1
|
Zou S, Liu B, Feng Y. CCL17, CCL22 and their receptor CCR4 in hematologic malignancies. Discov Oncol 2024; 15:412. [PMID: 39240278 PMCID: PMC11379839 DOI: 10.1007/s12672-024-01210-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 07/30/2024] [Indexed: 09/07/2024] Open
Abstract
Hematological malignancies (HM) are common malignant tumors with high morbidity and mortality rates, and are malignant diseases that seriously affect human health, with chemotherapy prone to recurrence and toxic side effects. Therefore, the development of precise, effective, and safe targeted therapeutic agents has become a hotspot in the current research of antitumor technology. More and more studies have shown that the interaction of C-C chemokine ligand 17 (CCL17) and C-C chemokine ligand 22 (CCL22) with the receptor C-C chemokine receptor type 4 (CCR4) promotes the immune escape of tumors and is closely related to the occurrence, development, and prognosis of hematological tumors. In this regard, we present a review on the expression and role of the CCL17/CCL22-CCR4 axis in HM, including lymphoma, leukemia, and multiple myeloma, with the aim of providing latest ideas and directions for the diagnosis and treatment of HM. In addition, we discuss the role and related mechanisms of HM therapeutic agents targeting the CCL17/CCL22-CCR4 axis and the potential of humanized anti-CCR4 antibodies for the treatment of HM.
Collapse
Affiliation(s)
- Shasha Zou
- Department of Hematology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Bo Liu
- Department of Key, Lab for Basic Pharmacology and Joint International Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Yonghuai Feng
- Department of Hematology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- Department of Hematology, Dongguan People's Hospital, Dongguan, China.
| |
Collapse
|
2
|
Freeman D, Diefenbach C, Lam L, Le T, Alexandre J, Raphael B, Grossbard M, Kaminetzky D, Ruan J, Chattopadhyay PK. terraFlow, a high-parameter analysis tool, reveals T cell exhaustion and dysfunctional cytokine production in classical Hodgkin's lymphoma. Cell Rep 2024; 43:114605. [PMID: 39128003 DOI: 10.1016/j.celrep.2024.114605] [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: 11/16/2023] [Revised: 03/19/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024] Open
Abstract
Immune cells express an incredible variety of proteins; by measuring combinations of these, cell types influencing disease can be precisely identified. We developed terraFlow, a platform that defines cell subsets exhaustively by combinatorial protein expression. Using high-parameter checkpoint-focused and function-focused panels, we studied classical Hodgkin's lymphoma (cHL), where systemic T cells have not been investigated in detail. terraFlow revealed immune perturbations in patients, including elevated activated, exhausted, and interleukin (IL)-17+ phenotypes, along with diminished early, interferon (IFN)γ+, and tumor necrosis factor (TNF)+ T cells before treatment; many perturbations remained after treatment. terraFlow identified more disease-associated differences than other tools, often with better predictive power, and included a non-gating approach, eliminating time-consuming and subjective manual thresholds. It also reports a method to identify the smallest set of markers distinguishing study groups. Our results provide mechanistic support for past reports of immune deficiency in cHL and demonstrate the value of terraFlow in immunotherapy and biomarker studies.
Collapse
Affiliation(s)
- Daniel Freeman
- terraFlow Bioinformatics, Cambridge, MA, USA; Biology and Biomedical Sciences, Harvard Medical School, Boston, MA, USA; Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | | | - Linda Lam
- terraFlow Bioinformatics, Cambridge, MA, USA
| | - Tri Le
- BD Biosciences, San Jose, CA, USA
| | | | - Bruce Raphael
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | | | | | - Jia Ruan
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | | |
Collapse
|
3
|
Engku Abd Rahman ENS, Irekeola AA, Shueb RH, Mat Lazim N, Mohamud R, Chen X, Ghazali L, Awang NMSH, Haron A, Chan YY. Aberrant frequency of TNFR2-expressing CD4+ FoxP3+ regulatory T cells in nasopharyngeal carcinoma patients. Cytokine 2023; 170:156341. [PMID: 37657236 DOI: 10.1016/j.cyto.2023.156341] [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: 05/07/2023] [Revised: 07/28/2023] [Accepted: 08/18/2023] [Indexed: 09/03/2023]
Abstract
TNFR2 is a surface marker of highly suppressive subset of CD4+ FoxP3+ regulatory T cells (Tregs) in humans and mice. This study examined the TNFR2 expression by Tregs of nasopharyngeal carcinoma (NPC) patients and healthy controls. The proliferation, migration, survival of TNFR2+ Tregs, and association with clinicopathological characteristics were assessed. The expression levels of selected cytokines were also determined. The results demonstrated that in both peripheral blood (PB) (10.45 ± 5.71%) and tumour microenvironment (TME) (54.38 ± 16.15%) of NPC patients, Tregs expressed TNFR2 at noticeably greater levels than conventional T cells (Tconvs) (3.91 ± 2.62%, p < 0.0001), akin to healthy controls. Expression of TNFR2 (1.06 ± 0.99%) was correlated better than CD25+ (0.40 ± 0.46%) and CD127-/low (1.00 ± 0.83% ) with FoxP3 expression in NPC PB (p = 0.0005). Though there was no significant association between TNFR2 expression with the functional capacity (proliferation, migration and survival) of Tregs (p > 0.05), the proportions of PB and TME TNFR2+ Tregs in NPC patients showed more proliferative, higher migration capacity, and better survival ability, as compared to those in healthy controls. Furthermore, TNFR2+ Tregs from NPC patients expressed significantly higher amounts of IL-6 (p = 0.0077), IL-10 (p = 0.0001), IFN-γ (p = 0.0105) and TNF-α (p < 0.0001) than those from healthy controls. Most significantly, TNFR2 expression in maximally suppressive Tregs population were linked to WHO Type III histological type, distant metastasis, progressive disease status, and poor prognosis for NPC patients. Hence, our research implies that TNFR2 expression by PB and TME Tregs may be a useful predictive indicator in NPC patients.
