451
|
Mikkilineni L, Whitaker-Menezes D, Domingo-Vidal M, Sprandio J, Avena P, Cotzia P, Dulau-Florea A, Gong J, Uppal G, Zhan T, Leiby B, Lin Z, Pro B, Sotgia F, Lisanti MP, Martinez-Outschoorn U. Hodgkin lymphoma: A complex metabolic ecosystem with glycolytic reprogramming of the tumor microenvironment. Semin Oncol 2017; 44:218-225. [PMID: 29248133 PMCID: PMC5737784 DOI: 10.1053/j.seminoncol.2017.10.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 10/05/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Twenty percent of patients with classical Hodgkin Lymphoma (cHL) have aggressive disease defined as relapsed or refractory disease to initial therapy. At present we cannot identify these patients pre-treatment. The microenvironment is very important in cHL because non-cancer cells constitute the majority of the cells in these tumors. Non-cancer intra-tumoral cells, such as tumor-associated macrophages (TAMs) have been shown to promote tumor growth in cHL via crosstalk with the cancer cells. Metabolic heterogeneity is defined as high mitochondrial metabolism in some tumor cells and glycolysis in others. We hypothesized that there are metabolic differences between cancer cells and non-cancer tumor cells, such as TAMs and tumor-infiltrating lymphocytes in cHL and that greater metabolic differences between cancer cells and TAMs are associated with poor outcomes. METHODS A case-control study was conducted with 22 tissue samples of cHL at diagnosis from a single institution. The case samples were from 11 patients with aggressive cHL who had relapsed after standard treatment with adriamycin, bleomycin, vinblastine, and dacarbazine (ABVD) or were refractory to this treatment. The control samples were from 11 patients with cHL who achieved a remission and never relapsed after ABVD. Reactive non-cancerous lymph nodes from four subjects served as additional controls. Samples were stained by immunohistochemistry for three metabolic markers: translocase of the outer mitochondrial membrane 20 (TOMM20), monocarboxylate transporter 1 (MCT1), and monocarboxylate transporter 4 (MCT4). TOMM20 is a marker of mitochondrial oxidative phosphorylation (OXPHOS) metabolism. Monocarboxylate transporter 1 (MCT1) is the main importer of lactate into cells and is a marker of OXPHOS. Monocarboxylate transporter 4 (MCT4) is the main lactate exporter out of cells and is a marker of glycolysis. The immunoreactivity for TOMM20, MCT1, and MCT4 was scored based on staining intensity and percentage of positive cells, as follows: 0 for no detectable staining in > 50% of cells; 1+ for faint to moderate staining in > 50% of cells, and 2+ for high or strong staining in > 50% of cells. RESULTS TOMM20, MCT1, and MCT4 expression was significantly different in Hodgkin and Reed Sternberg (HRS) cells, which are the cancerous cells in cHL compared with TAMs and tumor-associated lymphocytes. HRS have high expression of TOMM20 and MCT1, while TAMs have absent expression of TOMM20 and MCT1 in all but two cases. Tumor-infiltrating lymphocytes have low TOMM20 expression and absent MCT1 expression. Conversely, high MCT4 expression was found in TAMs, but absent in HRS cells in all but one case. Tumor-infiltrating lymphocytes had absent MCT4 expression. Reactive lymph nodes in contrast to cHL tumors had low TOMM20, MCT1, and MCT4 expression in lymphocytes and macrophages. High TOMM20 and MCT1 expression in cancer cells with high MCT4 expression in TAMs is a signature of high metabolic heterogeneity between cancer cells and the tumor microenvironment. A high metabolic heterogeneity signature was associated with relapsed or refractory cHL with a hazard ratio of 5.87 (1.16-29.71; two-sided P < .05) compared with the low metabolic heterogeneity signature. CONCLUSION Aggressive cHL exhibits features of metabolic heterogeneity with high mitochondrial metabolism in cancer cells and high glycolysis in TAMs, which is not seen in reactive lymph nodes. Future studies will need to confirm the value of these markers as prognostic and predictive biomarkers in clinical practice. Treatment intensity may be tailored in the future to the metabolic profile of the tumor microenvironment and drugs that target metabolic heterogeneity may be valuable in this disease.
Collapse
Affiliation(s)
- Lekha Mikkilineni
- Department of Medical Oncology, National Cancer Institute, Bethesda, MD
| | | | | | - John Sprandio
- Department of Medical Oncology, Chester County Memorial Hospital, West Chester, PA
| | - Paola Avena
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Calabria, Italy
| | - Paolo Cotzia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Jerald Gong
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA
| | - Guldeep Uppal
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA
| | - Tingting Zhan
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA
| | - Benjamin Leiby
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA
| | - Zhao Lin
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA
| | - Barbara Pro
- Division of Medical Oncology, Northwestern University, Evanston, IL
| | - Federica Sotgia
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Michael P Lisanti
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | | |
Collapse
|
452
|
Kang SP, Gergich K, Lubiniecki GM, de Alwis DP, Chen C, Tice MAB, Rubin EH. Pembrolizumab KEYNOTE-001: an adaptive study leading to accelerated approval for two indications and a companion diagnostic. Ann Oncol 2017; 28:1388-1398. [PMID: 30052728 PMCID: PMC5452070 DOI: 10.1093/annonc/mdx076] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
| | | | | | | | - C. Chen
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | |
Collapse
|
453
|
Li H, Yu J, Liu C, Liu J, Subramaniam S, Zhao H, Blumenthal GM, Turner DC, Li C, Ahamadi M, de Greef R, Chatterjee M, Kondic AG, Stone JA, Booth BP, Keegan P, Rahman A, Wang Y. Time dependent pharmacokinetics of pembrolizumab in patients with solid tumor and its correlation with best overall response. J Pharmacokinet Pharmacodyn 2017; 44:403-414. [PMID: 28573468 DOI: 10.1007/s10928-017-9528-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/23/2017] [Indexed: 12/26/2022]
Abstract
Pembrolizumab is a monoclonal antibody that targets the programmed death-1 receptor to induce immune-mediated clearance (CL) of tumor cells. Originally approved by the US Food and Drug Administration in 2014 for treating patients with unresectable or metastatic melanoma, pembrolizumab is now also used to treat patients with non-small-cell lung cancer, classical Hodgkin lymphoma, head and neck cancer, and urothelial cancer. This paper describes the recently identified feature of pembrolizumab pharmacokinetics, the time-dependent or time-varying CL. Overall results indicate that CL decreases over the treatment period of a typical patient in a pattern well described by a sigmoidal function of time with three parameters: the maximum proportion change in CL from baseline (approximately Imax or exactly eImax - 1), the time to reach Imax/2 (TI50), and a Hill coefficient. Best overall response per response evaluation criteria in solid tumor category was found to be associated with the magnitude of Imax.
