagnosed HL and 47 healthy pediatric controls. We found that the plasma protein profile was distinct from controls, and unique proteins were associated with high-risk disease (IL-10, TNF-α, IFN-γ, IL-8), slow early therapy response (CCL13, IFN-λ1, IL-8), and relapse (TNFSF10). These proteins could be used to improve risk stratification, and thus optimize outcomes and minimize unnecessary toxic exposures for those with childhood HL. Abstract Hodgkin lymphoma (HL) histopathology is characterized by rare malignant Reed–Sternberg cells among an inflammatory infiltrate. We hypothesized that characteristics of inflammation in pediatric HL lesions would be reflected by the levels of inflammatory cytokines or chemokines in pre-therapy plasma of children with HL. The study objectives were to better define the inflammatory pre-therapy plasma proteome and identify plasma biomarkers associated with extent of disease and clinical outcomes in pediatric HL. Pre-therapy plasma samples were obtained from pediatric subjects with newly diagnosed HL and healthy pediatric controls. Plasma concentrations of 135 cytokines/chemokines were measured with the Luminex platform. Associations between protein concentration and disease characteristics were determined using multivariate permutation tests with false discovery control. Fifty-six subjects with HL (mean age: 13 years, range 3–18) and 47 controls were analyzed. The cytokine/chemokine profiles of subjects with HL were distinct from controls, and unique cytokines/chemokines were associated with high-risk disease (IL-10, TNF-α, IFN-γ, IL-8) and slow early response (CCL13, IFN-λ1, IL-8). TNFSF10 was significantly elevated among those who ultimately relapsed and was significantly associated with worse event-free survival. These biomarkers could be incorporated into biologically based risk stratification to optimize outcomes and minimize toxicities in pediatric HL.
Collapse
|
17
|
Viviani S, Mazzocchi A, Pavoni C, Taverna F, Rossi A, Patti C, Romano A, Trentin L, Sorasio R, Guidetti A, Gottardi D, Tarella C, Cimminiello M, Zanotti R, Farina L, Ferreri AJM, Galbiati M, Corradini P, Gianni AM, Gallamini A, Rambaldi A. Early serum TARC reduction predicts prognosis in advanced-stage Hodgkin lymphoma patients treated with a PET-adapted strategy. Hematol Oncol 2020; 38:501-508. [PMID: 32602970 DOI: 10.1002/hon.2775] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/11/2022]
Abstract
Among patients with advanced-stage classical Hodgkin lymphoma (cHL) receiving ABVD chemotherapy, PET performed after the first two treatment cycles (PET-2) has prognostic value. However, 15% of patients with a negative PET-2 will experience treatment failure. Here we prospectively evaluated serum thymus and activation-regulated chemokine (TARC) levels, to improve risk assessment in patients treated according to HD0607 PET-driven trial (#NCT00795613). In 266 patients with available serum samples, who have agreed to participate in a sub-study for assessment of the role of TARC monitoring, serum TARC levels were measured at baseline and at time of PET-2 by commercially available ELISA test kits. The primary end-point was to evaluate the association between TARC after 2 ABVD cycles and PFS. Median TARC-2 values were significantly higher in PET-2-positive patients compared to PET-2-negative patients (P = .001), and in patients with treatment failure compared to those in continuous CR (P = .01). The 4-year PFS significantly differed between patients with TARC-2 >800 pg/mL vs ≤800 pg/mL (64% vs 86%, P = .0001). Moreover, among PET-2-negative patients, elevated TARC-2 identified those with a worse prognosis (74% vs 89%; P = .01). In multivariable analysis, TARC-2 >800 pg/mL was a significant independent predictor of PFS in the whole study population (HR 2.39, P = .004) and among the PET-2-negative patients (HR 2.49, P = .02). In conclusion, our results indicate that TARC-2 serum levels above 800 pg/mL suggest the need for a stringent follow-up in PET-2-negative patients, and the evaluation of new drugs in PET-2-positive, who will likely fail to respond to intensification with escalated BEACOPP.
Collapse
Affiliation(s)
- Simonetta Viviani
- Department of Hemato-Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Arabella Mazzocchi
- Immuno-hematology and Transfusion Medicine Service, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara Pavoni
- Department of Oncology and Hematology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Francesca Taverna
- Immuno-hematology and Transfusion Medicine Service, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Rossi
- Department of Oncology and Hematology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Caterina Patti
- Division of Hematology 1, Azienda Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Alessandra Romano
- Department of Medical and Surgery Science, Hematology Unit, University of Catania, Catania, Italy
| | - Livio Trentin
- Department of Hematology, University of Padova, Padova, Italy
| | - Roberto Sorasio
- Division of Hematology, Azienda Ospedaliera Santa Croce e Carle, Cuneo, Italy
| | - Anna Guidetti
- Department of Hemato-Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Daniela Gottardi
- Division of Hematology, Ospedale Mauriziano Umberto I di Torino, Turin, Italy
| | - Corrado Tarella
- Hemato-Oncology Division, European Institute of Oncology IRCCS, Milan, Italy.,University Department "Scienze della Salute" (DISS), University of Milan, Milan, Italy
| | | | - Roberta Zanotti
- Hematology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Lucia Farina
- Department of Hemato-Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrés José Maria Ferreri
- Lymphoma Unit, Department of Onco-Hematology, IRCCS Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Marina Galbiati
- Immuno-hematology and Transfusion Medicine Service, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Corradini
- Department of Hemato-Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Andrea Gallamini
- Division of Hematology, Azienda Ospedaliera Santa Croce e Carle, Cuneo, Italy
| | - Alessandro Rambaldi
- Department of Oncology and Hematology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy.,Department of Oncology and Hematology, University of Milan, Milan, Italy
| |
Collapse
|
18
|
Zijtregtop EAM, Meyer‐Wentrup F, Wong W, Hoogendijk R, Lopez‐Yurda M, Zwaan CM, Beishuizen A. Plasma thymus and activation-regulated chemokine (TARC) as diagnostic marker in pediatric Hodgkin lymphoma. EJHAEM 2020; 1:152-160. [PMID: 35847691 PMCID: PMC9176129 DOI: 10.1002/jha2.41] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022]
Abstract
Pediatric classical Hodgkin's lymphoma (cHL) is characterized by Hodgkin Reed-Sternberg cells located in an inflammatory microenvironment. Blood biomarkers result from active crosstalk between these cells. One promising biomarker in adult cHL patients is "thymus-and-activation-regulated chemokine" (TARC). The objectives of this study were to define normal TARC values in non-cHL children and to investigate and correlate pretherapy TARC as diagnostic marker in pediatric cHL. In this multicenter prospective study, plasma and serum samples were collected of newly diagnosed cHL patients before start of treatment (n = 49), and from randomly selected non-cHL patients (n = 81). TARC levels were measured by enzyme-linked immunosorbent assay. The non-cHL patients had a median plasma TARC value of 71 pg/mL (range: 18-762), compared to 14 619 pg/mL (range: 380-73 174) in cHL patients (P < .001). TARC values had a high discriminatory power (AUC = .999; 95% confidence interval, .998-1). A TARC cutoff level of 942 pg/mL maximized the sum of sensitivity (97.9%) and specificity (100%). TARC plasma levels were associated with age, treatment level, bulky disease, B-symptoms, and erythrocyte sedimentation rate. TARC was found to be a highly specific and sensitive diagnostic marker for pediatric cHL. This noninvasive marker could be of great value as screening test in the work-up for pediatric patients with lymphadenopathy.