Collapse
Affiliation(s)
- Engku Nur Syafirah Engku Abd Rahman
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Ahmad Adebayo Irekeola
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia; Microbiology Unit, Department of Biological Sciences, College of Natural and Applied Sciences, Summit University Offa, PMB 4412, Offa Kwara State, Nigeria
| | - Rafidah Hanim Shueb
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Norhafiza Mat Lazim
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, 999078 Macau
| | - Liyana Ghazali
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nik Mohd Syahrul Hafizzi Awang
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Ali Haron
- Department of Otorhinolaryngology, Hospital Raja Perempuan Zainab II, Jalan Hospital, 15200 Kota Bharu, Kelantan, Malaysia
| | - Yean Yean Chan
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
4
|
Kilsdonk M, Veldman C, Rosati S, Plattel W, Diepstra A. The value of thymus and activation related chemokine immunohistochemistry in classic Hodgkin lymphoma diagnostics. Histopathology 2023; 82:495-503. [PMID: 36345263 PMCID: PMC10100154 DOI: 10.1111/his.14836] [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: 07/22/2022] [Revised: 10/10/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022]
Abstract
AIMS Classic Hodgkin lymphoma (cHL) should be distinguished from its wide variety of histological mimics, including reactive conditions and mature B and T cell neoplasms. Thymus and activation-related chemokine (TARC) is produced in extremely high quantities by the Hodgkin/Reed-Sternberg (HRS) tumour cells and is largely responsible for the attraction of CD4+ T cells into the cHL tumour micro-environment. In the current study we evaluated the diagnostic potential of TARC immunohistochemistry in daily practice in a tertiary referral centre in the Netherlands. METHODS AND RESULTS A total of 383 cases, approximately half of which were cHL mimics, were prospectively evaluated in the period from June 2014 to November 2020. In 190 cHL cases, 92% were TARC-positive and the majority of cases showed strong and highly specific staining in all HRS cells (77%). In most cases, TARC could discriminate between nodular lymphocyte-predominant and lymphocyte-rich Hodgkin lymphoma. HRS-like cells in mature lymphoid neoplasms were rarely positive (6.4%) and there was no TARC staining at all in 64 reactive lymphadenopathies. CONCLUSIONS TARC immunohistochemistry has great value in differentiating between cHL and its mimics, including nodular lymphocyte-predominant Hodgkin lymphoma, reactive lymphadenopathies and mature lymphoid neoplasms with HRS-like cells.
Collapse
Affiliation(s)
- Melvin Kilsdonk
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Carlijn Veldman
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Stefano Rosati
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wouter Plattel
- Department of Haematology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
5
|
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
|
6
|
Zhang Y, Chen K, Li L, Mao W, Shen D, Yao N, Zhang L. CCR4 is a prognostic biomarker and correlated with immune infiltrates in head and neck squamous cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1443. [PMID: 34733995 PMCID: PMC8506764 DOI: 10.21037/atm-21-3936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/02/2021] [Indexed: 11/06/2022]
Abstract
Background Increased evidence has indicated that the tumour microenvironment plays an essential in the development, treatment and prognosis of head and neck squamous cell carcinoma (HNSC). Recent studies have indicated CC chemokine receptor 4 (CCR4) plays an essential role in tumor invasion and other adverse biological behavior. This study used data from the Cancer Genome Atlas (TCGA) database to explore the role of CCR4 in HNSC and its clinical significance. Methods The gene expression and clinical data of HNSC patients in the TCGA database were extracted. Gene Expression Profiling Interactive Analysis (GEPIA) was used to analyze the expression of CCR4 in tumor and non-tumor tissue. Kaplan-Meier survival analysis was used to analyze the relationship between CCR4 expression and overall survival rate (OS), disease-specific survival (DSS), and progression-free interval (PFI) in HNSC. A logistic regression model was used to analyze the relationships between various clinical factors and CCR4 expression. Gene Set Enrichment Analysis (GSEA) was used to explore the potential role of CCR4 in HNSC. Additionally, we explored the relationship between CCR4 and immune infiltration. Results The expression of CCR4 in HNSC was not significantly different from that in normal tissue. The expression level of CCR4 in wild-type TP53 was higher than that in mutant TP53. Cox regression analysis showed the expression level of CCR4 was related to the patient's tumor grade and Tumor-Node-Metastasis (TNM) stage. CCR4 expression level is an independent prognostic factor. CCR4 is positively correlated with immune infiltration and immune checkpoints expression levels. The results of GSEA revealed that the high CCR4 expression group genes were enriched in allograft rejection, inflammatory response, IL-6/JAK/STAT3 signaling, interferon gamma response, and KRAS signaling up. Low CCR4 expression group genes were enriched in oxidative phosphorylation, MYC targets v1, DNA repair, reactive oxygen species pathway, and P53 pathway. Further, our study indicated CCR4 can also predict the prognosis of radiotherapy patients. Conclusions Our study found that CCR4 was a prognostic marker related to HNSC immune infiltration, and patients with high expression of CCR4 had a better prognosis.
Collapse
Affiliation(s)
- Yijian Zhang
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Kai Chen
- Department of Radiotherapy, First People's Hospital of Yancheng, Yancheng, China
| | - Li Li
- Department of Oncology, Huaian Hospital, Huai'an, China
| | - Weidong Mao
- Department of Oncology, Jiangyin People's Hospital, Wuxi, China
| | - Dong Shen
- Department of Oncology, Jiangyin People's Hospital, Wuxi, China
| | - Ninghua Yao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Lei Zhang
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
Abstract
Classic Hodgkin lymphoma (cHL) is one of the most common lymphomas in the Western world. Advances in the management of cHL have led to high cure rates exceeding 80%. Nevertheless, relapse or refractory disease in a subset of patients and treatment-related toxicity still represents unsolved clinical problems. The introduction of targeted treatments such as PD-1 blockade and the CD30 antibody drug conjugate, brentuximab vedotin, has broadened treatment options in cHL, emphasizing the critical need to identify biomarkers with the goal to provide rationales for treatment selection, increase effective drug utilization, and minimize toxicity. The unique biology of cHL featuring low abundant tumor cells and numerous nonmalignant immune cells in the tumor microenvironment can provide various types of promising biomarkers related to the tumor cells directly, tumor microenvironment cross-talk, and host immune response. Here, we comprehensively review novel biomarkers including circulating tumor DNA and gene expression-based prognostic models that might guide the ideal management of cHL in the future.
Collapse
|
14
|
Zaucha JM, Chauvie S, Zaucha R, Biggii A, Gallamini A. The role of PET/CT in the modern treatment of Hodgkin lymphoma. Cancer Treat Rev 2019; 77:44-56. [PMID: 31260900 DOI: 10.1016/j.ctrv.2019.06.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/06/2019] [Accepted: 06/09/2019] [Indexed: 12/12/2022]
Abstract
Classical Hodgkin Lymphoma is distinguished from other lymphomas by its peculiar biology and heterogeneous chemosensitivity. Most of the patients respond to the standard first-line treatment and are cured, however, in selected cases, the disease relapses or remains primarily refractory. Among predictive/prognostic factors 18FDG positron emission tomography (PET), fully integrated with computed tomography (PET/CT) proved to be extremely useful in identifying patients with poor prognosis at the time of diagnosis, during and at the end of treatment. The aim of this review is to present the current role of PET/CT in cHL at staging, interim and end of therapy assessment and its ability to guide treatment with a response- and risk-adapted strategy in clinical practice. Finally, quantitative PET measurement and the concurrent use of PET with selected biomarkers are discussed.