Collapse
Affiliation(s)
- Hongshan Li
- Food and Drug Administration, Silver Spring, MD, USA
| | - Jingyu Yu
- Food and Drug Administration, Silver Spring, MD, USA
| | - Chao Liu
- Food and Drug Administration, Silver Spring, MD, USA
| | - Jiang Liu
- Food and Drug Administration, Silver Spring, MD, USA
| | | | - Hong Zhao
- Food and Drug Administration, Silver Spring, MD, USA
| | | | | | - Claire Li
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Rik de Greef
- Quantitative Solutions, A Certara Company, Oss, The Netherlands
| | | | | | | | - Brian P Booth
- Food and Drug Administration, Silver Spring, MD, USA
| | | | - Atiqur Rahman
- Food and Drug Administration, Silver Spring, MD, USA
| | - Yaning Wang
- Food and Drug Administration, Silver Spring, MD, USA. .,Division of Pharmacometrics, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA.
| |
Collapse
|
454
|
Biasoli I, Spector N. New agents in relapsed/refractory Hodgkin's lymphoma. Rev Bras Hematol Hemoter 2017; 39:193-196. [PMID: 28830595 PMCID: PMC5568577 DOI: 10.1016/j.bjhh.2017.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 11/17/2022] Open
Affiliation(s)
- Irene Biasoli
- Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
| | - Nelson Spector
- Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
455
|
Liao J, Luan Y, Ren Z, Liu X, Xue D, Xu H, Sun Z, Yang K, Peng H, Fu YX. Converting Lymphoma Cells into Potent Antigen-Presenting Cells for Interferon-Induced Tumor Regression. Cancer Immunol Res 2017; 5:560-570. [PMID: 28533311 DOI: 10.1158/2326-6066.cir-16-0221] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/09/2016] [Accepted: 05/15/2017] [Indexed: 12/11/2022]
Abstract
Anti-hCD20 is a therapeutic mAb that is clinically used to treat B-cell lymphoma. Some lymphomas are resistant to anti-hCD20; others relapse after treatment with anti-hCD20. Using a syngeneic immunocompetent mouse model, we observed that targeting lymphoma with interferon-α (IFNα) abolished resistance of B-cell lymphoma to anti-CD20 while limiting interferon (IFN)-associated systemic toxicity in the host. Control of tumors by a fusion of anti-CD20 and IFNα (anti-CD20-IFNα) depended on existing tumor-infiltrating CD8+ T cells. Although lymphomas were resistant to IFN-directed killing, IFN-exposed tumor cells became the dominant antigen-presenting cells (APC) for the reactivation of tumor-infiltrating CD8+ T cells that then controlled those lymphomas. Anti-CD20-IFNα also abolished checkpoint blockade resistance in advanced B-cell lymphoma. Our findings indicate that anti-CD20-IFNα eradicates B-cell lymphoma by employing tumor cells as APCs to reactivate tumor-infiltrating CD8+ T cells and synergizing with anti-PD-L1 treatment. Cancer Immunol Res; 5(7); 560-70. ©2017 AACR.
Collapse
Affiliation(s)
- Jing Liao
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yan Luan
- DingFu Biotarget Co. Ltd., Suzhou, Jiangsu, China
| | - Zhenhua Ren
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaojuan Liu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Diyuan Xue
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hairong Xu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhichen Sun
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Kaiting Yang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hua Peng
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
456
|
Siniard RC, Harada S. Immunogenomics: using genomics to personalize cancer immunotherapy. Virchows Arch 2017; 471:209-219. [DOI: 10.1007/s00428-017-2140-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/21/2017] [Accepted: 04/27/2017] [Indexed: 01/06/2023]
|
457
|
Curran EK, Godfrey J, Kline J. Mechanisms of Immune Tolerance in Leukemia and Lymphoma. Trends Immunol 2017; 38:513-525. [PMID: 28511816 DOI: 10.1016/j.it.2017.04.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/05/2017] [Accepted: 04/18/2017] [Indexed: 12/18/2022]
Abstract
The mechanisms through which immune responses are generated against solid cancers are well characterized and knowledge of the immune evasion pathways exploited by these malignancies has grown considerably. However, for hematological cancers, which develop and disseminate quite differently than solid tumors, the pathways that regulate immune activation or tolerance are less clear. Growing evidence suggests that, while numerous immune escape pathways are shared between hematological and solid malignancies, several unique pathways are exploited by leukemia and lymphoma. Below we discuss immune evasion mechanisms in leukemia and lymphoma, highlighting key differences from solid tumors. A more complete characterization of the mechanisms of immune tolerance in hematological malignancies is critical to inform the development of future immunotherapeutic approaches.
Collapse
Affiliation(s)
- Emily K Curran
- Department of Medicine, Section of Hematology, University of Chicago, Chicago, IL, USA; Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL, USA; University of Chicago Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA
| | - James Godfrey
- Department of Medicine, Section of Hematology, University of Chicago, Chicago, IL, USA
| | - Justin Kline
- Department of Medicine, Section of Hematology, University of Chicago, Chicago, IL, USA; University of Chicago Comprehensive Cancer Center, University of Chicago, Chicago, IL, USA; Committee on Immunology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
458
|
Rocha TMBDSD, Fortier SC, Fischer TRDC, Perini GF, Gaiolla RD, Fogliatto L, Delamain MT, Costa AFD, Castro NSD, Barretos WG, Souza CAD, Buccheri V, Chiattone CS. Everolimus as a single agent in refractory or relapsed Hodgkin's lymphoma: the Brazilian Named Patient Program Experience. Rev Bras Hematol Hemoter 2017; 39:216-222. [PMID: 28830600 PMCID: PMC5567422 DOI: 10.1016/j.bjhh.2017.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 03/13/2017] [Accepted: 03/23/2017] [Indexed: 11/22/2022] Open
Abstract
Background Despite all the scientific progress that has been made on understanding the disease, prognosis for patients with relapsed and refractory Hodgkin's lymphoma remains poor and the treatment is palliative in the majority of the cases. Thus, the aim of this study was to present the results on the compassionate use of everolimus in a group of patients who were monitored at nine different centers in Brazil. Methods A 10-mg oral dose of everolimus was given to each patient daily. Response time was evaluated from the beginning of medication use until loss of response, toxicity or medical decision to cease treatment. Results Thirty-three patients were evaluated. The median age at the beginning of medication administration was 29 years. Patients had received a median of five prior therapies. Overall response rate was 45.4%, with 13 patients achieving partial response, two achieved clinical response, 14 remained with stable disease, two had disease progression, and two were not evaluated. Patients received a median of 14 cycles. Progression-free survival was nine months, and overall survival was estimated to be 36 months. Three patients used the medication for more than four years. The most frequently reported adverse events were thrombocytopenia and hypercholesterolemia. Three patients had pulmonary toxicity. Grade III and IV adverse events occurred in 39% of the patients. Conclusion Everolimus was found to provide a response in a group of patients with refractory or relapsed Hodgkin's lymphoma who had adequate tolerability to the drug.
Collapse
Affiliation(s)
| | - Sergio Costa Fortier
- Faculdade de Ciências Médicas da Santa Casa de São Paulo (FCMSCSP), São Paulo, SP, Brazil
| | | | | | - Rafael Dezen Gaiolla
- Faculdade de Ciências Médicas da Santa Casa de São Paulo (FCMSCSP), São Paulo, SP, Brazil
| | - Laura Fogliatto
- Faculdade de Ciências Médicas da Santa Casa de São Paulo (FCMSCSP), São Paulo, SP, Brazil
| | | | | | | | - Wolney Gois Barretos
- Faculdade de Ciências Médicas da Santa Casa de São Paulo (FCMSCSP), São Paulo, SP, Brazil
| | | | - Valéria Buccheri
- Faculdade de Ciências Médicas da Santa Casa de São Paulo (FCMSCSP), São Paulo, SP, Brazil
| | | |
Collapse
|
459
|
Bond DA, Alinari L. Emerging treatment options for the management of Hodgkin's lymphoma: clinical utility of nivolumab. J Blood Med 2017; 8:41-54. [PMID: 28546779 PMCID: PMC5436782 DOI: 10.2147/jbm.s117452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Classical Hodgkin's lymphoma (cHL) is a B-cell malignancy comprised of pathologic Reed Sternberg cells with a surrounding immune-tolerant inflammatory milieu. RS cells evade immune recognition in part through programmed death ligand 1 (PD-L1) overexpression, which is genetically programmed through copy number alterations, polysomy, and amplification of the 9p24.1 locus encoding PD-L1. By engaging with PD-1+ T-cells, PD-L1 delivers a potent immune suppressive signal promoting immunologic escape of the tumor cell. Enhancing antitumor immune response by targeting PD-1 with the monoclonal antibody nivolumab has proved to be effective in multiple solid tumors, but the highest response rates to date have been reported in patients with cHL, with over 65% of treated patients achieving an objective clinical response. In this review, we will summarize the published evidence regarding the activity of nivolumab in cHL as well as its current place in therapy. We will review the pharmacology, mechanism of action, and side effects of nivolumab as well as the emerging data indicating possible increased risk of graft versus host disease in patients treated with PD-1 inhibitors either pre- or post-allogeneic stem cell transplant. Given the remarkable single-agent activity and safety profile of PD-1 inhibitors in heavily pretreated patients with cHL, the possibility of employing nivolumab in combination with other active agents and earlier in therapy is a promising area of active investigation, and we will briefly summarize current clinical trials.