Collapse
Affiliation(s)
- Eline A. M. Zijtregtop
- Department of Pediatric Hematology and OncologyErasmus Medical Centre ‐ Sophia Children's HospitalRotterdamThe Netherlands
- Department of Hemato‐oncologyPrincess Máxima Centre for Pediatric OncologyUtrechtThe Netherlands
| | - Friederike Meyer‐Wentrup
- Department of Hemato‐oncologyPrincess Máxima Centre for Pediatric OncologyUtrechtThe Netherlands
| | - Wai‐Chu Wong
- Department of Pediatric Hematology and OncologyErasmus Medical Centre ‐ Sophia Children's HospitalRotterdamThe Netherlands
| | - Raoull Hoogendijk
- Department of Hemato‐oncologyPrincess Máxima Centre for Pediatric OncologyUtrechtThe Netherlands
| | - Marta Lopez‐Yurda
- Department of Hemato‐oncologyPrincess Máxima Centre for Pediatric OncologyUtrechtThe Netherlands
- Department of BiometricsNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Christian M. Zwaan
- Department of Pediatric Hematology and OncologyErasmus Medical Centre ‐ Sophia Children's HospitalRotterdamThe Netherlands
- Department of Hemato‐oncologyPrincess Máxima Centre for Pediatric OncologyUtrechtThe Netherlands
| | - Auke Beishuizen
- Department of Pediatric Hematology and OncologyErasmus Medical Centre ‐ Sophia Children's HospitalRotterdamThe Netherlands
- Department of Hemato‐oncologyPrincess Máxima Centre for Pediatric OncologyUtrechtThe Netherlands
| |
Collapse
|
19
|
Abstract
Introduction: Hodgkin Lymphoma (HL) carries an overall excellent prognosis for young patients treated with multimodal therapy. Predicting an individual patient's prognosis is currently heavily dependent on imaging modalities such as Positron Emission Tomography (PET).Areas covered: Potential biomarkers from serum, tissue, circulating nucleic acids and non-tumor derived cells have all been reported to be of prognostic relevance in HL. We review a range of these biomarkers and discuss the integration of new biomarkers into individualized patient care.Expert opinion: Better prognostic markers are needed to predict an individuals response to HL therapy. Interim PET-scan improves the ability to predict long-term treatment responders. However, it is our opinion that supplementation of PET results with additional biomarkers (including circulating tumor DNA, protein biomarkers, tissue genotyping and metabolic tumor volume) are likely to improve risk stratification for future patients with HL.
Collapse
Affiliation(s)
- Melita Cirillo
- Faculty of Medicine and University Hospital of Cologne, Department I Of Internal Medicine, GHSG, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Centre for Molecular Medicine, University of Cologne, Cologne, Germany.,Department of Hematology, Royal Perth Hospital, Perth, Australia.,University of Western Australia, Perth, Australia
| | - Sven Borchmann
- Faculty of Medicine and University Hospital of Cologne, Department I Of Internal Medicine, GHSG, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Centre for Molecular Medicine, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Else Kröner Forschungskolleg Clonal Evolution in Cancer, University of Cologne, Cologne, Germany
| |
Collapse
|
20
|
Kahle XU, Montes de Jesus FM, Kwee TC, van Meerten T, Diepstra A, Rosati S, Glaudemans AWJM, Noordzij W, Plattel WJ, Nijland M. Relationship between semiquantitative 18F-fluorodeoxyglucose positron emission tomography metrics and necrosis in classical Hodgkin lymphoma. Sci Rep 2019; 9:11073. [PMID: 31363153 PMCID: PMC6667466 DOI: 10.1038/s41598-019-47453-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 07/16/2019] [Indexed: 11/08/2022] Open
Abstract
Semiquantitative 18F-fluoro-2-deoxy-D-glucose positron emission tomography (18F-FDG PET) parameters have been proposed as prognostic markers in classical Hodgkin lymphoma (cHL). In non-Hodgkin lymphoma necrosis as assessed by 18F-FDG PET or computed tomography (CT) (necrosisvisual) correlates with an adverse prognosis. We investigated whether semiquantitative 18F-FDG PET metrics correlate with necrosisvisual, determined the incidence of necrosisvisual and explored the prognostic impact of these factors in cHL. From 87 cHL cases treated with ABVD, (escalated) BEACOPP or CHOP chemotherapy between 2010 and 2017, 71 had both a NEDPAS/EARL accredited 18F-FDG PET and a contrast enhanced CT scan. Semiquantitative 18F-FDG PET parameters were determined using Hermes Hybrid 3D software. Necrosisvisual, defined by photopenic tumor areas on 18F-FDG PET and attenuation values between 10 and 30 Hounsfield units (HUs) on CT, was assessed blinded to outcome. Univariate Cox regression survival analyses of progression free survival (PFS) were performed. Necrosisvisual was observed in 18.3% of cHL patients. Bulky disease (tumor mass >10 cm in any direction) (P = 0.002) and TLG (P = 0.041) but no other semiquantitative parameters were significantly associated with necrosisvisual. In exploratory univariate survival analysis for PFS the covariates IPS, bulky disease, MTV and TLG were prognostic, while necrosisvisual was not.
Collapse
Affiliation(s)
- X U Kahle
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - F M Montes de Jesus
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - T C Kwee
- Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - T van Meerten
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - S Rosati
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - W Noordzij
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - W J Plattel
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M Nijland
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
21
|
Serum levels of TARC, MDC, IL-10, and soluble CD163 in Hodgkin lymphoma: a SWOG S0816 correlative study. Blood 2019; 133:1762-1765. [PMID: 30723079 DOI: 10.1182/blood-2018-08-870915] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 01/22/2019] [Indexed: 11/20/2022] Open
Abstract
Serum soluble chemokines/cytokines produced by Hodgkin cells and the tumor microenvironment might be of value as biomarkers in classic Hodgkin lymphoma (cHL). We assessed serum thymus and activation-related chemokine (TARC), macrophage-derived chemokine (MDC), interleukin-10 (IL-10), and soluble CD163 (sCD163) levels at baseline, time of interim fluorodeoxyglucose positron emission tomography (PET), and after therapy in cHL patients treated on S0816, an intergroup phase 2 response-adapted study evaluating escalated therapy for interim PET (PET2)-positive patients (www.clinicaltrials.gov #NCT00822120). Epstein-Barr virus (EBV) status was assessed, and 559 serum samples were evaluated for TARC, MDC, IL-10, and sCD163 by immunoassay. EBV positivity correlated with higher sCD163 and IL-10 levels but lower TARC levels. While baseline biomarker levels were not associated with outcome, sCD163 levels at the time of PET2 were associated with favorable progression-free survival (PFS), adjusting for PET2 status. After therapy TARC, MDC, and IL-10 correlated with PFS and overall survival (OS) on univariable analysis, which remained significant adjusting for international prognostic score. When also adjusting for end-of-therapy PET results, TARC and IL-10 remained significantly associated with shorter PFS and OS. Exploratory analysis in PET2-negative patients showed that elevated posttherapy TARC and IL-10 levels were associated with PFS. Serum cytokine levels correlate with outcome in cHL and should be investigated further in risk-adapted cHL trials.
Collapse
|
22
|
Cirillo M, Reinke S, Klapper W, Borchmann S. The translational science of hodgkin lymphoma. Br J Haematol 2018; 184:30-44. [DOI: 10.1111/bjh.15658] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/20/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Melita Cirillo
- Department of Haematology; Royal Perth Hospital; Perth Australia
- Department I of Internal Medicine; German Hodgkin Study Group (GHSG); Cologne Germany
| | - Sarah Reinke
- Department of Pathology; Hematopathology Section; University Hospital Schleswig-Holstein, Campus Kiel; Kiel Germany
| | - Wolfram Klapper
- Department of Pathology; Hematopathology Section; University Hospital Schleswig-Holstein, Campus Kiel; Kiel Germany
| | - Sven Borchmann
- Department I of Internal Medicine; German Hodgkin Study Group (GHSG); Cologne Germany
- Else Kröner Forschungskolleg Clonal Evolution in Cancer; University Hospital of Cologne; Cologne Germany
- Centre for Molecular Medicine Cologne; University of Cologne; Cologne Germany
| |
Collapse
|
23
|
Dörsam B, Bösl T, Reiners KS, Barnert S, Schubert R, Shatnyeva O, Zigrino P, Engert A, Hansen HP, von Strandmann EP. Hodgkin Lymphoma-Derived Extracellular Vesicles Change the Secretome of Fibroblasts Toward a CAF Phenotype. Front Immunol 2018; 9:1358. [PMID: 29967610 PMCID: PMC6015880 DOI: 10.3389/fimmu.2018.01358] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/31/2018] [Indexed: 12/21/2022] Open
Abstract
Secretion of extracellular vesicles (EVs) is a ubiquitous mechanism of intercellular communication based on the exchange of effector molecules, such as growth factors, cytokines, and nucleic acids. Recent studies identified tumor-derived EVs as central players in tumor progression and the establishment of the tumor microenvironment (TME). However, studies on EVs from classical Hodgkin lymphoma (cHL) are limited. The growth of malignant Hodgkin and Reed–Sternberg (HRS) cells depends on the TME, which is actively shaped by a complex interaction of HRS cells and stromal cells, such as fibroblasts and immune cells. HRS cells secrete cytokines and angiogenic factors thus recruiting and inducing the proliferation of surrounding cells to finally deploy an immunosuppressive TME. In this study, we aimed to investigate the role of tumor cell-derived EVs within this complex scenario. We observed that EVs collected from Hodgkin lymphoma (HL) cells were internalized by fibroblasts and triggered their migration capacity. EV-treated fibroblasts were characterized by an inflammatory phenotype and an upregulation of alpha-smooth muscle actin (α-SMA), a marker of cancer-associated fibroblasts. Analysis of the secretome of EV-treated fibroblast revealed an enhanced release of pro-inflammatory cytokines (e.g., IL-1α, IL-6, and TNF-α), growth factors (G-CSF and GM-CSF), and pro-angiogenic factors such as VEGF. These soluble factors are known to promote HL progression. In line, ingenuity pathway analysis identified inflammatory pathways, including TNF-α/NF-κB-signaling, as key factors directing the EV-dependent phenotype changes in fibroblasts. Confirming the in vitro data, we demonstrated that EVs promote α-SMA expression in fibroblasts and the expression of proangiogenic factors using a xenograft HL model. Collectively, we demonstrate that HL EVs alter the phenotype of fibroblasts to support tumor growth, and thus shed light on the role of EVs for the establishment of the tumor-promoting TME in HL.