Collapse
Affiliation(s)
- Jan Maciej Zaucha
- Department of Hematology and Transplantology, Medical University of Gdańsk, Poland.
| | - Stephane Chauvie
- Department of Medical Physics, Santa Croce e Carle Hospital, Cuneo, Italy
| | - Renata Zaucha
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Poland
| | - Alberto Biggii
- Department of Nuclear Medicine, Santa Croce e Carle Hospital, Cuneo, Italy
| | - Andrea Gallamini
- Department of Research and Clinical Innovation, A. Lacassagne Cancer Center, Nice, France
| |
Collapse
|
15
|
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
|
16
|
Locatelli SL, Careddu G, Serio S, Consonni FM, Maeda A, Viswanadha S, Vakkalanka S, Castagna L, Santoro A, Allavena P, Sica A, Carlo-Stella C. Targeting Cancer Cells and Tumor Microenvironment in Preclinical and Clinical Models of Hodgkin Lymphoma Using the Dual PI3Kδ/γ Inhibitor RP6530. Clin Cancer Res 2018; 25:1098-1112. [DOI: 10.1158/1078-0432.ccr-18-1133] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/31/2018] [Accepted: 10/19/2018] [Indexed: 11/16/2022]
|
17
|
Walewski J, Paszkiewicz-Kozik E, Borsaru G, Hellmann A, Janikova A, Warszewska A, Mais A, Ammendola A, Herz T, Krauss B, Henning SW. Resminostat in patients with relapsed or refractory Hodgkin lymphoma: results of the phase II SAPHIRE study. Leuk Lymphoma 2018; 60:675-684. [DOI: 10.1080/10428194.2018.1492122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Jan Walewski
- Department of Lymphoid Malignancies, Maria Skłodowska-Curie Institute – Oncology Center, Warszawa, Poland
| | - Ewa Paszkiewicz-Kozik
- Department of Lymphoid Malignancies, Maria Skłodowska-Curie Institute – Oncology Center, Warszawa, Poland
| | | | - Andrzej Hellmann
- Department for Hematology and Transplantology, University Clinical Centre, Medical University of Gdansk, Gdansk, Poland
| | - Andrea Janikova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Agnieszka Warszewska
- Department of Lymphoid Malignancies, Maria Skłodowska-Curie Institute – Oncology Center, Warszawa, Poland
| | - Anna Mais
- 4SC AG, Martinsried, Planegg, Germany
| | | | | | | | | |
Collapse
|
18
|
Li Z, Ju X, Lee K, Clarke C, Hsu JL, Abadir E, Bryant CE, Pears S, Sunderland N, Heffernan S, Hennessy A, Lo TH, Pietersz GA, Kupresanin F, Fromm PD, Silveira PA, Tsonis C, Cooper WA, Cunningham I, Brown C, Clark GJ, Hart DNJ. CD83 is a new potential biomarker and therapeutic target for Hodgkin lymphoma. Haematologica 2018; 103:655-665. [PMID: 29351987 PMCID: PMC5865416 DOI: 10.3324/haematol.2017.178384] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/10/2018] [Indexed: 11/30/2022] Open
Abstract
Chemotherapy and hematopoietic stem cell transplantation are effective treatments for most Hodgkin lymphoma patients, however there remains a need for better tumor-specific target therapy in Hodgkin lymphoma patients with refractory or relapsed disease. Herein, we demonstrate that membrane CD83 is a diagnostic and therapeutic target, highly expressed in Hodgkin lymphoma cell lines and Hodgkin and Reed-Sternberg cells in 29/35 (82.9%) Hodgkin lymphoma patient lymph node biopsies. CD83 from Hodgkin lymphoma tumor cells was able to trogocytose to surrounding T cells and, interestingly, the trogocytosing CD83+T cells expressed significantly more programmed death-1 compared to CD83-T cells. Hodgkin lymphoma tumor cells secreted soluble CD83 that inhibited T-cell proliferation, and anti-CD83 antibody partially reversed the inhibitory effect. High levels of soluble CD83 were detected in Hodgkin lymphoma patient sera, which returned to normal in patients who had good clinical responses to chemotherapy confirmed by positron emission tomography scans. We generated a human anti-human CD83 antibody, 3C12C, and its toxin monomethyl auristatin E conjugate, that killed CD83 positive Hodgkin lymphoma cells but not CD83 negative cells. The 3C12C antibody was tested in dose escalation studies in non-human primates. No toxicity was observed, but there was evidence of CD83 positive target cell depletion. These data establish CD83 as a potential biomarker and therapeutic target in Hodgkin lymphoma.
Collapse
Affiliation(s)
- Ziduo Li
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Australia
| | - Xinsheng Ju
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Australia
| | - Kenneth Lee
- Sydney Medical School, University of Sydney, Australia
- Department of Anatomical Pathology, Concord Repatriation General Hospital, Sydney, Australia
| | - Candice Clarke
- Department of Anatomical Pathology, Concord Repatriation General Hospital, Sydney, Australia
| | - Jennifer L Hsu
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Australia
| | - Edward Abadir
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Australia
| | - Christian E Bryant
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Suzanne Pears
- Animal Facility, Royal Prince Alfred Hospital, Sydney, Australia
| | | | - Scott Heffernan
- Animal Facility, Royal Prince Alfred Hospital, Sydney, Australia
| | | | - Tsun-Ho Lo
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Australia
| | - Geoffrey A Pietersz
- Burnet Institute, Melbourne, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Fiona Kupresanin
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
| | - Phillip D Fromm
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Australia
| | - Pablo A Silveira
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Australia
| | - Con Tsonis
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
| | - Wendy A Cooper
- Sydney Medical School, University of Sydney, Australia
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, Australia
- School of Medicine, University of Western Sydney, Australia
| | - Ilona Cunningham
- Department of Haematology, Concord Repatriation General Hospital, Sydney, Australia
| | - Christina Brown
- Sydney Medical School, University of Sydney, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Georgina J Clark
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Australia
| | - Derek N J Hart
- Dendritic Cell Research, ANZAC Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Australia
| |
Collapse
|
19
|
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
|
20
|
Abstract
The choice of treatment for advanced Hodgkin’s lymphoma has traditionally been made using an assessment of the baseline risk factors and a judgement of the balance between efficacy and toxicity for the group in question. The use of functional imaging with 2-(18F)-fluoro-2-deoxy-D-glucose (FDG)-positron emission tomography (PET) early in the course of therapy offers a way to make treatment better adjusted to the most important feature of Hodgkin’s lymphoma: the response to therapy. Recent studies have shown that excellent results can be achieved by using early FDG-PET to modulate therapy, with escalation for those with an unsatisfactory response and treatment reduction for those with the most chemosensitive disease. The results of these trials indicate that response-adapted therapy should now become part of the standard approach to care, offering opportunities to improve the results further by indicating those subgroups in need of new approaches such as the emerging antibody-based treatments.