Collapse
Affiliation(s)
- David A Bond
- Department of Internal Medicine, Division of Hematology, Arthur G James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lapo Alinari
- Department of Internal Medicine, Division of Hematology, Arthur G James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
460
|
Safety and tolerability of pembrolizumab in patients with relapsed/refractory primary mediastinal large B-cell lymphoma. Blood 2017; 130:267-270. [PMID: 28490569 DOI: 10.1182/blood-2016-12-758383] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/24/2017] [Indexed: 01/13/2023] Open
Abstract
Treatment options for relapsed/refractory primary mediastinal large B-cell lymphoma (rrPMBCL) are limited, and prognosis is generally poor (overall response rate [ORR] 0% to 25%; 2-year overall survival 15%). PMBCL frequently involves PD-1 ligand overexpression, potentially making PMBCL particularly susceptible to PD-1 blockade. We evaluated safety and antitumor activity of pembrolizumab, an anti-PD-1 antibody, in rrPMBCL as part of the KEYNOTE-013 multicohort phase 1b trial. At time of data cutoff, 18 patients (median age 30 years; median 3 prior lines of therapy) had been enrolled and treated, of whom 17 were included in the efficacy analyses. Eleven patients (61%) experienced drug-related adverse events (mostly grade 1-2); none discontinued treatment due to adverse events. ORR was 41% (7/17); 6 additional patients (35%) had stable disease. Of patients evaluable by imaging, 13 out of 16 (81%) had decreases in target lesions. With a median follow-up of 11.3 months, median duration of response was not reached. Two patients reached the maximum 2-year treatment duration and remain in remission. Median overall survival was not reached for treated patients overall; all responders were still alive at data cutoff. These results in heavily pretreated rrPMBCL patients demonstrate that PD-1 blockade with pembrolizumab has a manageable safety profile and promising antitumor activity. This trial was registered at www.clinicaltrials.gov as #NCT01953692.
Collapse
|
461
|
Ma C, Horlad H, Pan C, Yano H, Ohnishi K, Fujiwara Y, Matsuoka M, Lee A, Niidome T, Yamanaka R, Takeya M, Komohara Y. Stat3 inhibitor abrogates the expression of PD-1 ligands on lymphoma cell lines. J Clin Exp Hematop 2017; 57:21-25. [PMID: 28496056 DOI: 10.3960/jslrt.17006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Recent studies have indicated the significance of immune checkpoint molecules including programmed death-1 (PD-1), cytotoxic T-lymphocyte associated protein 4, and T-cell immunoglobulin and mucin domain-containing molecule-3 for anti-tumor immune responses. We previously investigated PD-1 ligand 1/2 (PD-L1/2) expression in lymphoma cell lines, and found that PD-L1/2 is expressed on the adult T-cell leukemia/lymphoma (ATL-T) and B-cell lymphoma (SLVL) cell lines. In the present study, we investigated whether the Stat3 inhibitor WP1066 abrogated PD-L1/2 expression in lymphoma cell lines. Incubation with WP1066 inhibited lymphoma cell growth and induced cell apoptosis. PD-L1/2 expression in the ATL-T, SLVL, and human brain malignant lymphoma (HKBML) cell lines was significantly abrogated by WP1066 treatment. These data indicated that a Stat3 inhibitor abrogated PD-L1/2 expression in lymphoma cells. Such an inhibitor is therefore considered to be useful for additional immunotherapy in patients with advanced lymphoma.
Collapse
Affiliation(s)
- Chaoya Ma
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Hasita Horlad
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Cheng Pan
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Hiromu Yano
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Koji Ohnishi
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Masao Matsuoka
- Department of Hematology, Graduate School of Medical Sciences, Kumamoto University.,Laboratory of Virus Control, Institute for Virus Research, Kyoto University
| | - Aeju Lee
- International Research Organization for Advanced Science and Technology (IROAST), Kumamoto University.,Magnesium Research Center, Kumamoto University
| | - Takuro Niidome
- Faculty of Advanced Science and Technology, Kumamoto University
| | - Ryuya Yamanaka
- Kyoto Prefectural University of Medicine, Graduate School for Health Care Science
| | - Motohiro Takeya
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| |
Collapse
|
462
|
Karantanos T, Politikos I, Boussiotis VA. Advances in the pathophysiology and treatment of relapsed/refractory Hodgkin's lymphoma with an emphasis on targeted therapies and transplantation strategies. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2017; 7:37-52. [PMID: 28701859 PMCID: PMC5502320 DOI: 10.2147/blctt.s105458] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Hodgkin’s lymphoma (HL) is highly curable with first-line therapy. However, a minority of patients present with refractory disease or experience relapse after completion of frontline treatment. These patients are treated with salvage chemotherapy followed by autologous stem cell transplantation (ASCT), which remains the standard of care with curative potential for refractory or relapsed HL. Nevertheless, a significant percentage of such patients will progress after ASCT, and allogeneic hematopoietic stem cell transplantation remains the only curative approach in that setting. Recent advances in the pathophysiology of refractory or relapsed HL have provided the rationale for the development of novel targeted therapies with potent anti-HL activity and favorable toxicity profile, in contrast to cytotoxic chemotherapy. Brentuximab vedotin and programmed cell death-1-based immunotherapy have proven efficacy in the management of refractory or relapsed HL, whereas several other agents have shown promise in early clinical trials. Several of these agents are being incorporated with transplantation strategies in order to improve the outcomes of refractory or relapsed HL. In this review we summarize the current knowledge regarding the mechanisms responsible for the development of refractory/relapsed HL and the outcomes with current treatment strategies, with an emphasis on targeted therapies and hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Theodoros Karantanos
- General Internal Medicine Section, Boston Medical Center, Boston University School of Medicine, Boston, MA
| | - Ioannis Politikos
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
463
|
Abstract
Inhibitory molecules such as PD-1, CTLA-4, LAG-3, or TIM-3 play a role to keep a balance in immune function. However, many cancers exploit such molecules to escape immune surveillance. Accumulating data support that their functions are dysregulated in lymphoid neoplasms, including plasma cell myeloma, myelodysplastic syndrome, and acute myeloid leukemia. In lymphoid neoplasms, aberrations in 9p24.1 (PD-L1, PD-L2, and JAK2 locus), latent Epstein-Barr virus infection, PD-L1 3'-untranslated region disruption, and constitutive JAK-STAT pathway are known mechanisms to induce PD-L1 expression in lymphoma cells. Clinical trials demonstrated that PD-1 blockade is an attractive way to restore host's immune function in hematological malignancies, particularly classical Hodgkin lymphoma. Numerous clinical trials exploring PD-1 blockade as a single therapy or in combination with other immune checkpoint inhibitors in patients with hematologic cancers are under way. Although impressive clinical response is observed with immune checkpoint inhibitors in patients with certain cancers, not all patients respond to immune checkpoint inhibitors. Therefore, to identify best candidates who would have excellent response to checkpoint inhibitors is of utmost importance. Several possible biomarkers are available, but consensus has not been made and pursuit to discover the best biomarker is ongoing.