Collapse
Affiliation(s)
- Bastian Dörsam
- Clinic for Hematology, Oncology and Immunology, Experimental Tumor Research, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Teresa Bösl
- Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Katrin S Reiners
- Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Sabine Barnert
- Department of Pharmaceutical Technology and Biopharmacy, Albert-Ludwigs-University, Freiburg, Germany
| | - Rolf Schubert
- Department of Pharmaceutical Technology and Biopharmacy, Albert-Ludwigs-University, Freiburg, Germany
| | - Olga Shatnyeva
- Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Paola Zigrino
- Department of Dermatology, University Hospital of Cologne, Cologne, Germany
| | - Andreas Engert
- Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Hinrich P Hansen
- Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Elke Pogge von Strandmann
- Clinic for Hematology, Oncology and Immunology, Experimental Tumor Research, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.,Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
24
|
Spinner MA, Advani RH, Connors JM, Azzi J, Diefenbach C. New Treatment Algorithms in Hodgkin Lymphoma: Too Much or Too Little? Am Soc Clin Oncol Educ Book 2018; 38:626-636. [PMID: 30231319 DOI: 10.1200/edbk_200679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Hodgkin lymphoma treatment continues to evolve as new means of assessing response to treatment, new appreciation of important risk factors, and more effective therapeutic agents become available. Treatment algorithms integrating functional imaging now provide the opportunity to modify therapy during its delivery, allowing adjustment of duration and intensity of chemotherapy and rationale identification of patients who may benefit from the addition of therapeutic irradiation. Novel agents, including the antibody drug conjugate brentuximab vedotin and checkpoint inhibitors such as nivolumab and pembrolizumab can improve the effectiveness of treatment while keeping toxicity within acceptable limits. Carefully designed clinical trials permit the identification of superior approaches in which efficacy is enhanced and toxicity minimized. Clinicians treating patients with Hodgkin lymphoma now have access to novel treatment approaches, which will require detailed assessment of each patient and careful discussion of the goals and risks of treatment at the time of planning primary treatment, again during delivery of that treatment as data indicating ongoing effectiveness become available, at the conclusion of initial intervention, and, when the need arises, at the time of recurrence of disease.
Collapse
Affiliation(s)
- Michael A Spinner
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Ranjana H Advani
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Joseph M Connors
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Jacques Azzi
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Catherine Diefenbach
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| |
Collapse
|
25
|
Lim SH, Johnson PWM. Optimizing therapy in advanced-stage Hodgkin lymphoma. Blood 2018; 131:1679-1688. [PMID: 29500173 DOI: 10.1182/blood-2017-09-772640] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/25/2017] [Indexed: 02/07/2023] Open
Abstract
The treatment of Hodgkin lymphoma has evolved continuously since the introduction of extended-field radiotherapy in the 1960s to involved-field and then involved-node radiotherapy, multiagent chemotherapy, combined chemoradiotherapy, risk-adapted and response-adapted modulation, and, most recently, introduction of antibody-drug conjugates and immune checkpoint-blocking antibodies. These changes have translated into progressively increasing cure rates, so that 10-year survival figures now exceed 80%, compared with <50% 40 years ago. The challenge now is how to improve upon success while maintaining, or if possible improving, the quality of life for survivors. Steering between undertreatment, with the risk of avoidable recurrences, and overtreatment, with the risk of unnecessary toxicity, remains complex because control of the lymphoma and the probability of survival are no longer closely linked. This requires trials with long follow-up and continuous reappraisal of the interaction between the illness; the method used to define risk, and the type of treatment involved. One important factor in this is age: outcomes in older patients have not improved at the same rate as those in the population under 60 years of age, reflecting the need for different approaches. Recently, treatment has moved from being primarily risk-based, using baseline characteristics such as anatomical stage and severity of the illness, to a more dynamic approach that takes account of the response to therapy, using functional imaging to make an early appraisal, with the option to modulate subsequent treatment. The results of several trials indicate that this has advantages, but a combination of risk- and response-adaptation is probably ideal.
Collapse
Affiliation(s)
- Sean H Lim
- Antibody and Vaccine Group and
- Cancer Sciences Unit, Cancer Research UK Centre, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Peter W M Johnson
- Cancer Sciences Unit, Cancer Research UK Centre, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
26
|
O'Connor OA, Lue JK, Sawas A, Amengual JE, Deng C, Kalac M, Falchi L, Marchi E, Turenne I, Lichtenstein R, Rojas C, Francescone M, Schwartz L, Cheng B, Savage KJ, Villa D, Crump M, Prica A, Kukreti V, Cremers S, Connors JM, Kuruvilla J. Brentuximab vedotin plus bendamustine in relapsed or refractory Hodgkin's lymphoma: an international, multicentre, single-arm, phase 1-2 trial. Lancet Oncol 2017; 19:257-266. [PMID: 29276022 PMCID: PMC9098158 DOI: 10.1016/s1470-2045(17)30912-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 11/26/2022]
Abstract
Background: The major objective of this study was to explore the safety and clinical activity of Brentuximab vedotin (Bv) and bendamustine in combination in patients with relapsed or refractory Hodgkin Lymphoma. Bv produces high response rates and durable progression-free survival (PFS) in CD30-expressing lymphomas and is approved for the treatment of relapsed Hodgkin lymphoma (HL) and relapsed ALCL. Bendamustine (B) is active agent across the lymphoproliferative malignancies, though the PFS among patients with HL and PTCL is modest. Methods: This was an international, multicenter, single-arm, Phase 1–2 study of BvB in patients with relapsed or refractory HL and ALCL. Eligible patients were required to have relapsed/refractory CD30+ biopsy proven HL or ALCL and an ECOG Performance Status ≤2. In the Phase 1, HL patients were deemed eligible if they developed progressive disease following or after declining ASCT, or had at least 2 prior multi-agent chemotherapy regimens. In the Phase 2, patients with HL were eligible if they had relapsed or refractory disease after one line of therapy. Eligible ALCL patients were required to have relapsed after at least one prior multi-agent chemotherapy regimen and if they were not eligible for or have declined ASCT. The primary objective of the Phase I portion of this study was to identify the maximum tolerated dose (MTD) and dose limiting toxicity (DLT). The primary endpoint of the Phase 2 portion was to determine the overall response rate (ORR; complete response [CR] plus partial response [PR])) based on an intention to treat analysis (ITT). Secondary objectives of Phase 1–2 included assessing for duration of response, progression free survival and overall survival. Response was evaluated using International Harmonization Project Group 2007 Revised Response Criteria. Bv was escalated from 1.2mg/kg Day 1, and B from 70mg/m2 Days 1 and 2 every 21 days until the MTD or recommended phase 2 dose (RP2D) was reached. The study is ongoing but no longer recruiting patients. This trial is registered with ClinicalTrials.gov number NCT01657331. Findings: 65 patients (only 1 ALCL) were treated, 28 on the Phase 1 and 37 on the phase 2. While the MTD of the combination was not reached, the single agent MTD of Bv (1.8mg/kg Day 1) and RP2D of B (90mg/m2 Days 1 and 2) were identified as the RP2D of the combination. Patients were heavily treated, 65% (42 of 65) had an autologous or allogeneic stem cell transplant or both. The Phase 1 revealed modest toxicity. The major Grade 3/4 toxicities included Grade 3 lung infection in 5 (14%) patients in the Phase 2, and Grade 3/4 neutropenia in 13 (24%) patients across the Phase 1 and 2. The Phase 1 and 2 overall response rates (ORR) were 61% and 78% respectively, with 43% (16 of 27) patients treated in the Phase 2 attaining a complete remission (CR). In the Phase 2, the median PFS has not been reached and duration of response (DOR) was 3.4 months. There was a total of 23 deaths with 21 due to progression of disease, 2 occurring after being transplanted, and none of which were treatment related. Interpretation: This demonstrates that BvB might be an effective salvage regimen for patients with HL, with a favorable safety profile. Funding: Seattle Genetics, The Lymphoma Research Fund of Columbia University and National Center for Advancing Translational Sciences, National Institutes of Health, through Grant Number UL1TR001873 provided support for this investigator initiated sponsored trial. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.