Collapse
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
Gallamini A, Hutchings M, Borra A. Functional Imaging in Hodgkin Lymphoma. HODGKIN LYMPHOMA 2015. [DOI: 10.1007/978-3-319-12505-3_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
27
|
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]
|
28
|
Identification of chemokines and growth factors in proliferative diabetic retinopathy vitreous. BIOMED RESEARCH INTERNATIONAL 2014; 2014:486386. [PMID: 25401103 PMCID: PMC4226181 DOI: 10.1155/2014/486386] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 11/17/2022]
Abstract
Associations were investigated between levels of chemokines and growth factors in the vitreous and proliferative diabetic retinopathy (PDR). Enrolled were 58 patients (58 eyes) requiring pars plana vitrectomy (PPV), with PDR (n=32, none with traction retinal detachment) or not (non-PDR). In the latter, 16 had macular hole (MH) and 10 had epiretinal membrane (ERM). With a multiplex bead immunoassay, levels of 11 chemokines and growth factors were measured from the undiluted vitreous sample from each patient. In the non-PDR eyes, the levels of the 11 chemokines and growth factors tested were similar between patients with MH and those with ERM. However, the levels of all 11 were significantly higher in the PDR eyes relative to the non-PDR; CCL17, CCL19, and TGFβ3 were markedly upregulated and have not been investigated in PDR previously. The significantly higher levels of CCL4 and CCL11 in PDR contradict the results of previous reports. Based on Spearman's nonparametric test, moderate-to-strong correlations were found between VEGF and other mediators. Our results indicate that these chemokines and growth factors could be candidates for research into targeted therapies applied either singly or in combination with anti-VEGF drugs for the treatment of PDR.
Collapse
|
29
|
Agur A, Amir G, Paltiel O, Klein M, Dann EJ, Goldschmidt H, Goldschmidt N. CD68 staining correlates with the size of residual mass but not with survival in classical Hodgkin lymphoma. Leuk Lymphoma 2014; 56:1315-9. [PMID: 25204373 DOI: 10.3109/10428194.2014.963081] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The prognostic role of CD68 tumor-associated macrophages in classic Hodgkin lymphoma (cHL) remains controversial. We stained diagnostic biopsies and scored for CD68 using the PGM1 antibody among 98 consecutive patients with cHL from our center followed over a median of 45 months for progression-free survival (PFS). Among 79 patients we assessed interim and post-treatment positron emission tomography-computed tomography (PET-CT). Residual mass (RM) size was based on the greatest diameter of the largest mass seen in post-treatment imaging, and percent reduction was calculated by comparing RM size with its greatest pretreatment diameter. We found a significant association between CD68 positivity and absolute size of initial disease mass (p = 0.014) and residual mass at the end of therapy (p = 0.006) but no association was observed with interim PET-CT results or PFS. Our findings suggest that macrophages may influence tumor size by altering the microenvironment. This study does not support a prognostic role of CD68 positivity in predicting survival.
Collapse
Affiliation(s)
- Ariel Agur
- Hematology Department, Hadassah-Hebrew University Medical Center , Jerusalem , Israel
| | | | | | | | | | | | | |
Collapse
|
30
|
Lim JB, Kim DK, Chung HW. Clinical significance of serum thymus and activation-regulated chemokine in gastric cancer: potential as a serum biomarker. Cancer Sci 2014; 105:1327-33. [PMID: 25154912 PMCID: PMC4462361 DOI: 10.1111/cas.12505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 08/04/2014] [Accepted: 08/13/2014] [Indexed: 01/26/2023] Open
Abstract
Thymus and activation-regulated chemokine (TARC) can stimulate cancer cell proliferation and migration. The present study evaluated the clinical significance of serum TARC in gastric cancer (GC). We measured serum TARC, macrophage-derived chemokine, monocyte chemotactic protein-1 and stem cell factor (SCF) levels using a chemiluminescent immunoassay along the GC carcinogenesis (normal, high-risk, early GC [EGC] and advanced GC [AGC]) in both training (N = 25 per group) and independent validation datasets (90 normal, 30 high-risk, 50 EGC and 50 AGC). Serum levels were compared among groups using one-way analysis of variance. To evaluate the diagnostic potential of serum TARC for GC, receiver operating characteristic curve and logistic regression analyses were performed. Correlations between serum TARC and GC clinicopathological features were analyzed using Spearman's correlation. In the training dataset, serum TARC correlated with serum MDC, MCP-1 and SCF. However, only serum TARC and SCF were significantly higher in cancer groups than non-cancer groups (P < 0.001). In the validation dataset, serum TARC also increased along the GC carcinogenesis; the AGC group (167.2 ± 111.1 ng/mL) had significantly higher levels than the EGC (109.1 ± 67.7 ng/mL), the high-risk (66.2 ± 47.7 ng/mL) and the normal (67.5 ± 36.2 ng/mL) groups (Bonferroni, all P < 0.001). Receiver operating characteristic curves and logistic regression demonstrated the remarkable diagnostic potential of serum TARC as a single marker (72.0% sensitivity and 71.1% specificity; cutoff point, 0.37; logistic regression) and in a multiple-marker panel (72.6% sensitivity and 88.2% specificity; cutoff point, 0.54). Spearman's correlation showed that serum TARC was closely correlated with tumor size (γs = 0.227, P = 0.028), T-stage (γs = 0.340, P = 0.001), N-stage (γs = 0.318, P = 0.002) and M-stage (γs = 0.346, P = 0.001). Serum TARC is a promising serum biomarker for GC.
Collapse
Affiliation(s)
- Jong-Baeck Lim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | | | | |
Collapse
|
31
|
Romano A, Vetro C, Caocci G, Greco M, Parrinello NL, Di Raimondo F, La Nasa G. Immunological deregulation in classic hodgkin lymphoma. Mediterr J Hematol Infect Dis 2014; 6:e2014039. [PMID: 24959336 PMCID: PMC4063611 DOI: 10.4084/mjhid.2014.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/06/2014] [Indexed: 01/19/2023] Open
Abstract
Classic Hodgkin Lymphoma (cHL) has a unique histology since only a few neoplastic cells are surrounded by inflammatory accessory cells that in the last years have emerged as crucial players in sustaining the course of disease. In addition, recent studies suggest that the abnormal activity of these inflammatory cells (such as deregulation in regulatory T cells signaling, expansion of myeloid derived suppressor cells, HLA-G signaling and natural killer cells dysfunction) may have prognostic significance. This review is focused on summarizing recent advanced in immunological defects in cHL with translational implications.