Collapse
Affiliation(s)
- Chi Young Ok
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009 USA
| | - Ken H. Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009 USA
| |
Collapse
|
464
|
Pai SI, Faquin WC. Programmed cell death ligand 1 as a biomarker in head and neck cancer. Cancer Cytopathol 2017; 125:529-533. [PMID: 28472542 DOI: 10.1002/cncy.21872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 12/24/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Sara I Pai
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - William C Faquin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
465
|
PD-1 blockade for relapsed lymphoma post-allogeneic hematopoietic cell transplant: high response rate but frequent GVHD. Blood 2017; 130:221-228. [PMID: 28468799 DOI: 10.1182/blood-2017-01-761346] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/24/2017] [Indexed: 12/21/2022] Open
Abstract
Given the limited treatment options for relapsed lymphoma post-allogeneic hematopoietic cell transplantation (post-allo-HCT) and the success of programmed death 1 (PD-1) blockade in classical Hodgkin lymphoma (cHL) patients, anti-PD-1 monoclonal antibodies (mAbs) are increasingly being used off-label after allo-HCT. To characterize the safety and efficacy of PD-1 blockade in this setting, we conducted a multicenter retrospective analysis of 31 lymphoma patients receiving anti-PD-1 mAbs for relapse post-allo-HCT. Twenty-nine (94%) patients had cHL and 27 had ≥1 salvage therapy post-allo-HCT and prior to anti-PD-1 treatment. Median follow-up was 428 days (range, 133-833) after the first dose of anti-PD-1. Overall response rate was 77% (15 complete responses and 8 partial responses) in 30 evaluable patients. At last follow-up, 11 of 31 patients progressed and 21 of 31 (68%) remain alive, with 8 (26%) deaths related to new-onset graft-versus-host disease (GVHD) after anti-PD-1. Seventeen (55%) patients developed treatment-emergent GVHD after initiation of anti-PD-1 (6 acute, 4 overlap, and 7 chronic), with onset after a median of 1, 2, and 2 doses, respectively. GVHD severity was grade III-IV acute or severe chronic in 9 patients. Only 2 of these 17 patients achieved complete response to GVHD treatment, and 14 of 17 required ≥2 systemic therapies. In conclusion, PD-1 blockade in relapsed cHL allo-HCT patients appears to be highly efficacious but frequently complicated by rapid onset of severe and treatment-refractory GVHD. PD-1 blockade post-allo-HCT should be studied further but cannot be recommended for routine use outside of a clinical trial.
Collapse
|
466
|
Filippi AR, Levis M, Parikh R, Hoppe B. Optimal Therapy for Early-Stage Hodgkin's Lymphoma: Risk Adapting, Response Adapting, and Role of Radiotherapy. Curr Oncol Rep 2017; 19:34. [PMID: 28365830 DOI: 10.1007/s11912-017-0592-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PURPOSE OF REVIEW The aim of this article is to discuss the current role of radiotherapy (RT) for early-stage Hodgkin's lymphoma (HL) in the context of risk-adapted and response-adapted treatment strategy, and describe changes in RT technical approach. RECENT FINDINGS In low-risk patients, RT could be omitted but, at the price of a lower progression-free survival, and its role is still debated. Ongoing trials are combining new agents with chemotherapy alone or response-adapted combined modality therapy, and results are awaited. Modern RT incorporates lower doses and smaller fields, together with the implementation of sophisticated delivery techniques aimed to reducing the dose to critical structures such as the heart. The role of RT for early-stage HL is still under debate, and new combinations are emerging; an individualized approach should be recommended, considering all RT technical opportunities to minimize toxicity while maintaining efficacy.
Collapse
Affiliation(s)
- Andrea Riccardo Filippi
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Torino, Regione Gonzole 10, 10043, Orbassano, Turin, Italy.
| | - Mario Levis
- Department of Oncology, San Luigi Gonzaga University Hospital, University of Torino, Regione Gonzole 10, 10043, Orbassano, Turin, Italy
| | - Rahul Parikh
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, USA
| | - Bradford Hoppe
- Department of Radiation Oncology, University of Florida, Gainesville, USA.,University of Florida Health Proton Therapy Institute, Jacksonville, USA
| |
Collapse
|
467
|
Hude I, Sasse S, Bröckelmann PJ, von Tresckow B, Momotow J, Engert A, Borchmann S. Leucocyte and eosinophil counts predict progression-free survival in relapsed or refractory classical Hodgkin Lymphoma patients treated with PD1 inhibition. Br J Haematol 2017; 181:837-840. [PMID: 28439879 DOI: 10.1111/bjh.14705] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Ida Hude
- German Hodgkin Study Group (GHSG), Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany.,Department of Internal Medicine, Division of Haematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Stephanie Sasse
- German Hodgkin Study Group (GHSG), Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Paul J Bröckelmann
- German Hodgkin Study Group (GHSG), Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Bastian von Tresckow
- German Hodgkin Study Group (GHSG), Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Jesko Momotow
- German Hodgkin Study Group (GHSG), Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Andreas Engert
- German Hodgkin Study Group (GHSG), Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Sven Borchmann
- German Hodgkin Study Group (GHSG), Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
468
|
Chen R, Zinzani PL, Fanale MA, Armand P, Johnson NA, Brice P, Radford J, Ribrag V, Molin D, Vassilakopoulos TP, Tomita A, von Tresckow B, Shipp MA, Zhang Y, Ricart AD, Balakumaran A, Moskowitz CH. Phase II Study of the Efficacy and Safety of Pembrolizumab for Relapsed/Refractory Classic Hodgkin Lymphoma. J Clin Oncol 2017; 35:2125-2132. [PMID: 28441111 DOI: 10.1200/jco.2016.72.1316] [Citation(s) in RCA: 726] [Impact Index Per Article: 103.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Hodgkin Reed-Sternberg cells harbor alterations in chromosome 9p24.1, leading to overexpression of programmed death-ligand 1 (PD-L1) and PD-L2. Pembrolizumab, a programmed death 1-blocking antibody, demonstrated a high overall response rate (ORR) in patients with relapsed or refractory classic Hodgkin lymphoma (rrHL) in phase I testing. Methods KEYNOTE-087 ( ClinicalTrials.gov identifier, NCT02453594) was a single-arm phase II study of pembrolizumab in three cohorts of patients with rrHL, defined on the basis of lymphoma progression after (1) autologous stem cell transplantation (ASCT) and subsequent brentuximab vedotin (BV); (2) salvage chemotherapy and BV, and thus, ineligible for ASCT because of chemoresistant disease; and (3) ASCT, but without BV after transplantation. Patients received pembrolizumab 200 mg once every 3 weeks. Response was assessed every 12 weeks. The primary end points were ORR by central review and safety. Results A total of 210 patients were enrolled and treated (69 in cohort 1, 81 in cohort 2, and 60 in cohort 3). At the time of analysis, patients received a median of 13 treatment cycles. Per central review, the ORR was 69.0% (95% CI, 62.3% to 75.2%), and the complete response rate was 22.4% (95% CI, 16.9% to 28.6%). By cohort, ORRs were 73.9% for cohort 1, 64.2% for cohort 2, and 70.0% for cohort 3. Thirty-one patients had a response ≥ 6 months. The safety profile was largely consistent with previous pembrolizumab studies. Conclusion Pembrolizumab was associated with high response rates and an acceptable safety profile in patients with rrHL, offering a new treatment paradigm for this disease.
Collapse
Affiliation(s)
- Robert Chen
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pier Luigi Zinzani
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michelle A Fanale
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Philippe Armand
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nathalie A Johnson
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pauline Brice
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - John Radford
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Vincent Ribrag
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel Molin
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Theodoros P Vassilakopoulos
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Akihiro Tomita
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bastian von Tresckow
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Margaret A Shipp
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yinghua Zhang
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alejandro D Ricart
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Arun Balakumaran
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Craig H Moskowitz
- Robert Chen, City of Hope National Medical Center, Duarte, CA; Pier Luigi Zinzani, University of Bologna, Bologna, Italy; Michelle A. Fanale, The University of Texas MD Anderson Cancer Center, Houston, TX; Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Nathalie A. Johnson, Jewish General Hospital, Montreal, Canada; Pauline Brice, Hôpital Saint-Louis, Paris; Vincent Ribrag, Institut Gustave Roussy, Villejuif, France; John Radford, The University of Manchester and Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom; Daniel Molin, Uppsala University, Uppsala, Sweden; Theodoros P. Vassilakopoulos, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; Akihiro Tomita, Nagoya University Graduate School of Medicine, Nagoya, Japan; Bastian von Tresckow, University Hospital Cologne, Cologne, Germany; Yinghua Zhang, Alejandro D. Ricart, and Arun Balakumaran, Merck, Kenilworth, NJ; Craig H. Moskowitz, Memorial Sloan Kettering Cancer Center, New York, NY
| | | |
Collapse
|
469
|
Im A, Pavletic SZ. Immunotherapy in hematologic malignancies: past, present, and future. J Hematol Oncol 2017; 10:94. [PMID: 28434396 PMCID: PMC5402171 DOI: 10.1186/s13045-017-0453-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/29/2017] [Indexed: 12/25/2022] Open
Abstract
The field of immunotherapy in cancer treatments has been accelerating over recent years and has entered the forefront as a leading area of ongoing research and promising therapies that have changed the treatment landscape for a variety of solid malignancies. Prior to its designation as the Science Breakthrough of the Year in 2013, cancer immunotherapy was active in the treatment of hematologic malignancies. This review provides a broad overview of the past, present, and potential future of immunotherapy in hematologic malignancies.