Collapse
Affiliation(s)
- Owen A O'Connor
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA.
| | - Jennifer K Lue
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Ahmed Sawas
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Jennifer E Amengual
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Changchun Deng
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Matko Kalac
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Lorenzo Falchi
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Enrica Marchi
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Ithamar Turenne
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Renee Lichtenstein
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Celeste Rojas
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Mark Francescone
- Department of Radiology, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Lawrence Schwartz
- Department of Radiology, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Bin Cheng
- Department of Biostatistics, Mailman School of Public Health, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA; Clinical Translational Research Center, Laboratory of Analytical Pharmacology, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Kerry J Savage
- BC Cancer Agency, Centre for Lymphoid Cancer, Vancouver, BC, Canada
| | - Diego Villa
- BC Cancer Agency, Centre for Lymphoid Cancer, Vancouver, BC, Canada
| | - Michael Crump
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Anca Prica
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - Serge Cremers
- Department of Biostatistics, Mailman School of Public Health, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA; Clinical Translational Research Center, Laboratory of Analytical Pharmacology, Columbia University Medical Center-College of Physicians and Surgeons, New York, NY, USA
| | - Joseph M Connors
- BC Cancer Agency, Centre for Lymphoid Cancer, Vancouver, BC, Canada
| | | |
Collapse
|
27
|
Moskowitz AJ, Schöder H, Gavane S, Thoren KL, Fleisher M, Yahalom J, McCall SJ, Cadzin BR, Fox SY, Gerecitano J, Grewal R, Hamlin PA, Horwitz SM, Kumar A, Matasar M, Ni A, Noy A, Palomba ML, Perales MA, Portlock CS, Sauter C, Straus D, Younes A, Zelenetz AD, Moskowitz CH. Prognostic significance of baseline metabolic tumor volume in relapsed and refractory Hodgkin lymphoma. Blood 2017; 130:2196-2203. [PMID: 28874350 PMCID: PMC5691245 DOI: 10.1182/blood-2017-06-788877] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022] Open
Abstract
Identification of prognostic factors for patients with relapsed/refractory Hodgkin lymphoma (HL) is essential for optimizing therapy with risk-adapted approaches. In our phase 2 study of positron emission tomography (PET)-adapted salvage therapy with brentuximab vedotin (BV) and augmented ifosfamide, carboplatin, and etoposide (augICE), we assessed clinical factors, quantitative PET assessments, and cytokine and chemokine values. Transplant-eligible patients with relapsed/refractory HL received 2 (cohort 1) or 3 (cohort 2) cycles of weekly BV; PET-negative patients (Deauville score ≤2) proceeded to autologous stem cell transplantation (ASCT) whereas PET-positive patients received augICE before ASCT. Serum cytokine and chemokine levels were measured at baseline and after BV. Metabolic tumor volume (MTV) and total lesion glycolysis were measured at baseline, after BV, and after augICE. Sixty-five patients enrolled (45, cohort 1; 20, cohort 2); 49 (75%) achieved complete response and 64 proceeded to ASCT. Three-year overall survival and event-free survival (EFS) were 95% and 82%, respectively. Factors predictive for EFS by multivariable analysis were baseline MTV (bMTV) (P < .001) and refractory disease (P = .003). Low bMTV (<109.5 cm3) and relapsed disease identified a favorable group (3-year EFS, 100%). For patients who received a transplant, bMTV and pre-ASCT PET were independently prognostic; 3-year EFS for pre-ASCT PET-positive patients with low bMTV was 86%. In this phase 2 study of PET-adapted therapy with BV and augICE for relapsed/refractory HL, bMTV and refractory disease were independent prognostic factors for EFS. Furthermore, bMTV improved the predictive power of pre-ASCT PET. Future studies should optimize efficacy and tolerability of salvage therapy by stratifying patients according to risk factors such as bMTV.
Collapse
Affiliation(s)
| | - Heiko Schöder
- Nuclear Medicine Department, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Somali Gavane
- Nuclear Medicine Department, Mt. Sinai Hospital, New York, NY; and
| | | | | | | | | | | | | | | | - Ravinder Grewal
- Nuclear Medicine Department, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | - Andy Ni
- Biostatistics Department, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Fülle L, Steiner N, Funke M, Gondorf F, Pfeiffer F, Siegl J, Opitz FV, Haßel SK, Erazo AB, Schanz O, Stunden HJ, Blank M, Gröber C, Händler K, Beyer M, Weighardt H, Latz E, Schultze JL, Mayer G, Förster I. RNA Aptamers Recognizing Murine CCL17 Inhibit T Cell Chemotaxis and Reduce Contact Hypersensitivity In Vivo. Mol Ther 2017; 26:95-104. [PMID: 29103909 PMCID: PMC5763148 DOI: 10.1016/j.ymthe.2017.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 10/04/2017] [Accepted: 10/04/2017] [Indexed: 01/21/2023] Open
Abstract
The chemokine CCL17, mainly produced by dendritic cells (DCs) in the immune system, is involved in the pathogenesis of various inflammatory diseases. As a ligand of CCR4, CCL17 induces chemotaxis and facilitates T cell-DC interactions. We report the identification of two novel RNA aptamers, which were validated in vitro and in vivo for their capability to neutralize CCL17. Both aptamers efficiently inhibited the directed migration of the CCR4+ lymphoma line BW5147.3 toward CCL17 in a dose-dependent manner. To study the effect of these aptamers in vivo, we used a murine model of contact hypersensitivity. Systemic application of the aptamers significantly prevented ear swelling and T cell infiltration into the ears of sensitized mice after challenge with the contact sensitizer. The results of this proof-of-principle study establish aptamers as potent inhibitors of CCL17-mediated chemotaxis. Potentially, CCL17-specific aptamers may be used therapeutically in humans to treat or prevent allergic and inflammatory diseases.
Collapse
Affiliation(s)
- Lorenz Fülle
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Nancy Steiner
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Markus Funke
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany
| | - Fabian Gondorf
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Franziska Pfeiffer
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany
| | - Julia Siegl
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany
| | - Friederike V Opitz
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Silvana K Haßel
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany
| | - Anna Belen Erazo
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Oliver Schanz
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - H James Stunden
- Institute of Innate Immunity, University Hospital Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany
| | - Michael Blank
- AptaIT, Am Klopferspitz 19a, 82152 Planegg-Martinsried, Germany
| | - Carsten Gröber
- AptaIT, Am Klopferspitz 19a, 82152 Planegg-Martinsried, Germany
| | - Kristian Händler
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Sigmund-Freud-Straße 27, 53127 Bonn, Germany
| | - Marc Beyer
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Sigmund-Freud-Straße 27, 53127 Bonn, Germany; Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Straße 27, 53127 Bonn, Germany
| | - Heike Weighardt
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany; Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Sigmund-Freud-Straße 27, 53127 Bonn, Germany
| | - Günter Mayer
- Chemical Biology and Chemical Genetics, Life and Medical Sciences (LIMES) Institute, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany; Centre of Aptamer Research and Development, University of Bonn, Gerhard-Domagk-Straße 1, 53121 Bonn, Germany.