Collapse
Affiliation(s)
- Alessandra Romano
- Division of Haematology, Azienda Ospedaliera “Policlinico-Vittorio Emanuele”, University of Catania. Via Citelli 6, 95124 Catania, Italy
| | - Calogero Vetro
- Division of Haematology, Azienda Ospedaliera “Policlinico-Vittorio Emanuele”, University of Catania. Via Citelli 6, 95124 Catania, Italy
| | - Giovanni Caocci
- Hematology Unit, Department of Medical Sciences “Mario Aresu,” University of Cagliari, Italy
| | - Marianna Greco
- Hematology Unit, Department of Medical Sciences “Mario Aresu,” University of Cagliari, Italy
| | - Nunziatina Laura Parrinello
- Division of Haematology, Azienda Ospedaliera “Policlinico-Vittorio Emanuele”, University of Catania. Via Citelli 6, 95124 Catania, Italy
| | - Francesco Di Raimondo
- Division of Haematology, Azienda Ospedaliera “Policlinico-Vittorio Emanuele”, University of Catania. Via Citelli 6, 95124 Catania, Italy
| | - Giorgio La Nasa
- Hematology Unit, Department of Medical Sciences “Mario Aresu,” University of Cagliari, Italy
| |
Collapse
|
32
|
Jona A, Szodoray P, Illés A. Immunologic pathomechanism of Hodgkin's lymphoma. Exp Hematol 2013; 41:995-1004. [PMID: 24099823 DOI: 10.1016/j.exphem.2013.09.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 09/16/2013] [Accepted: 09/29/2013] [Indexed: 12/16/2022]
Abstract
Hodgkin's lymphoma is a lymphoid malignancy of the immune system. The pathognomonic Hodgkin and Reed-Sternberg cells (HRS) are derived mainly from monoclonal, preapoptotic B cells, and they carry rearranged, somatically mutated immunoglobulin heavy chains. In an appropriate microenvironment, HRS cells escape from apoptosis by several mechanisms, including single mutations, aberrant signaling pathways. Eventually, weakened immune surveillance leads to uncontrolled, disproportional B cell proliferation. This review summarizes the latest findings on the pathogenesis of Hodgkin lymphoma, with a special emphasis on immunologic processes, and depicts current and future immunotherapeutic regimens, which improve treatment outcomes and reduce late toxicities.
Collapse
Affiliation(s)
- Adam Jona
- Department of Hematology, Institute for Internal Medicine; University of Debrecen Medical and Health Science Center, Debrecen, Hungary.
| | | | | |
Collapse
|
33
|
Marri PR, Hodge LS, Maurer MJ, Ziesmer SC, Slager SL, Habermann TM, Link BK, Cerhan JR, Novak AJ, Ansell SM. Prognostic significance of pretreatment serum cytokines in classical Hodgkin lymphoma. Clin Cancer Res 2013; 19:6812-9. [PMID: 24141626 DOI: 10.1158/1078-0432.ccr-13-1879] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE Although the International Prognostic Score (IPS) is the gold standard for risk-stratifying patients with classical Hodgkin lymphoma (cHL), these criteria do not accurately predict outcome. As cytokines are critically involved in driving cHL, we tested whether pretreatment serum cytokine levels could provide additional prognostic information. EXPERIMENTAL DESIGN Thirty cytokines were measured in pretreatment serum from 140 patients with cHL and compared with 50 nonlymphoma controls. Patients were followed for event-free survival (EFS) and overall survival (OS), and Cox proportional hazards regression models were used to assess the association of individual cytokines and the cytokine profiles with outcome via unadjusted and IPS-adjusted HR. RESULTS Twelve cytokines (EGF, bFGF, G-CSF, HGF, IL-6, IL-8, IL-12, IL-2R, IP-10, MIG, TNF-α, and VEGF) were significantly (P < 0.05) higher in patients with cHL than controls; elevated levels of HGF, IL-6, IL-2R, IP-10, and MIG were all associated with poorer EFS. Only interleukin-2 receptor (IL-2R; P = 0.002) and interleukin (IL)-6 (P < 0.001) were independently prognostic. Patients with increased IL-6 and IL-2R had a significantly higher risk of early relapse and death, a finding that remained significant even after IPS-based risk stratification. Although elevated IL-6 and IL-2R correlated with the IPS, soluble CD30 (sCD30), and thymus and activation-related chemokine (TARC) levels, the two-cytokine model remained independently predictive of prognosis. CONCLUSIONS Elevated pretreatment serum cytokines are associated with increased disease relapse and inferior survival in cHL. Thus, the pretreatment cytokine profile, particularly serum levels of IL-6 and IL-2R, may be used to identify patients with cHL at high risk for early-disease relapse.
Collapse
Affiliation(s)
- Preethi Reddy Marri
- Authors' Affiliations: Divisions of Hematology and Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota; and Department of Internal Medicine, College of Medicine, University of Iowa, Iowa City, Iowa
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Provencio M, Sánchez A, Sánchez-Beato M. New drugs and targeted treatments in Hodgkin's lymphoma. Cancer Treat Rev 2013; 40:457-64. [PMID: 24095205 DOI: 10.1016/j.ctrv.2013.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/24/2013] [Accepted: 09/04/2013] [Indexed: 12/26/2022]
Abstract
New drugs are being developed in recent years that may change the handling of relapsed and refractory Hodgkin's lymphoma patients. Brentuximab vedotin treatment has already been approved by the FDA; and other drugs are promising, such as histone deacetylase inhibitors, bendamustine, lenalidomide and m-TOR inhibitors.
Collapse
Affiliation(s)
- Mariano Provencio
- Medical Oncology Service, Onco-hematology Research Unit, Instituto de Investigación Sanitaria Puerta de Hierro, Hospital Universitario Puerta de Hierro-Majadahonda, Spain.
| | - Antonio Sánchez
- Medical Oncology Service, Onco-hematology Research Unit, Instituto de Investigación Sanitaria Puerta de Hierro, Hospital Universitario Puerta de Hierro-Majadahonda, Spain
| | - Margarita Sánchez-Beato
- Medical Oncology Service, Onco-hematology Research Unit, Instituto de Investigación Sanitaria Puerta de Hierro, Hospital Universitario Puerta de Hierro-Majadahonda, Spain
| |
Collapse
|
35
|
Ghesquières H, Maurer MJ, Casasnovas O, Ansell SM, Larrabee BR, Lech-Maranda E, Novak AJ, Borrel AL, Slager SL, Brice P, Allmer C, Brion A, Ziesmer SC, Morschhauser F, Habermann TM, Gaillard I, Link BK, Stamatoullas A, Fermé C, Dogan A, Macon WR, Audouin J, Cerhan JR, Salles G. Cytokine gene polymorphisms and progression-free survival in classical Hodgkin lymphoma by EBV status: results from two independent cohorts. Cytokine 2013; 64:523-31. [PMID: 24008079 DOI: 10.1016/j.cyto.2013.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 06/18/2013] [Accepted: 08/06/2013] [Indexed: 12/29/2022]
Abstract
BACKGROUND Cytokines are important immune mediators of classical Hodgkin lymphoma (CHL) pathogenesis, and circulating levels at diagnosis may help predict prognosis. Germline single nucleotide polymorphisms (SNPs) in immune genes have been correlated with cytokine production and function. METHODS We investigated whether selected germline SNPs in IL10 (rs1800890, rs1800896, rs1800871, rs1800872), TNFA (rs1800629), IL6 (rs1800795), ILRN (rs419598), INFG (rs2430561) and CCL17 (rs223828) were associated with circulating levels of related cytokines at diagnosis and progression-free survival (PFS) in CHL. Patients were from France (GELA, N=464; median age=32years) and the United States (Iowa/Mayo Specialized Program Of Research Excellence [SPORE], N=239; median age=38years); 22% of 346 CHL cases with EBV tumor status were positive. RESULTS There was no association with any of the SNPs with cytokine levels. Overall, there was no association of any of the SNPs with PFS. In exploratory analyses by EBV status, TNFA rs1800629 (HRAA/AG=2.41; 95%CI, 1.17-4.94) was associated with PFS in EBV-negative GELA patients, with similar trends in the SPORE patients (HRAA/AG=1.63; 95%CI, 0.61-4.40). In a meta-analysis of the two studies, TNFA (HRAA/AG=2.11; 95%CI, 1.18-3.77; P=0.01) was statistically significant, and further adjustment for the international prognostic system did not alter this result. CONCLUSIONS This study showed that germline variation in TNFA was associated with CHL prognosis for EBV-negative patients, which will require confirmation. These results support broader studies on the differential impact of genetic variation in immune genes on EBV-positive vs. EBV-negative CHL pathogenesis.