Collapse
Affiliation(s)
- Annie Im
- University of Pittsburgh Cancer Institute, 5150 Centre Ave, Suite 554, Pittsburgh, PA 15213 USA
| | - Steven Z. Pavletic
- National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
470
|
Low-dose nivolumab induced remission in refractory classical Hodgkin lymphoma. Ann Hematol 2017; 96:1219-1220. [DOI: 10.1007/s00277-017-3007-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 04/18/2017] [Indexed: 10/19/2022]
|
471
|
Guldbrandsen KF, Hendel HW, Langer SW, Fischer BM. Nuclear Molecular Imaging Strategies in Immune Checkpoint Inhibitor Therapy. Diagnostics (Basel) 2017; 7:diagnostics7020023. [PMID: 28430133 PMCID: PMC5489943 DOI: 10.3390/diagnostics7020023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 04/12/2017] [Accepted: 04/18/2017] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitor therapy (ICT) is a new treatment strategy developed for the treatment of cancer. ICT inhibits pathways known to downregulate the innate immune response to cancer cells. These drugs have been shown to be effective in the treatment of a variety of cancers, including metastatic melanoma and lung cancer. Challenges in response evaluation of patients in ICT have risen as immune related side effects and immune cell infiltration may be confused with progressive disease. Furthermore, the timing of the evaluation scan may be challenged by relatively slow responses. To overcome this, new response criteria for evaluating these patients with morphologic imaging have been proposed. The aim of this paper is to review and discuss the current evidence for the use of molecular imaging, e.g., PET/CT (Positron Emission Tomography/Computer Tomography) with 18F-Fluorodeoxyglucoes (FDG) as an alternative imaging method for monitoring patients undergoing ICT. Following the currently available evidence, this review will primarily focus on patients with malignant melanoma.
Collapse
Affiliation(s)
- Kasper F Guldbrandsen
- Department of Pulmonary and Infectious Diseases, Nordsjællands Hospital Hillerød, 3400 Hillerød, Denmark.
| | - Helle W Hendel
- Department of Clinical Physiology and Nuclear Medicine, Herlev and Gentofte Hospital, 2750 Herlev, Denmark.
| | - Seppo W Langer
- Department of Oncology 5073, Rigshospitalet, 2100 Copenhagen, Denmark.
| | - Barbara M Fischer
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, 2100 Copenhagen, Denmark.
| |
Collapse
|
472
|
Rosner S, Sen F, Postow M. Response after treatment with pembrolizumab in a patient with myelophthisis due to melanoma: the role of checkpoint inhibition in the bone. J Immunother Cancer 2017; 5:34. [PMID: 28428883 PMCID: PMC5394614 DOI: 10.1186/s40425-017-0236-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/24/2017] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Myelophthisis due to melanoma is a rare phenomenon. Treatment strategies for patients with this serious complication of malignancy have not been well documented, and none have previously reported efficacy of immune checkpoint inhibition. Since bone metastases are not measurable lesions per standard response criteria, the efficacy of immune checkpoint inhibition in the bones is also not well described. CASE PRESENTATION We describe a patient with widespread melanoma metastases involving the bone marrow causing myelophthisis and pancytopenia who responded to immune checkpoint inhibition with the anti-programmed cell death-1 (PD-1) inhibitor pembrolizumab. CONCLUSIONS This is the first report to our knowledge of disease response to immune checkpoint inhibition in a patient with myelophthisis. Clinical trials have recently emerged describing the efficacy of PD-1 inhibition for disorders regularly involving the bone marrow, such as hematologic malignancies, suggesting the importance of better understanding the bone marrow as an immunologically active compartment. Clinicians should be aware that immune checkpoint inhibition alone may be effective in treating malignancy involving the bone marrow, even in cases of extensive involvement resulting in pancytopenia due to myelophthisis from a solid tumor as our case suggests.
Collapse
Affiliation(s)
- Samuel Rosner
- Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461 USA
| | - Filiz Sen
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 USA
| | - Michael Postow
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 USA
- Weill Cornell Medical College, New York, USA
| |
Collapse
|
473
|
Donato EM, Fernández-Zarzoso M, De La Rubia J. Immunotherapy for the treatment of Hodgkin lymphoma. Expert Rev Hematol 2017; 10:417-423. [PMID: 28359170 DOI: 10.1080/17474086.2017.1313701] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Most patients with Hodgkin lymphoma (HL) enjoy durable remissions following front-line treatment but 30% of patients are refractory or relapse after first line therapy. Salvage chemotherapy followed by autologous stem cell transplantation (ASCT) can cure an additional 50-55% of relapsing patients but new treatments are needed for patients with HL who are refractory or relapse after ASCT. Immunotherapy has emerged as a promising treatment for the management of these patients. The availability of the anti-CD30 antibody brentuximab vedotin and new targeted drugs such as immune checkpoint inhibitors, show promising clinical activity in patients with HL and are important milestones for the management of patients with HL particularly for those who have progressed after standard initial therapy and ASCT. Areas covered: Overview of the results from the most relevant clinical trials including monoclonal antibody-based therapy in HL. Phase 2 and phase 3 trials including brentuximab vedotin and immune checkpoints inhibitors in patients with Hodgkin lymphoma have been reviewed. In addition, the potential impact of these new therapies in the management of patients with newly diagnosed HL has also been addressed. Expert commentary: Anti-CD30 antibody brentuximab vedotin and immune checkpoint inhibitors have shown promising results in patients with relapsed and refractory HL. Administration of these therapies earlier in the course of the disease might reduce the proportion of relapsed or refractory patients and, subsequently, minimize the number of patients undergoing high-dose therapy and autologous stem cell transplantation. We have little doubt that this will have substantial effects on the outcome for future generations of HL patients.
Collapse
Affiliation(s)
- Eva M Donato
- a Hematology Service , University Hospital Doctor Peset , Valencia , Spain
| | | | - Javier De La Rubia
- a Hematology Service , University Hospital Doctor Peset , Valencia , Spain.,b Department of Internal Medicine , Universidad Católica de Valencia "San Vicente Mártir" , Valencia , Spain
| |
Collapse
|
474
|
El Cheikh J, Massoud R, Abudalle I, Haffar B, Mahfouz R, Kharfan-Dabaja MA, Jisr T, Mougharbel A, Ibrahim A, Bazarbachi A. Nivolumab salvage therapy before or after allogeneic stem cell transplantation in Hodgkin lymphoma. Bone Marrow Transplant 2017; 52:1074-1077. [PMID: 28394366 DOI: 10.1038/bmt.2017.69] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- J El Cheikh
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - R Massoud
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - I Abudalle
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - B Haffar
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - R Mahfouz
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
| | - M A Kharfan-Dabaja
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - T Jisr
- Division of Hematology and Oncology, Makassed University Hospital, Beirut, Lebanon
| | - A Mougharbel
- Division of Hematology and Oncology, Makassed University Hospital, Beirut, Lebanon
| | - A Ibrahim
- Division of Hematology and Oncology, Makassed University Hospital, Beirut, Lebanon
| | - A Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
475
|
Iwai Y, Hamanishi J, Chamoto K, Honjo T. Cancer immunotherapies targeting the PD-1 signaling pathway. J Biomed Sci 2017; 24:26. [PMID: 28376884 PMCID: PMC5381059 DOI: 10.1186/s12929-017-0329-9] [Citation(s) in RCA: 455] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 03/20/2017] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has recently emerged as the fourth pillar of cancer treatment, joining surgery, radiation, and chemotherapy. While early immunotherapies focused on accelerating T-cell activity, current immune-checkpoint inhibitors take the brakes off the anti-tumor immune responses. Successful clinical trials with PD-1 monoclonal antibodies and other immune-checkpoint inhibitors have opened new avenues in cancer immunology. However, the failure of a large subset of cancer patients to respond to these new immunotherapies has led to intensified research on combination therapies and predictive biomarkers. Here we summarize the development of PD-1-blockade immunotherapy and current issues in its clinical use.