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Straße 31, 53115 Bonn, Germany.
| |
Collapse
|
29
|
Guidetti A, Mazzocchi A, Miceli R, Paterno' E, Taverna F, Spina F, Crippa F, Farina L, Corradini P, Gianni AM, Viviani S. Early reduction of serum TARC levels may predict for success of ABVD as frontline treatment in patients with Hodgkin Lymphoma. Leuk Res 2017; 62:91-97. [PMID: 28992524 DOI: 10.1016/j.leukres.2017.09.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 09/01/2017] [Accepted: 09/25/2017] [Indexed: 01/23/2023]
Abstract
BACKGROUND Many efforts have been made to predict prognosis of newly diagnosed Hodgkin Lymphoma (HL) patients. Objective of this study was to investigate the association between early reduction of Thymus and Activation-Regulated Chemokine after the first ABVD cycle (TARC-1) and prognosis of HL patients. METHODS Serum samples of 116 HL patients were collected at baseline, after every ABVD cycle and during follow-up. The 99th centile of TARC distribution in a group of 156 independent healthy subjects (800pg/ml) was considered as cut-off for discriminating between abnormal and normal TARC values. FINDINGS 101 patients out of 116 had baseline TARC above 800pg/ml (median value 27515pg/ml (IQR, 11001-68139)) and were the object of this analysis. TARC-1 significantly decreased to a median value of 556pg/ml (IQR, 378-977pg/ml). TARC-1 values below 800pg/ml were associated with success of therapy (p=0.0003) and PET-2 negativity (p=0.001). TARC-1≤800pg/ml identified a population with a significantly higher 5-years PFS in the whole cohort (90.1% vs 55.6%; p<0.0001) and in both subgroups of advanced (p=0.003) and early stage patients (p=0.021). At multivariable analysis, TARC-1 was significant independent predictor of PFS (p=0.0035). INTERPRETATION Early reduction of TARC serum levels can predict success of treatment, being associated with achievement of interim PET-2 negative and favorable long-term outcome in HL patients receiving ABVD as front-line therapy.
Collapse
Affiliation(s)
- A Guidetti
- Department of Hematology and Pediatric Onco-Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Oncology and Onco-Hematology, University of Milano, Milan, Italy
| | - A Mazzocchi
- Immunohematology and Transfusion Medicine Service, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - R Miceli
- Department of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - E Paterno'
- Department of Hematology and Pediatric Onco-Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - F Taverna
- Immunohematology and Transfusion Medicine Service, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - F Spina
- Department of Hematology and Pediatric Onco-Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - F Crippa
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - L Farina
- Department of Hematology and Pediatric Onco-Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - P Corradini
- Department of Hematology and Pediatric Onco-Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Oncology and Onco-Hematology, University of Milano, Milan, Italy
| | - A M Gianni
- Department of Hematology and Pediatric Onco-Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S Viviani
- Department of Hematology and Pediatric Onco-Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
30
|
Yu WY, Geng M, Hao J, Chen M, Zhang SJ, Wang J, Mi JQ. Clinical Features and Prognosis Analysis of Hodgkin Lymphoma: A Multicenter Retrospective Study Over a Decade of Patients in China. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:274-282. [PMID: 28292586 DOI: 10.1016/j.clml.2017.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/22/2016] [Accepted: 02/07/2017] [Indexed: 10/20/2022]
Abstract
OBJECTIVE There is little information available regarding Chinese patients with Hodgkin lymphoma (HL). We analyzed the clinical features, outcome, and prognostic factors of Chinese patients with HL, aiming to establish a new risk model for better risk-adapted therapeutic strategy. PATIENTS AND METHODS Patients with newly diagnosed HL at 4 medical centers from January 2000 to August 2014 were recruited. RESULTS A total of 150 patients were reviewed. The median age was 30 years (range, 15-91 years). At completion of initial therapy, 73.65% of patients achieved complete remission. The 5-year event-free survival (EFS) of the entire cohort was 61.1%, the overall survival was 84.7%, and the disease-free survival was 78.8%. B symptoms, extranodal involvement, and International Prognostic Score ≥ 3 remained as independent prognostic factors of EFS. Patients who failed to reach complete remission on interim positron emission tomography/computed tomography or computed tomography had a significantly worse outcome than those who did. A new risk model incorporating traditional risk factors and interim response stratified patients into 3 classes, with a 5-year EFS of 100%, 83.1%, and 33.1%, respectively (P < .0001). CONCLUSIONS General clinical features were comparable with those of Western patients, whereas therapeutic outcomes were slightly inferior. The novel risk assessment model showed potential as a more powerful prognostic tool by identifying 3 subsets of patients with significantly distinct outcomes, which warrants further validations.
Collapse
Affiliation(s)
- Wen-Yan Yu
- Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mei Geng
- Department of Oncology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Hao
- Department of Hematology, Shanghai North Station Hospital, Shanghai, China
| | - Mei Chen
- Department of Hematology, Shanghai Yang Pu Central Hospital Affiliated to Shanghai Tong Ji University, Shanghai, China
| | - Su-Jiang Zhang
- Department of Hematology, Rui Jin North Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Wang
- Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Qing Mi
- Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
31
|
Diefenbach CS, Connors JM, Friedberg JW, Leonard JP, Kahl BS, Little RF, Baizer L, Evens AM, Hoppe RT, Kelly KM, Persky DO, Younes A, Kostakaglu L, Bartlett NL. Hodgkin Lymphoma: Current Status and Clinical Trial Recommendations. J Natl Cancer Inst 2016; 109:2742050. [PMID: 28040700 DOI: 10.1093/jnci/djw249] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/24/2016] [Accepted: 09/26/2016] [Indexed: 12/12/2022] Open
Abstract
The National Clinical Trials Network lymphoid malignancies Clinical Trials Planning Meeting (CTPM) occurred in November of 2014. The scope of the CTPM was to prioritize across the lymphoid tumors clinically significant questions and to foster strategies leading to biologically informed and potentially practice changing clinical trials. This review from the Hodgkin lymphoma (HL) subcommittee of the CTPM discusses the ongoing clinical challenges in HL, outlines the current standard of care for HL patients from early to advanced stage, and surveys the current science with respect to biomarkers and the landscape of ongoing clinical trials. Finally, we suggest areas of unmet need in HL and elucidate promising therapeutic strategies to guide future HL clinical trials planning across the NCTN.
Collapse
Affiliation(s)
- Catherine S Diefenbach
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Joseph M Connors
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Jonathan W Friedberg
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - John P Leonard
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Brad S Kahl
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Richard F Little
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Lawrence Baizer
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Andrew M Evens
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Richard T Hoppe
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Kara M Kelly
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Daniel O Persky
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Anas Younes
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Lale Kostakaglu
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| | - Nancy L Bartlett
- Affiliations of authors: NYU Perlmutter Cancer Center, New York, NY (CSD); BC Cancer Agency Centre for Lymphoid Cancer, Vancouver, BC, Canada (JMC); Wilmot Cancer Center and Division of Hematology/Oncology, University of Rochester Medical Center, Rochester, NY (JWF); Department of Medicine, Weil Cornell University, New York, NY (JPL); Oncology Division, Department of Medicine, Washington University, St. Louis, MO (BSK, NLB); Division of Cancer Treatment and Diagnosis (RFL) and Coordinating Center for Clinical Trials (LB), Tufts Cancer Center and Division of Hematology/Oncology, Tufts University School of Medicine, Boston, MA (AME); Stanford Cancer Institute, Stanford University Medical School, Stanford, CA (RTH); Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Medical Center, New York, NY (KMK); Department of Medicine, University of Arizona Cancer Center, Tucson, AZ (DOP); Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY (AY); Department of Radiology, Mount Sinai Hospital, New York, NY (LK)
| |
Collapse
|
32
|
Combined prognostic value of absolute lymphocyte/monocyte ratio in peripheral blood and interim PET/CT results in Hodgkin lymphoma. Int J Hematol 2016; 103:63-9. [PMID: 26462809 DOI: 10.1007/s12185-015-1884-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 10/02/2015] [Accepted: 10/05/2015] [Indexed: 12/16/2022]
Abstract
Decreased absolute lymphocyte/monocyte ratio (LMR) in peripheral blood has been reported as an unfavorable prognostic marker in Hodgkin lymphoma. We aimed to investigate whether combining LMR and interim PET/CT scan result (PET2) confers stronger prognostic value than PET2 alone. 121 HL patients were investigated. LMR was calculated from a blood sample taken at the time of diagnosis. PET2 was carried out after the second chemotherapy cycle. Survival was calculated using the Kaplan-Meier method and significance was determined by log-rank test. Effect of variants on survival results was examined using univariate and multivariate analyses. Best LMR cut-off value was determined by receiver operating characteristic (ROC) curve. Best LMR cut-off value was 2.11 in the case of our patients (LMR > 2.11: favorable, LMR ≤ 2.11: unfavorable). Overall and progression-free survivals (OS/PFS) were significantly worse both in lower LMR (≤ 2.11) (OS: P = 0.041, PFS: P = 0.044) and PET2 positive groups (OS: P < 0.001, PFS: P < 0.001). In PET2 positive patient group (n = 32) the low LMR result meant a significantly worse OS (0.030) and PFS (0.001). Both LMR and PET2 proved to be independent prognostic factors on multivariate analysis, and strengthened each other's effect.