Collapse
Affiliation(s)
- Hervé Ghesquières
- Onco-Hematology, Centre Léon Bérard, UMR CNRS 5239, Université Lyon 1, Lyon, France; Health Sciences Research, Mayo Clinic, Rochester, MN, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Diefenbach C, Steidl C. New strategies in Hodgkin lymphoma: better risk profiling and novel treatments. Clin Cancer Res 2013; 19:2797-803. [PMID: 23447000 PMCID: PMC3928836 DOI: 10.1158/1078-0432.ccr-12-3064] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent advances in Hodgkin lymphoma research are expected to prelude a promising new treatment era for patients and their treating physicians. Scientific investigations over the last few years have provided new insights into risk stratification, and, simultaneously, a plethora of novel targeted therapies are emerging for patients with relapsed and refractory disease. These novel therapies will be tested primarily in high-risk patients because 75% of the patients are cured with conventional therapies. The challenges, as Hodgkin lymphoma therapy moves forward, will be using these biologic insights to identify the patients who may benefit earlier in treatment from these novel agents, and tailoring the therapy to the tumor biology of the patient. These dual aims are intertwined; as our therapeutic arsenal increases, these biologic determinants of risk may themselves inform the design of therapies and the choice of treatments for high-risk patients.
Collapse
Affiliation(s)
- Catherine Diefenbach
- Department of Medicine, New York University School of Medicine, NYU Cancer Institute, New York, New York 10016, USA.
| | | |
Collapse
|
37
|
Karube K, Niino D, Kimura Y, Ohshima K. Classical Hodgkin lymphoma, lymphocyte depleted type: clinicopathological analysis and prognostic comparison with other types of classical Hodgkin lymphoma. Pathol Res Pract 2013; 209:201-7. [PMID: 23478005 DOI: 10.1016/j.prp.2012.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 11/05/2012] [Accepted: 11/26/2012] [Indexed: 10/27/2022]
Abstract
The lymphocyte-depleted type (LD) is a morphological subtype of classical Hodgkin lymphoma (CHL), but its rarity and heterogeneous morphological character makes a definite clinicopathological identification difficult. To characterize this disease, LD cases were compared with other types of CHL. From 1982 to 2006, we collected 310 CHL cases. Among them, 29 cases were diagnosed as LD. We could additionally analyze clinical data of 157 CHL cases (including 28 LD cases) and the immunophenotype of 150 CHL cases (including 28 LD cases). We compared clinicopathological data between LD cases and other types of CHL cases and determined prognostic factors by univariate and multivariate analysis. LD showed a more progressive disease stage (stage 3/4, 64%) than other types of CHL (stage 3/4, 30%; P<0.001), more frequent B symptoms (89% vs. 40%; P<0.001) and extranodal invasion (50% vs. 11%; P<0.001), older age (median: 66 vs. 33; P<0.001), higher serum soluble interleukin 2 receptor levels (median: 8240U/ml vs. 1705U/ml; P<0.001), and much poorer prognosis regardless of the international prognostic score (IPS) (five-year overall survival: 29% vs. 86%; P<0.001). The morphological subtype of LD represented an independent prognostic factor as did age and IPS by multivariate analysis. Immunohistochemistry showed that the characteristics of Hodgkin and Reed-Sternberg (HRS) cells of LD are basically not different from those of other types of CHL in terms of CD30, CD15, and chemokine receptors, except for high-level EB-virus infection (72% vs. 41%; P=0.002). LD should be distinguished from other types of classical Hodgkin lymphoma because of its definitive clinicopathological characters.
Collapse
Affiliation(s)
- Kennosuke Karube
- Department of Pathology, School of Medicine, Kurume University, Japan.
| | | | | | | |
Collapse
|
38
|
Sauer M, Plütschow A, Jachimowicz RD, Kleefisch D, Reiners KS, Ponader S, Engert A, von Strandmann EP. Baseline serum TARC levels predict therapy outcome in patients with Hodgkin lymphoma. Am J Hematol 2013; 88:113-5. [PMID: 23225085 DOI: 10.1002/ajh.23361] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/24/2012] [Accepted: 10/29/2012] [Indexed: 01/28/2023]
Abstract
Hodgkin lymphoma (HL) has become one of the best curable cancers. However, better biomarkers are needed for outcome prediction that would allow protecting patients from over- or under-dosing of treatment. Thymus and activation-regulated chemokine/CCL17 (TARC) is highly and specifically elevated in this disease and has been proposed as possible biomarker in HL patients. In this study, we show that pretreatment TARC levels were associated with established clinical risk factors and predictive for response to treatment in a large cohort of HL patients treated in clinical trials by the German Hodgkin Study Group. Moreover, TARC levels also significantly contributed to a novel multivariate model predicting treatment response. These data clearly suggest an important role for this chemokine as biomarker in HL.