Collapse
Affiliation(s)
- Yoshiko Iwai
- Department of Molecular Biology, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu-shi, Fukuoka, 807-8555, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Chamoto
- Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
476
|
PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood 2017; 129:3071-3073. [PMID: 28356247 DOI: 10.1182/blood-2017-01-764209] [Citation(s) in RCA: 329] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 02/25/2017] [Indexed: 12/16/2022] Open
Abstract
Primary central nervous system (CNS) lymphoma (PCNSL) and primary testicular lymphoma (PTL) are rare extranodal large B-cell lymphomas with similar genetic signatures. There are no standard-of-care treatment options for patients with relapsed and refractory PCNSL and PTL, and the overall prognosis is poor. PCNSLs and PTLs exhibit frequent 9p24.1 copy-number alterations and infrequent translocations of 9p24.1 and associated increased expression of the programmed cell death protein 1 (PD-1) ligands, PD-L1 and PD-L2. The activity of PD-1 blockade in other lymphomas with 9p24.1 alterations prompted us to test the efficacy of the anti-PD1 antibody, nivolumab, in 4 patients with relapsed/refractory PCNSL and 1 patient with CNS relapse of PTL. All 5 patients had clinical and radiographic responses to PD-1 blockade, and 3 patients remain progression-free at 13+ to 17+ months. Our data suggest that nivolumab is active in relapsed/refractory PCNSL and PTL and support further investigation of PD-1 blockade in these diseases.
Collapse
|
477
|
Gay ND, Okada CY, Chen AI, Scott EC. Targeting the programmed cell death 1 pathway in Hodgkin lymphoma: the place of nivolumab. Ther Adv Hematol 2017; 8:175-180. [PMID: 28473905 DOI: 10.1177/2040620717695723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Nivolumab is a humanized immunoglobulin gamma-4 kappa anti-programmed cell death 1 monoclonal antibody that is currently approved in the treatment of several solid tumors and recently gained accelerated approval in classical Hodgkin lymphoma (cHL) that has relapsed or progressed following autologous hematopoietic stem-cell transplantation and post-transplantation brentuximab vedotin. The purpose of this article is to review the immunophysiologic basis, clinical efficacy, and toxicity of nivolumab in the treatment of cHL. In addition, we will review ongoing clinical trials and potential future directions of checkpoint inhibition in the treatment of cHL.
Collapse
Affiliation(s)
- Nathan D Gay
- Knight Cancer Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Craig Y Okada
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Andy I Chen
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Emma C Scott
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
478
|
Covut F, Pinto R, Cooper BW, Tomlinson B, Metheny L, Malek E, Lazarus HM, de Lima M, Caimi PF. Nivolumab before and after allogeneic hematopoietic cell transplantation. Bone Marrow Transplant 2017; 52:1054-1056. [PMID: 28346414 DOI: 10.1038/bmt.2017.44] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- F Covut
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - R Pinto
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - B W Cooper
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - B Tomlinson
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - L Metheny
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - E Malek
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - H M Lazarus
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - M de Lima
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - P F Caimi
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Cleveland, OH, USA
| |
Collapse
|
479
|
Abstract
Classical Hodgkin lymphoma (cHL) is the most common hematological malignancy in young adults and can be cured in most cases. However, relapsed and refractory Hodgkin lymphoma, certain patient groups, such as elderly patients, and toxicity of first-line treatment still pose significant challenges. Consequently, new treatment options are needed. Recently, many new treatment concepts have been evaluated in clinical trials. Targeted drug-antibody conjugates and immune checkpoint inhibitors have decisively changed treatment approaches. This review aims to give a comprehensive overview of novel agents in Hodgkin lymphoma that have been recently or are currently being evaluated in clinical trials. In addition to dedicated sections on brentuximab vedotin (BV) and immune checkpoint inhibitors, other emerging substances and concepts are discussed. In doing so, this review compares trial results regarding safety and efficacy. A special focus lies on the effect novel agents will have on the different treatment settings faced by clinicians involved in the treatment of Hodgkin lymphoma.
Collapse
Affiliation(s)
- Sven Borchmann
- a German Hodgkin Study Group (GHSG), Department I of Internal Medicine , University Hospital Cologne , Cologne , Germany
| | - Bastian von Tresckow
- a German Hodgkin Study Group (GHSG), Department I of Internal Medicine , University Hospital Cologne , Cologne , Germany
| |
Collapse
|
480
|
The role of anti-PD-1 and anti-PD-L1 agents in the treatment of diffuse large B-cell lymphoma: The future is now. Crit Rev Oncol Hematol 2017; 113:52-62. [PMID: 28427522 DOI: 10.1016/j.critrevonc.2017.02.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 02/07/2017] [Accepted: 02/10/2017] [Indexed: 11/23/2022] Open
Abstract
Immune checkpoints inhibitors have been incorporated into standard treatment protocols for advanced solid tumors. The aim of T-cell-based immune therapy in cancer has been to generate durable clinical benefits for patients, paired with enhanced side effect profiles. The beneficial antitumoral activity of programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) has been thoroughly demonstrated in certain metastatic malignancies (e.g. melanoma, non-small cell lung cancer, renal cell carcinoma); however, the therapeutic role in lymphoid cancers is complex. Nonetheless, the striking clinical activity seen in early clinical trials of various subtypes of relapsed lymphoma have paved the way for these exciting innovative therapeutic alternatives in these tumors. In this article we assess the literature on the role of the PD-1/PD-L1 pathway in Diffuse Large B-cell lymphoma (DLBCL), and describe future strategies involving these new anticancer agents in this lymphoid neoplasm.
Collapse
|
481
|
Ok CY, Young KH. Targeting the programmed death-1 pathway in lymphoid neoplasms. Cancer Treat Rev 2017; 54:99-109. [PMID: 28242522 PMCID: PMC5815314 DOI: 10.1016/j.ctrv.2017.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 01/23/2017] [Accepted: 01/29/2017] [Indexed: 12/31/2022]
Abstract
Programmed death-1 (PD-1) is a co-inhibitory molecule and is seen in CD4+ and CD8+ T cells. Upon binding to its ligands, programmed death ligand-1 (PD-L1) and -2 (PD-L2), PD-1 negatively regulates interleukin 2 (IL-2) production and T cell proliferation. Activated effector T-cells, which kill cancer cells, can be affected by PD-1 signaling in some lymphoid neoplasm that express PD-L1 or PD-L2. PD-L1 expression in tumor cells can be induced by extrinsic signal (i.e. interferon gamma) or intrinsic signals, such as genetic aberrations involving 9p24.1, latent Epstein-Barr virus infection, PD-L1 3'- untranslated region disruptions, and activated Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. Anti-PD-1 therapy improves the overall response rate to treatment in patients with lymphoid neoplasms, particularly relapsed/refractory classical Hodgkin lymphoma. Inspired by their success in treating patients with classical Hodgkin lymphoma, medical practitioners have expanded PD-1 therapy, given as a single therapy or in combination with other drugs, to patients with other types of lymphoma. In this review, current clinical trials with anti-PD-1 or anti-PD-L1 drugs are summarized. The results of numerous clinical trials will broaden our understanding of PD-1 pathway and shall expand the list of patients who will get benefit from these agents including those who suffer from lymphoid neoplasms.