Collapse
|
33
|
Allen PB, Gordon LI. PD-1 blockade in Hodgkin's lymphoma: learning new tricks from an old teacher. Expert Rev Hematol 2016; 9:939-49. [PMID: 27622603 DOI: 10.1080/17474086.2016.1235970] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Classical Hodgkin's Lymphoma (cHL) is characterized by genetic reliance on the PD-1 pathway. Rapid accumulation of data describing the role and efficacy of PD-1 and its blockade warrants a focused review. AREAS COVERED In this article, we will review the unique biologic features that predispose cHL to PD-1 inhibition, current data regarding the safety and efficacy of PD-1 inhibitors in the treatment of cHL, biomarkers of immune response, ongoing clinical trials with PD-1 inhibitors, as well as areas of uncertainty. Expert commentary: The biologic and genetic underpinnings of cHL make it unique among all malignancies in its exquisite sensitivity to PD-1 inhibition. High response rates to single agent PD-1 inhibitors in early phase clinical trials serve as further proof of concept. These data strongly support continued clinical investigation of the evolving role of PD-1 inhibition in classical Hodgkin's lymphoma, including the optimal sequence, setting, and combination to best exploit the immunologic properties of this disease.
Collapse
Affiliation(s)
- Pamela Blair Allen
- a Lymphoma Program, Division of Hematology/Oncology, Department of Medicine , Northwestern University Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center , Chicago , IL , USA
| | - Leo I Gordon
- a Lymphoma Program, Division of Hematology/Oncology, Department of Medicine , Northwestern University Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center , Chicago , IL , USA
| |
Collapse
|
34
|
Abstract
The prognosis of patients with classical Hodgkin lymphoma following chemo- and radiotherapy has been excellent during the last 4 decades. However, the development of secondary malignancies is of major concern. Therefore, the reduction of radiotherapy application is a major objective of ongoing clinical trials. De-escalation of treatment may increase the risk of relapses and thus may lead to reappearance of prognostic factors. Prognostic biomarkers might help to identify patients who are at increased risk of relapse. This review summarizes the current knowledge about potential prognostic biomarkers for patients with classical Hodgkin lymphoma.
Collapse
Affiliation(s)
- Martin S Staege
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Stefanie Kewitz
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Toralf Bernig
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Caspar Kühnöl
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Christine Mauz-Körholz
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| |
Collapse
|
35
|
Plattel WJ, Alsada ZND, van Imhoff GW, Diepstra A, van den Berg A, Visser L. Biomarkers for evaluation of treatment response in classical Hodgkin lymphoma: comparison of sGalectin-1, sCD163 and sCD30 with TARC. Br J Haematol 2016; 175:868-875. [PMID: 27610595 DOI: 10.1111/bjh.14317] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/13/2016] [Indexed: 01/03/2023]
Abstract
Soluble Galectin-1 (sGal-1, also termed LGALS1), soluble CD163 (sCD163) and soluble CD30 (sCD30) have been reported to be elevated in plasma or serum of patients with classical Hodgkin lymphoma (cHL). We aimed to determine the clinical utility of these biomarkers for evaluation of treatment response compared to thymus and activation regulated chemokine (TARC, also termed CCL17). Plasma or serum samples were prospectively collected among 103 newly diagnosed cHL patients before and after treatment. Levels of sGal-1, sCD163, sCD30 and TARC were correlated with disease characteristics and clinical treatment response. Elevated plasma levels of sGal-1, sCD163, sCD30 and TARC were found in 67%, 21%, 91% and 93% of cHL patients respectively. Mean plasma levels of sGal-1 and sCD30 decreased after treatment but sCD163 did not decrease after treatment. There was no correlation with change of these markers and clinical treatment response in individual patients. TARC levels strongly correlated with disease characteristics and metabolic volume. TARC remained high in 6 out of 7 non-responsive patients and dramatically decreased in 95 out of 96 responsive patients. In summary, elevated pre-treatment levels of sGal-1, sCD163, sCD30 and TARC can be found in patients with cHL. However, only plasma TARC accurately reflects disease activity and correlates with clinical treatment response.
Collapse
Affiliation(s)
- Wouter J Plattel
- Department of Haematology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Zainab N D Alsada
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Gustaaf W van Imhoff
- Department of Haematology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Lydia Visser
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
36
|
Bröckelmann PJ, Angelopoulou MK, Vassilakopoulos TP. Prognostic factors in Hodgkin lymphoma. Semin Hematol 2016; 53:155-64. [DOI: 10.1053/j.seminhematol.2016.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
Vardhana S, Younes A. The immune microenvironment in Hodgkin lymphoma: T cells, B cells, and immune checkpoints. Haematologica 2016; 101:794-802. [PMID: 27365459 PMCID: PMC5004458 DOI: 10.3324/haematol.2015.132761] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/13/2016] [Indexed: 12/11/2022] Open
Abstract
Classical Hodgkin lymphoma is curable in the majority of cases with chemotherapy and/or radiation. However, 15-20% of patients ultimately relapse and succumb to their disease. Pathologically, classical Hodgkin lymphoma is characterized by rare tumor-initiating Reed-Sternberg cells surrounded by a dense immune microenvironment. However, the role of the immune microenvironment, particularly T and B cells, in either promoting or restricting Classical Hodgkin lymphoma growth remains undefined. Recent dramatic clinical responses seen using monoclonal antibodies against PD-1, a cell surface receptor whose primary function is to restrict T cell activation, have reignited questions regarding the function of the adaptive immune system in classical Hodgkin lymphoma. This review summarizes what is known regarding T cells, B cells, and immune checkpoints in classical Hodgkin lymphoma.
Collapse
Affiliation(s)
- Santosha Vardhana
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anas Younes
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
38
|
Rapidly Progressing Refractory Hodgkin Lymphoma: A Case Report and a Possible Explanation. Case Rep Med 2016; 2016:7698624. [PMID: 27429620 PMCID: PMC4939189 DOI: 10.1155/2016/7698624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 06/05/2016] [Indexed: 11/17/2022] Open
Abstract
Introduction. Hodgkin lymphoma is a highly curable lymphoid malignancy; however treatment of a significant number of patients remains challenging. Case Report. The authors present an unusually rapidly progressing case of refractory advanced stage classical nodular sclerosis subtype Hodgkin lymphoma with unfavorable prognosis. A 66-year-old male patient was refractory for first-line doxorubicin, bleomycin, vinblastin, dacarbazine (ABVD) treatment with persistent disease; therefore physicians changed treatment for dexamethasone, cytarabine, and cisplatin (DHAP) and later ifosfamide, gemcitabine, and vinorelbine (IGEV) regimen. Unfortunately the patient developed acute kidney and respiratory failure and died after 6 months of treatment. Current and retrospective histological examination of the patient's lymph node biopsy, skin lesion, and autopsy revealed the same aberrantly expressing CD4 positive nodular sclerosis subtype Hodgkin lymphoma. Conclusion. Aberrant expression of T-cell antigens on the Hodgkin and Reed/Sternberg cells could be associated with inferior outcome. T-cell associated antigens should be investigated more often in patients not responding sufficiently to treatment and hence treatment should be intensified or targeted therapy (brentuximab vedotin) should be considered.