Collapse
Affiliation(s)
- Maike Sauer
- Department of Internal Medicine I; Laboratory for Immunotherapy; University Hospital Cologne; Cologne; Germany
| | - Annette Plütschow
- Department of Internal Medicine I; German Hodgkin Study Group; University Hospital Cologne; Cologne; Germany
| | | | - Dominik Kleefisch
- Department of Internal Medicine I; Laboratory for Immunotherapy; University Hospital Cologne; Cologne; Germany
| | - Katrin S. Reiners
- Department of Internal Medicine I; Laboratory for Immunotherapy; University Hospital Cologne; Cologne; Germany
| | | | - Andreas Engert
- Department of Internal Medicine I; German Hodgkin Study Group; University Hospital Cologne; Cologne; Germany
| | - Elke Pogge von Strandmann
- Department of Internal Medicine I; Laboratory for Immunotherapy; University Hospital Cologne; Cologne; Germany
| |
Collapse
|
39
|
Jones K, Vari F, Keane C, Crooks P, Nourse JP, Seymour LA, Gottlieb D, Ritchie D, Gill D, Gandhi MK. Serum CD163 and TARC as disease response biomarkers in classical Hodgkin lymphoma. Clin Cancer Res 2012; 19:731-42. [PMID: 23224400 DOI: 10.1158/1078-0432.ccr-12-2693] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Candidate circulating disease response biomarkers for classical Hodgkin lymphoma (cHL) might arise from Hodgkin-Reed-Sternberg (HRS) cells or nonmalignant tumor-infiltrating cells. HRS cells are sparse within the diseased node, whereas benign CD163(+) M2 tissue-associated macrophages (TAM) are prominent. CD163(+) cells within the malignant node may be prognostic, but there is no data on serum CD163 (sCD163). The HRS-specific serum protein sTARC shows promise as a disease response biomarker. Tumor-specific and tumor-infiltrating circulating biomarkers have not been compared previously. EXPERIMENTAL DESIGN We prospectively measured sCD163 and sTARC in 221 samples from 47 patients with Hodgkin lymphoma and 21 healthy participants. Blood was taken at five fixed time-points prior, during, and after first-line therapy. Results were compared with radiological assessment and plasma Epstein-Barr virus DNA (EBV-DNA). Potential sources of circulating CD163 were investigated, along with immunosuppressive properties of CD163. RESULTS Pretherapy, both sCD163 and sTARC were markedly elevated compared with healthy and complete remission samples. sCD163 better reflected tumor burden during therapy, whereas sTARC had greater value upon completion of therapy. sCD163 correlated with plasma EBV-DNA, and associated with B symptoms, stage, and lymphopenia. Circulating CD163(+) monocytes were elevated in patients, indicating that sCD163 are likely derived from circulating and intratumoral cells. Depletion of cHL CD163(+) monocytes markedly enhanced T-cell proliferation, implicating monocytes and/or TAMs as potential novel targets for immunotherapeutic manipulation. CONCLUSION The combination of circulating tumor-infiltrate (sCD163) and tumor-specific (sTARC) proteins is more informative than either marker alone as disease response biomarkers in early and advanced disease during first-line therapy for cHL.
Collapse
Affiliation(s)
- Kimberley Jones
- Clinical Immunohaematology Laboratory, Queensland Institute of Medical Research (QIMR), Queensland, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Advances in the treatment of Hodgkin lymphoma. Int J Hematol 2012; 96:535-43. [PMID: 23054655 DOI: 10.1007/s12185-012-1199-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 09/30/2012] [Indexed: 10/27/2022]
|
41
|
Interim FDG-PET in Hodgkin lymphoma: a compass for a safe navigation in clinical trials? Blood 2012; 120:4913-20. [PMID: 22932799 DOI: 10.1182/blood-2012-03-403790] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Despite the rewarding results achieved in the treatment of Hodgkin lymphoma (HL), concerns have been raised regarding the long-term complications induced by therapy. Hence, the current challenge is to develop a new therapeutic strategy maintaining excellent patient outcome while reducing potentially life-threatening late adverse effects. Therefore, it would be beneficial to identify chemoresistant or refractory patients early during therapy for appropriate and timely escalation of treatment. Recently, compelling data have emerged on the prognostic role of interim [18F]-fluoro-2-deoxy-D-glucose positron emission tomography (FDG-PET) performed early during the course of treatment to predict ultimate outcome, even proving superior to conventional prognostic factors. Several ongoing prospective trials are exploring the feasibility of treatment de-escalation strategies in patients with a negative interim PET, as well as therapy escalation in advanced-stage HL patients who have a positive interim PET result. In this article, the published reports on the contribution of interim PET to the design of ongoing response-adapted clinical trials are reviewed. Moreover, some of the unresolved issues revolving around the suboptimal positive predictive value of interim PET are addressed with an emphasis on the interpretation criteria. A final remark on the appropriate use of interim PET is also provided.
Collapse
|
42
|
Guidetti A, Carlo-Stella C, Locatelli SL, Malorni W, Pierdominici M, Barbati C, Mortarini R, Devizzi L, Matteucci P, Marchianò A, Lanocita R, Farina L, Dodero A, Tarella C, Di Nicola M, Corradini P, Anichini A, Gianni AM. Phase II study of sorafenib in patients with relapsed or refractory lymphoma. Br J Haematol 2012; 158:108-19. [PMID: 22571717 DOI: 10.1111/j.1365-2141.2012.09139.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 03/06/2012] [Indexed: 12/15/2022]
Abstract
The safety and activity of the multikinase inhibitor sorafenib were investigated in patients with relapsed or refractory lymphoproliferative disorders who received sorafenib (400 mg) twice daily until disease progression or appearance of significant clinical toxicity. The primary endpoint was overall response rate (ORR). Biomarkers of sorafenib activity were analysed at baseline and during treatment. Thirty patients (median age, 61 years; range, 18-74) received a median of 4 months of therapy. Grade 3-4 toxicities included hand/foot skin reactions (20%), infections (12%), neutropenia (20%) and thrombocytopenia (14%). Two patients achieved complete remission (CR), and two achieved partial remission (PR) for an ORR of 13%. Stable disease (SD) and progressive disease (PD) was observed in 15 (50%) and 11 patients (37%), respectively. The median overall survival (OS) for all patients was 16 months. For patients who achieved CR, PR and SD, the median time to progression and OS was 5 and 24 months, respectively. Compared with patients with PD, responsive patients had significantly higher baseline levels of extracellular signal-regulated kinase phosphorylation and autophagy and presented a significant reduction of these parameters after 1 month of therapy. Sorafenib was well tolerated and had a clinical activity that warrants development of combination regimens.
Collapse
Affiliation(s)
- Anna Guidetti
- Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Furtado M, Rule S. Emerging Pharmacotherapy for Relapsed or Refractory Hodgkin's Lymphoma: Focus on Brentuximab Vedotin. Clin Med Insights Oncol 2012; 6:31-9. [PMID: 22253553 PMCID: PMC3256979 DOI: 10.4137/cmo.s6637] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Hodgkins' lymphoma (HL) which has relapsed post or is refractory to autologous bone marrow transplant presents an ongoing treatment challenge. Development of monoclonal antibodies (mAb) for the treatment of HL has aimed to replicate the success of mAb therapy in the treatment on Non Hodgkins Lymphoma. The identification of CD30 as a potential target for treatment has led to the development of a new antibody-drug conjugate, brentuximab vedotin (SGN-35), which conjugates monomethyl auristatin E to an anti-CD30 antibody to deliver targeted toxicity to the malignant Reed Sternberg cells of HL. This review describes CD30 as an antibody target, and focuses on the antibody-drug conjugate brentuximab vedotin, including current knowledge of the mechanism of action, preclinical, clinical and pharmacokinetic data available for Brentuximab Vedotin.