Collapse
Affiliation(s)
- Chi Young Ok
- The Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ken H Young
- The Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
482
|
Abstract
Treatment of classical Hodgkin's lymphoma (HL) has been a success story, with cure of localized disease with radiotherapy in the 1930s, cure of advanced stages with combination chemotherapy with/without radiotherapy in the mid-1960s and continuous improvements since then. Nonetheless, at present approximately 2% of patients with classical HL are primarily refractory to conventional therapy with only 50% becoming long-term survivors. Another 13% of patients relapse, with only 60% being alive 10 years postrecurrence (as exemplified in this review in a Swedish cohort of 18- to 65-year-old patients diagnosed during the period 1992-2009). Recently, novel targeted drugs were approved for refractory/relapsed HL and here we review results of trials that form the basis for these approvals as well as new trials. In summary, brentuximab vedotin can be used in refractory patients (i) as a complement to high-dose chemotherapy with autologous stem cell transplantation (SCT) improving the chances of being able to proceed to an allogenic SCT and cure, (ii) as consolidation after autologous SCT and (iii) as palliative life-prolonging treatment. However, we have yet to determine whether this drug provides the greatest benefit in first- or second-line treatment, as consolidation or in refractory disease or relapse. Trials of immune checkpoint inhibitors, such as those targeting programmed death 1 (nivolumab and pembrolizumab), and thus not primarily the tumour cells, have shown overall response rates of >65%. Long-term results and data from Phase III trials are still lacking, but nivolumab recently gained approval in refractory patients already treated with brentuximab vedotin and autologous SCT. Other novel treatments of interest include T cells with a chimeric antigen receptor and combination therapies with histone deacetylase inhibitors.
Collapse
Affiliation(s)
- I Glimelius
- Department of Immunology, Genetics and Pathology, Section of Clinical and Experimental Oncology, Uppsala University, Uppsala, Sweden.,Department of Medicine, Clinical Epidemiology Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - A Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
483
|
Koh YW, Han JH, Park SY, Yoon DH, Suh C, Huh J. GLUT1 as a Prognostic Factor for Classical Hodgkin's Lymphoma: Correlation with PD-L1 and PD-L2 Expression. J Pathol Transl Med 2017; 51:152-158. [PMID: 28219001 PMCID: PMC5357756 DOI: 10.4132/jptm.2016.11.03] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/17/2016] [Accepted: 11/03/2016] [Indexed: 11/24/2022] Open
Abstract
Background Glucose transporter type 1 (GLUT1) expression is linked to glucose metabolism and tissue hypoxia. A recent study reported that GLUT1 was significantly associated with programmed death ligand 1 (PD-L1) as a therapeutic target in relapsed or refractory classical Hodgkin’s lymphoma (cHL). The purpose of this study was to measure the expression of GLUT1 and assess its prognostic significance and potential relationships with PD-L1, programmed death ligand 2 (PD-L2), and programmed death-1 (PD-1) expressions in cHL. Methods Diagnostic tissues from 125 patients with cHL treated with doxorubicin, bleomycin, vinblastine, and dacarbazine were evaluated retrospectively via immunohistochemical analysis of GLUT1, PD-L1, PD-L2, and PD-1 expression. Results The median follow-up time was 4.83 years (range, 0.08 to 17.33 years). GLUT1, PD-L1, PD-L2, and PD-1 were expressed in 44.8%, 63.2%, 9.6%, and 13.6% of the specimens, respectively. Positive correlations were found between GLUT1 and PD-L1 expression (p = .004) and between GLUT1 and PD-L2 expression (p = .031). GLUT1 expression in Hodgkin/Reed-Sternberg (HRS) cells was not associated with overall survival or event-free survival (EFS) in the entire cohort (p = .299 and p = .143, respectively). A subgroup analysis according to the Ann Arbor stage illustrated that GLUT1 expression in HRS cells was associated with better EFS in advanced-stage disease (p = .029). A multivariate analysis identified GLUT1 as a marginally significant prognostic factor for EFS (p = .068). Conclusions This study suggests that GLUT1 expression is associated with better clinical outcomes in advanced-stage cHL and is significantly associated with PD-L1 and PD-L2 expressions.
Collapse
Affiliation(s)
- Young Wha Koh
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Jae-Ho Han
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Seong Yong Park
- Department of Thoracic and Cardiovascular Surgery, Ajou University School of Medicine, Suwon, Korea
| | - Dok Hyun Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Cheolwon Suh
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jooryung Huh
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
484
|
Oncale MB, Maymani H, Nastoupil LJ. Harnessing the immune system through programmed death-1 blockade in the management of Hodgkin lymphoma. Blood Lymphat Cancer 2017; 7:1-7. [PMID: 31360081 PMCID: PMC6467338 DOI: 10.2147/blctt.s110665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Immunotherapy is a rapidly evolving therapeutic option in the treatment of lymphoma. Neoplastic cells evade immune recognition through the programmed death (PD)-1/PD-ligand immune checkpoint pathway. Several novel agents have been developed to restore the immune system's ability to recognize and destroy cancer cells. Nivolumab and pembrolizumab are two anti-PD-1 antibodies that have demonstrated success in the treatment of refractory Hodgkin lymphoma. Harnessing the immune system's ability to target neoplastic cells, ideally without the use of cytotoxic chemotherapeutic agents, is one way in which these novel agents are changing the therapeutic landscape in the treatment of lymphomas. Here, we review the emerging data regarding checkpoint inhibitors in the management of Hodgkin lymphoma, the unique adverse effects encountered with the use of these agents, and a practical approach to the management of these adverse effects. Additionally, we discuss upcoming trials that will further assess the promising future developments of checkpoint inhibition in the treatment of not only Hodgkin lymphoma but also other B cell lymphomas and myeloma. These agents offer immense promise of a future where many lymphomas can be treated without the toxic effects of chemotherapeutic agents.
Collapse
Affiliation(s)
- Melody B Oncale
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,
| | - Hossein Maymani
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,
| | - Loretta J Nastoupil
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,
| |
Collapse
|
485
|
Jethava Y, Guru Murthy GS, Hamadani M. Relapse of Hodgkin lymphoma after autologous transplantation: Time to rethink treatment? Hematol Oncol Stem Cell Ther 2017; 10:47-56. [PMID: 28183681 DOI: 10.1016/j.hemonc.2016.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/07/2016] [Accepted: 12/29/2016] [Indexed: 11/29/2022] Open
Abstract
Relapse of Hodgkin lymphoma after autologous hematopoietic cell transplantation (autologous HCT) is a major therapeutic challenge. Its management, at least in younger patients, traditionally involves salvage chemotherapy aiming to achieve disease remission followed by consolidation with allogeneic hematopoietic cell transplantation (allogeneic HCT) in eligible patients. The efficacy of salvage therapy is variable and newer combination chemotherapy regimens have improved the outcomes. Factors such as shorter time to relapse after autologous HCT and poor performance status have been identified as predictors of poor outcome. Newer agents such as immunoconjugate brentuximab vedotin, checkpoint inhibitors (e.g., pembrolizumab, nivolumab), lenalidomide, and everolimus are available for the treatment of patients relapsing after autologous HCT. With the availability of reduced intensity conditioning allogeneic HCT, more patients are eligible for this therapy with lesser toxicity and better efficacy due to graft versus lymphoma effects. Alternative donor sources such as haploidentical stem cell transplantation and umbilical cord blood transplantation are expanding this procedure to patients without HLA-matched donors. However, strategies aimed at reduction of disease relapse after reduced intensity conditioning allogeneic HCT are needed to improve the outcomes of this treatment. This review summarizes the current data on salvage chemotherapy and HCT strategies used to treat patients with relapsed Hodgkin lymphoma after prior autologous HCT.