Collapse
|
39
|
Cuccaro A, Annunziata S, Cupelli E, Martini M, Calcagni ML, Rufini V, Giachelia M, Bartolomei F, Galli E, D'Alò F, Voso MT, Leone G, Giordano A, Larocca LM, Hohaus S. CD68+ cell count, early evaluation with PET and plasma TARC levels predict response in Hodgkin lymphoma. Cancer Med 2016; 5:398-406. [PMID: 26758564 PMCID: PMC4799945 DOI: 10.1002/cam4.585] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/16/2015] [Accepted: 10/19/2015] [Indexed: 12/14/2022] Open
Abstract
Early response evaluation with [18F]fluordeoxyglucose (FDG) positron emission tomography after 2 cycles of chemotherapy (interim PET) has been indicated as the strongest predictor for outcome in classical Hodgkin lymphoma (HL). We studied the prognostic role of the number of tumor‐infiltrating CD68+ cells and of the plasma levels of TARC (thymus and activation‐regulated chemokine) in the context of interim PET in 102 patients with classical HL treated with Adriamycin, Bleomycin, Vinblastine, Dacarbazine (ABVD). After 2 ABVD cycles, interim PET according to Deauville criteria was negative (score 0–3) in 85 patients and positive (score 4–5) in 15 patients (2 patients technically not evaluable). TARC levels were elevated in 89% of patients at diagnosis, and decreased after 2 cycles in 82% of patients. Persistently elevated TARC levels in 18% of patients were significantly associated with a positive PET result (P = 0.007). Strong predictors for progression‐free survival (PFS) were a negative interim PET (85% vs. 28%, P < 0.0001) and CD68+ cell counts <5% (89% vs. 67%, P = 0.006), while TARC levels at diagnosis and at interim evaluation had no prognostic role. In multivariate analysis, interim PET, CD68+ cell counts and presence of B‐symptoms were independently associated with PFS. We conclude that although TARC levels are a biomarker for early response evaluation, they cannot substitute for interim PET as outcome predictor in HL. The evaluation of CD68 counts and B‐symptoms at diagnosis may help to identify low‐risk patients regardless positive interim PET.
Collapse
Affiliation(s)
- Annarosa Cuccaro
- Institute of Hematology, Catholic University of the Sacred Heart, Rome, Italy
| | - Salvatore Annunziata
- Institute of Nuclear Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Elisa Cupelli
- Institute of Hematology, Catholic University of the Sacred Heart, Rome, Italy
| | - Maurizio Martini
- Institute of Pathology, Catholic University of the Sacred Heart, Rome, Italy
| | - Maria L Calcagni
- Institute of Nuclear Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Vittoria Rufini
- Institute of Nuclear Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Manuela Giachelia
- Institute of Hematology, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Eugenio Galli
- Institute of Hematology, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco D'Alò
- Institute of Hematology, Catholic University of the Sacred Heart, Rome, Italy
| | - Maria T Voso
- Institute of Hematology, Catholic University of the Sacred Heart, Rome, Italy
| | - Giuseppe Leone
- Institute of Hematology, Catholic University of the Sacred Heart, Rome, Italy
| | - Alessandro Giordano
- Institute of Nuclear Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Luigi M Larocca
- Institute of Pathology, Catholic University of the Sacred Heart, Rome, Italy
| | - Stefan Hohaus
- Institute of Hematology, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
40
|
Lynch RC, Advani RH. Risk-Adapted Treatment of Advanced Hodgkin Lymphoma With PET-CT. Am Soc Clin Oncol Educ Book 2016; 35:e376-e385. [PMID: 27249744 DOI: 10.1200/edbk_159036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Although patients with advanced-stage classic Hodgkin lymphoma have excellent outcomes with contemporary therapy, the outcomes of patients with refractory disease is suboptimal. Identification of these high-risk patients at diagnosis is challenging as the differences in outcomes using clinical criteria are less marked using current modern therapy. Data suggest that an interim PET-CT may be a powerful tool in risk-stratifying patients. Retrospective studies show that a negative interim PET-CT after two to four cycles of ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine) is predictive of favorable outcome independent of IPS score. Currently, there are several ongoing trials that aim to determine whether early-response assessment can be used to select patients who might benefit from modifications of subsequent therapy, either by intensifying or abbreviating regimens and/or omitting radiotherapy with promising early results. Longer follow-up is required to assess whether this strategy impacts overall survival (OS). Herein, we review the results of recent trials using interim PET-CT-based adaptive design in the treatment of advanced HL.
Collapse
Affiliation(s)
- Ryan C Lynch
- From the Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Ranjana H Advani
- From the Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
41
|
Carlo-Stella C, Santoro A. Microenvironment-related biomarkers and novel targets in classical Hodgkin's lymphoma. Biomark Med 2015. [DOI: 10.2217/bmm.15.30] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Classical Hodgkin's lymphoma accounts for approximately 10% of all malignant lymphomas. Although most patients can be cured with modern treatment strategies, approximately 25% of them experience either primary or secondary chemorefractoriness or disease relapse, thus requiring novel treatments. Increasing preclinical and clinical evidences have demonstrated the role of microenvironment in the molecular pathogenesis of classical Hodgkin's lymphoma and elucidated the complex cross-talk between the malignant Hodgkin Reed–Sternberg cells and the nonmalignant, reactive cells of the microenvironment, strongly supporting novel therapeutic approaches aimed at targeting Hodgkin's Reed–Sternberg cells along with reactive cells in order to overcome chemorefractoriness. In the near future, these novel therapies will also be tested in chemosensitive patients to reduce long-term toxicities of chemo-radiotherapy.
Collapse
Affiliation(s)
- Carmelo Carlo-Stella
- Department of Hematology & Oncology, Humanitas Cancer Center, Humanitas Clinical & Research Center, Via Manzoni 56, 20089 Rozzano (Milano), Italy
- Department of Medical Biotechnology & Translational Medicine, University of Milano, Milano, Italy
| | - Armando Santoro
- Department of Hematology & Oncology, Humanitas Cancer Center, Humanitas Clinical & Research Center, Via Manzoni 56, 20089 Rozzano (Milano), Italy
| |
Collapse
|
42
|
Kataoka Y. Thymus and activation-regulated chemokine as a clinical biomarker in atopic dermatitis. J Dermatol 2015; 41:221-9. [PMID: 24628072 DOI: 10.1111/1346-8138.12440] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 01/17/2014] [Indexed: 11/28/2022]
Abstract
Thymus and activation-regulated chemokine (TARC/CCL17) is a member of the T-helper 2 chemokine family. In Japan, serum TARC level has been commercially measured since 2008. After years of experience, we realized that TARC is an extremely useful clinical biomarker for atopic dermatitis (AD) treatment. Usually, physicians conduct a visual examination to determine whether their treatment has been successful; however, the visual examination results may not always be accurate; in such cases, serum TARC levels should be measured to eliminate any ambiguity regarding the treatment outcome. When the waning and waxing of eczema and fluctuations in the serum TARC levels were considered, we frequently found that AD does not follow a natural course but follows non-regulated inflammatory floating caused by insufficient intermittent topical treatment. Serum TARC is a promising biomarker for remission and can be used for accurately monitoring proactive treatment for long-term control. Abnormally high serum TARC levels indicate accelerated pathogenesis of cutaneous inflammation. Rapid normalization and maintaining normal serum TARC levels using appropriate topical treatment is a reasonable strategy for alleviating inflammation without upregulating cytokine expression. Observing serum TARC levels during early intervention for severe infantile AD is worthwhile to determine initial disease activity and evaluate treatment efficacy. Appropriate control of severe early-onset infantile AD is important for improving prognosis of eczema and for preventing food allergies. Additionally, this biomarker is useful for improving patient adherence. Dermatologists will be able to make great progress in treating AD by adopting biomarkers such as TARC for accurately assessing non-visible subclinical disorders.