Collapse
Affiliation(s)
| | - Simon Rule
- Department of Haematology, Derriford Hospital, Plymouth, UK
| |
Collapse
|
45
|
|
46
|
|
47
|
Abstract
AbstractAdvanced-stage Hodgkin lymphoma (HL) has become a curable disease in the majority of patients. Research during the last decade has challenged chemotherapy with Adriamycin, bleomycin, vinblastine, dacarbazine (ABVD) as the standard of care and debates continue regarding the role of radiation therapy (RT) in this patient population. The incorporation of interim positron emission tomography (PET) imaging and, recently, further characterization of HL on cellular and molecular levels are emerging as tools for treatment stratification and predictors of disease status. Newer targeted therapies have emerged that are very effective in the relapsed setting and are actively being explored as frontline therapy. Lastly, the expanding population of survivors cured of HL outnumbers patients with the disease and needs to be monitored for therapy-related late effects.
Collapse
|
48
|
Fehniger TA, Larson S, Trinkaus K, Siegel MJ, Cashen AF, Blum KA, Fenske TS, Hurd DD, Goy A, Schneider SE, Keppel CR, Wagner-Johnston ND, Carson KR, Bartlett NL. A phase 2 multicenter study of lenalidomide in relapsed or refractory classical Hodgkin lymphoma. Blood 2011; 118:5119-25. [PMID: 21937701 PMCID: PMC3217400 DOI: 10.1182/blood-2011-07-362475] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Accepted: 08/29/2011] [Indexed: 01/27/2023] Open
Abstract
Relapsed or refractory (rel/ref) classical Hodgkin lymphoma (cHL) remains a clinical challenge, with limited effective treatment options available after stem cell transplantation. In a multicenter phase 2 study, the efficacy of lenalidomide in rel/ref cHL patients was evaluated at a dose of 25 mg/d on days 1-21 of a 28-day cycle. Patients remained on lenalidomide until disease progression or an unacceptable adverse event (AE) occurred. Thirty-eight cHL patients were enrolled with a median of 4 (range, 2-9) prior therapies; 87% had undergone prior stem cell transplantation and 55% of patients did not respond to their last prior therapy. Of 36 evaluable patients, responses were 1 complete remission (CR), 6 partial remissions (PRs), and 5 patients with stable disease (SD) for ≥ 6 months resulting in an International Working Committee (IWC) objective overall response rate (ORR) of 19% and a cytostatic ORR of 33%. Decreased chemokine (CCL17 and CCL22) plasma levels at 2 weeks were associated with a subsequent response. The treatment was well tolerated, and the most common grade 3/4 AEs were neutropenia (47%), anemia (29%), and thrombocytopenia (18%). Four patients discontinued lenalidomide because of rash, elevated transaminases/bilirubin, and cytopenias. We provide preliminary evidence of lenalidomide's activity in patients with rel/ref cHL, and therefore exploration of lenalidomide in combination with other active agents is warranted. This trial is registered at www.ClinicalTrials.gov as NCT00540007.
Collapse
Affiliation(s)
- Todd A Fehniger
- Division of Oncology, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Plattel WJ, van den Berg A, Visser L, van der Graaf AM, Pruim J, Vos H, Hepkema B, Diepstra A, van Imhoff GW. Plasma thymus and activation-regulated chemokine as an early response marker in classical Hodgkin's lymphoma. Haematologica 2011; 97:410-5. [PMID: 22058214 DOI: 10.3324/haematol.2011.053199] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Plasma thymus and activation-regulated chemokine is a potential biomarker for classical Hodgkin's lymphoma. To define its value as a marker to monitor treatment response, we correlated serial plasma thymus and activation-regulated chemokine levels with clinical response in newly diagnosed and relapsed classical Hodgkin's lymphoma patients. DESIGN AND METHODS Plasma was collected from 60 (39 early stage and 21 advanced stage) newly diagnosed classical Hodgkin's lymphoma patients before, during, and after treatment, and from 12 relapsed patients before and after treatment. Plasma thymus and activation-regulated chemokine levels were determined by enzyme-linked immunosorbent assay and were related to pre-treatment metabolic tumor volume, as measured by quantification of 2-[18F]fluoro-2-deoxyglucose positron emission tomography images, and to treatment response. RESULTS Baseline plasma thymus and activation-regulated chemokine levels correlated with stage of disease and bulky disease, and more closely with metabolic tumor volume. Response to treatment was observed among 38 of 39 early stage and 19 of 21 advanced stage patients. Reduction in plasma thymus and activation-regulated chemokine to normal range levels could be observed as early as after one cycle of chemotherapy in all responsive patients, while plasma levels remained elevated during and after treatment in the 3 non-responsive patients. Plasma thymus and activation-regulated chemokine was elevated in all 12 relapsed patients at time of relapse and remained elevated after salvage treatment in the 4 non-responsive patients. CONCLUSIONS Baseline plasma thymus and activation-regulated chemokine levels correlate with classical Hodgkin's lymphoma tumor burden and serial levels correlate with response to treatment in patients with classical Hodgkin's lymphoma.
Collapse
Affiliation(s)
- Wouter J Plattel
- Department of Hematology, University of Groningen, University Medical Center Groningen, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Pham MN, Hawa MI, Pfleger C, Roden M, Schernthaner G, Pozzilli P, Buzzetti R, Scherbaum WA, Seissler J, Kolb H, Hunter S, Leslie RDG, Schloot NC. Pro- and anti-inflammatory cytokines in latent autoimmune diabetes in adults, type 1 and type 2 diabetes patients: Action LADA 4. Diabetologia 2011; 54:1630-8. [PMID: 21347621 DOI: 10.1007/s00125-011-2088-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 01/24/2011] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS Systemic pro- and anti-inflammatory cytokines are associated with both type 1 and type 2 diabetes, while their role in latent autoimmune diabetes in adults (LADA) is unclear. Therefore, we compared cytokine concentrations in patients with LADA, type 1 or type 2 diabetes and healthy individuals to test the hypothesis that differences of cytokine concentrations between all groups are attributable to diabetes type and BMI. METHODS The pro-inflammatory cytokines IL-6 and TNF-α, and the anti-inflammatory cytokines IL-1 receptor antagonist (IL-1RA) and IL-10 were measured in 90 participants with type 1 diabetes, 61 with LADA, 465 with type 2 diabetes and 41 control participants using multiple regression models adjusted for BMI, sex, age, blood pressure and diabetes duration. RESULTS Patients with type 2 diabetes had higher concentrations of systemic IL-1RA, IL-6 and TNF-α cytokines than patients with either LADA or type 1 diabetes (p < 0.0001 for all differences). Cytokine concentrations in controls were lower than those in all diabetes types (p < 0.04). Increased BMI was positively associated with higher systemic cytokine concentrations in all diabetes types (p < 0.0001). Despite the association of cytokines with anthropometric data, differences between diabetes forms persisted also after adjusting analysis for the confounders BMI, age, sex, disease duration and blood pressure (p < 0.04). CONCLUSIONS/INTERPRETATION Although body mass associates positively with pro- and anti-inflammatory cytokine levels, patients with type 2 diabetes have higher cytokine levels independent of the prevailing BMI. LADA and type 1 diabetes could not be distinguished by systemic cytokines.
Collapse
Affiliation(s)
- M N Pham
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Duesseldorf, Auf'm Hennekamp 65, 40225 Duesseldorf, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|