Collapse
Affiliation(s)
- Yoko Kataoka
- Dermatology, Osaka Prefectural Medical Center for Respiratory and Allergic Diseases, Habikino, Osaka, Japan
| |
Collapse
|
43
|
Gloghini A, Bongarzone I. Cell-secreted signals shape lymphoma identity. Semin Cancer Biol 2015; 34:81-91. [PMID: 25837156 DOI: 10.1016/j.semcancer.2015.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/21/2015] [Accepted: 02/05/2015] [Indexed: 12/12/2022]
Abstract
Sequencing data show that both specific genes and a number of signaling pathways are recurrently mutated in various types of lymphoma. DNA sequencing analyses of lymphoma have identified several aberrations that might affect the interaction between malignant cells and the tumor microenvironment. Microenvironmental functions are essential to lymphoma; they provide survival and proliferation signals and license immune evasion. It is plausible that interventions that aim to destroy tumor-microenvironment interactions may improve responses to therapeutics. Accordingly, the identification of extrinsic factors and their downstream intracellular signaling targets has led to much progress in understanding tumor-microenvironment interactions. Lymphoma cells are differently influenced by cells' interactions with components of their microenvironment; these cell extrinsic factors include soluble and immobilized factors, the extracellular matrix, and signals presented by neighboring cells. Soluble factors, which are often cell-secreted autocrine and paracrine factors, comprise a significant fraction of targetable molecules. To begin to understand how intercellular communication is conducted in lymphoma, a first order of study is deciphering the soluble factors secreted by malignant cells and microenvironmental cells. These soluble factors are shed into the interstitial fluid in lymphoma and can be conveniently explored using mass spectrometry. Protein components can be detected and quantified, thus enabling the routine navigation of the soluble part of the microenvironment. Elucidating functional and signaling states affords a new paradigm for understanding cancer biology and devising new therapies. This review summarizes knowledge in this field and discusses the utility of studying tumor-secreted factors.
Collapse
Affiliation(s)
- Annunziata Gloghini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - Italia Bongarzone
- Proteomics Laboratory, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| |
Collapse
|
44
|
Abstract
Abstract
Treatment of Hodgkin lymphoma is associated with 2 major types of risk: that the treatment may fail to cure the disease or that the treatment will prove unacceptably toxic. Careful assessment of the amount of the lymphoma (tumor burden), its behavior (extent of invasion or specific organ compromise), and host related factors (age; coincident systemic infection; and organ dysfunction, especially hematopoietic, cardiac, or pulmonary) is essential to optimize outcome. Elaborately assembled prognostic scoring systems, such as the International Prognostic Factors Project score, have lost their accuracy and value as increasingly effective chemotherapy and supportive care have been developed. Identification of specific biomarkers derived from sophisticated exploration of Hodgkin lymphoma biology is bringing promise of further improvement in targeted therapy in which effectiveness is increased at the same time off-target toxicity is diminished. Parallel developments in functional imaging are providing additional potential to evaluate the efficacy of treatment while it is being delivered, allowing dynamic assessment of risk during chemotherapy and adaptation of the therapy in real time. Risk assessment in Hodgkin lymphoma is continuously evolving, promising ever greater precision and clinical relevance. This article explores the past usefulness and the emerging potential of risk assessment for this imminently curable malignancy.
Collapse
|
45
|
Abstract
Abstract
The development of curative systemic treatment of Hodgkin lymphoma was recently voted one of the top 5 achievements of oncology in the last 50 years (http://cancerprogress.net/top-5-advances-modern-oncology). The high expectation of cure (above 80%) with initial therapy, even for advanced disease, is tempered by the recognition of some important limitations: not all patients are cured, especially those in older age groups, and patients have suffered debilitating or, in some cases, fatal long-term side effects. The challenge for modern treatment approaches is to improve the cure rate and, at the same time, minimize the long-term damage resulting from treatment. After several decades during which we have tested a variety of different ways to combine conventional cytotoxic treatments with or without radiotherapy but have identified no effective new approaches, the field is once again moving forward. The developments that hold the greatest promise in this respect are the application of functional imaging with 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) to make an early judgment of the success of treatment and the introduction of some highly active new agents such as antibody-drug conjugates.
Collapse
|
46
|
Lymphomagenesis in Hodgkin lymphoma. Semin Cancer Biol 2015; 34:14-21. [PMID: 25725205 DOI: 10.1016/j.semcancer.2015.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 02/06/2015] [Accepted: 02/10/2015] [Indexed: 02/07/2023]
Abstract
Hodgkin lymphoma (HL) accounts for approximately 0.6% of all new cancer cases, 10% of all lymphomas in the USA, leading to an approximate 9000 new cases per year. It is very unique in that the neoplastic Hodgkin and Reed-Sternberg (HRS) cells of classical HL account for only 1% of the tumor tissue in most cases, with various inflammatory cells including B-cells, T-cells, mast cells, macrophages, eosinophils, neutrophils, and plasma cells comprising the tumor microenvironment. Recent research has identified germinal center B-cells to be the cellular origin of HRS cells. Various transcription factor dysregulation in these neoplastic cells that explains for the loss of B-cell phenotype as well as acquisition of survival and anti-apoptotic features of HRS cells has been identified. Aberrant activation of nuclear factor-kappa B (NF-κB), Janus kinase (JAK)/signal transducer and activator of transcription (STAT), and phosphoinositide 3-kinase (PI3K) pathways play a central role in HL pathogenesis. Both intrinsic genetic mechanisms as well as extrinsic signals have been identified to account for the constitutive activation of these pathways. The extrinsic factors that regulate the activation of transcription pathways in HRS cells have also been studied in detail. Cytokines and chemokines produced both by the HRS cells as well as cells of the microenvironment of HL work in an autocrine and/or paracrine manner to promote survival of HRS cells as well as providing mechanisms for immune escape from the body's antitumor immunity. The understanding of various mechanisms involved in the lymphomagenesis of HL including the importance of its microenvironment has gained much interest in the use of these microenvironmental features as prognostic markers as well as potential treatment targets. In this article, we will review the pathogenesis of HL starting with the cellular origin of neoplastic cells and the mechanisms supporting its pathogenesis, especially focusing on the microenvironment of HL and its associated cytokines.
Collapse
|
47
|
Mounier N, Nicolas M, Gisselbrecht C, Christian G. Decision-making in the management of adult classical Hodgkin's lymphoma: determining the optimal treatment. Expert Rev Hematol 2015; 8:205-16. [PMID: 25634543 DOI: 10.1586/17474086.2015.995622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This review discusses promising new approaches in classical Hodgkin's lymphoma that have been recently evaluated. There is a focus on the fluorodeoxyglucose PET scanning that is now considered crucial for staging and treatment evaluation, including interim evaluation after two cycles. An up-front treatment strategy is discussed, with the place of radiation therapy and the difficult choice of chemotherapy intensity emphasized. Indications for frail patients are also reviewed, particularly elderly or HIV-positive patients. Emerging data on the antibody drug conjugate brentuximab vedotin and its future potential in the transplantation framework for relapsed/refractory Hodgkin's lymphoma is also discussed.
Collapse
|
48
|
Farina L, Rezzonico F, Spina F, Dodero A, Mazzocchi A, Crippa F, Alessi A, Dalto S, Viviani S, Corradini P. Serum Thymus and Activation-Regulated Chemokine Level Monitoring May Predict Disease Relapse Detected by PET Scan after Reduced-Intensity Allogeneic Stem Cell Transplantation in Patients with Hodgkin Lymphoma. Biol Blood Marrow Transplant 2014; 20:1982-8. [DOI: 10.1016/j.bbmt.2014.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 08/19/2014] [Indexed: 01/31/2023]
|
49
|
Jachimowicz RD, Engert A. The challenging aspects of managing adolescents and young adults with Hodgkin's lymphoma. Acta Haematol 2014; 132:274-8. [PMID: 25228552 DOI: 10.1159/000360205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cancer in the adolescent and young adult (AYA) is the second leading cause of nonaccidental death with hematological malignancies spiking during this period. Treatment of AYAs with hematological malignancies usually follows either pediatric or adult protocols with sufficient information lacking on subgroup analyses regarding course and outcome. In this review we will outline up-to-date treatment possibilities for AYAs diagnosed with Hodgkin's lymphoma. Early and late toxicities will be addressed and future directions of research suggested.
Collapse
Affiliation(s)
- Ron D Jachimowicz
- First Department of Internal Medicine, German Hodgkin Study Group, University of Cologne, Cologne, Germany
| | | |
Collapse
|
50
|
The NF-κB inhibitor DHMEQ decreases survival factors, overcomes the protective activity of microenvironment and synergizes with chemotherapy agents in classical Hodgkin lymphoma. Cancer Lett 2014; 349:26-34. [DOI: 10.1016/j.canlet.2014.03.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 03/19/2014] [Accepted: 03/23/2014] [Indexed: 12/28/2022]
|