Collapse
Affiliation(s)
- Yogesh Jethava
- Division of Hematology-Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Mehdi Hamadani
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
486
|
Chan TSY, Luk TH, Lau JSM, Khong PL, Kwong YL. Low-dose pembrolizumab for relapsed/refractory Hodgkin lymphoma: high efficacy with minimal toxicity. Ann Hematol 2017; 96:647-651. [PMID: 28138786 DOI: 10.1007/s00277-017-2931-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 01/20/2017] [Indexed: 01/13/2023]
Abstract
Five patients with refractory/relapsed classical Hodgkin lymphoma (cHL), four having failed multiple lines of chemotherapy and brentuximab vedotin, were treated with low-dose pembrolizumab (median dose 100 mg, range: 100-200 mg, every 3 weeks). Complete response (CR) was achieved in four patients (80%), after a median cumulative dose of merely 495 (300-800) milligrams. Three CR patients have continued to receive pembrolizumab for a median of 16 (14-25) cycles, remaining in CR for a median of 18 (9-18) months. One CR patient underwent autologous hematopoietic stem cell transplantation and has remained in CR for 9 months. Partial response (PR) was achieved in one patient (20%), after a cumulative dose of 400 mg. The overall response rate was therefore 100% (CR: 80%; PR: 20%). Toxicity was virtually absent, with only grade 1 diarrhea and eczema each observed in one patient. Low-dose pembrolizumab was highly efficacious, achieving responses with minimal toxicity and at much lower costs.
Collapse
Affiliation(s)
- Thomas S Y Chan
- Department of Medicine, Professorial Block, Queen Mary Hospital, Pokfulam Road, Hong Kong, China
| | - Tsan-Hei Luk
- Department of Medicine, Queen Elizabeth Hospital, Hong Kong, China
| | - June S M Lau
- Department of Medicine, Queen Elizabeth Hospital, Hong Kong, China
| | - Pek-Lan Khong
- Department of Diagnostic Radiology, Queen Mary Hospital, Hong Kong, China
| | - Yok-Lam Kwong
- Department of Medicine, Professorial Block, Queen Mary Hospital, Pokfulam Road, Hong Kong, China.
| |
Collapse
|
487
|
Botticelli A, Zizzari I, Mazzuca F, Ascierto PA, Putignani L, Marchetti L, Napoletano C, Nuti M, Marchetti P. Cross-talk between microbiota and immune fitness to steer and control response to anti PD-1/PDL-1 treatment. Oncotarget 2017; 8:8890-8899. [PMID: 27806346 PMCID: PMC5352451 DOI: 10.18632/oncotarget.12985] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/13/2016] [Indexed: 12/22/2022] Open
Abstract
Immune Checkpoint Inhibitors (ICIs) are improving the survival of cancer patients, however only the 20-30% of treated patients present clinical benefits. Toxicity represents the major cause of reduced dosage, delayed drug administration and therapy discontinuation. Hence in the context of multiple treatment possibilities, the identification of predictive markers of response and toxicity is a challenging approach for drug selection in order to obtain the best clinical benefit while minimizing the side effects. The loss of the protective function of intestinal barriers that interacts with the environment measured as increased intestinal permeability and the changes occurring in the microbiota composition have been proposed as a mechanism potentially explaining the pathogenesis of immune related toxicity.In this review we discuss the new perspectives on the involvement of PD-1 and PDL-1 in the cross talk between gut microbiota and immune fitness and how gut microbiota impacts on the efficacy of anti-PD-1 and anti-PDL-1 treatments in cancer.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Clinical and Molecular Medicine, SantAndrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Ilaria Zizzari
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Mazzuca
- Department of Clinical and Molecular Medicine, SantAndrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy, and Innovative Therapy, Istituto nazionale Tumori Fondazione G Pascale, Napoli, Italy
| | - Lorenza Putignani
- Units of Parasitology and Human Microbiome, Bambino Ges Childrens Hospital and Research Institute, Rome, Italy
| | - Luca Marchetti
- Department of Clinical Oncology, Policlinico Umberto I, University of Rome Sapienza, Rome, Italy
| | - Chiara Napoletano
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marianna Nuti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, SantAndrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
488
|
Hendriks D, Choi G, de Bruyn M, Wiersma VR, Bremer E. Antibody-Based Cancer Therapy: Successful Agents and Novel Approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:289-383. [PMID: 28325214 DOI: 10.1016/bs.ircmb.2016.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since their discovery, antibodies have been viewed as ideal candidates or "magic bullets" for use in targeted therapy in the fields of cancer, autoimmunity, and chronic inflammatory disorders. A wave of antibody-dedicated research followed, which resulted in the clinical approval of a first generation of monoclonal antibodies for cancer therapy such as rituximab (1997) and cetuximab (2004), and infliximab (2002) for the treatment of autoimmune diseases. More recently, the development of antibodies that prevent checkpoint-mediated inhibition of T cell responses invigorated the field of cancer immunotherapy. Such antibodies induced unprecedented long-term remissions in patients with advanced stage malignancies, most notably melanoma and lung cancer, that do not respond to conventional therapies. In this review, we will recapitulate the development of antibody-based therapy, and detail recent advances and new functions, particularly in the field of cancer immunotherapy. With the advent of recombinant DNA engineering, a number of rationally designed molecular formats of antibodies and antibody-derived agents have become available, and we will discuss various molecular formats including antibodies with improved effector functions, bispecific antibodies, antibody-drug conjugates, antibody-cytokine fusion proteins, and T cells genetically modified with chimeric antigen receptors. With these exciting advances, new antibody-based treatment options will likely enter clinical practice and pave the way toward more successful control of malignant diseases.
Collapse
Affiliation(s)
- D Hendriks
- Department of Surgery, Translational Surgical Oncology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - G Choi
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - M de Bruyn
- Department of Obstetrics & Gynecology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - V R Wiersma
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
| | - E Bremer
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands; University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
489
|
Galanina N, Kline J, Bishop MR. Emerging role of checkpoint blockade therapy in lymphoma. Ther Adv Hematol 2017; 8:81-90. [PMID: 28203344 DOI: 10.1177/2040620716673787] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Following the successful application of immune checkpoint blockade therapy (CBT) in refractory solid tumors, it has recently gained momentum as a promising modality in the treatment of relapsed lymphoma. This significant therapeutic advance stems from decades of research that elucidated the role of immune regulation pathways and the mechanisms by which tumors can engage these critical pathways to escape immune detection. To date, two main pathways, the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed death 1 (PD-1), have emerged as key targets of CBT demonstrating unprecedented activity particularly in heavily pretreated relapsed/refractory Hodgkin lymphoma and some forms of non-Hodgkin disease. Herein we provide a brief discussion of checkpoint blockade in the context of lymphoma biology with a specific focus on novel checkpoint inhibitors and their therapeutic activity. We discuss current clinical trials and the landscape of CBT to underscore both the remarkable progress and foreseeable limitations of this novel treatment strategy. In particular, we build upon state-of-the-art knowledge and clinical insights gained from the early trials to review potential approaches to how CBT may be integrated with other treatment modalities, including chemoimmunotherapy to improve patient outcomes in the future. Finally, as the role of CBT evolves to potentially become a cornerstone of therapy in refractory/relapsed lymphoma, we briefly emphasize the importance of predictive biomarkers in an effort to select appropriate patients who are most likely to derive benefit from CBT.
Collapse
Affiliation(s)
- Natalie Galanina
- Department of Hematology/Oncology, UC San Diego Moores Cancer Center, La Jolla, California, USA
| | - Justin Kline
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Michael R Bishop
- Section of Hematology and Oncology, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637, USA
| |
Collapse
|
490
|
Greil R, Hutterer E, Hartmann TN, Pleyer L. Reactivation of dormant anti-tumor immunity - a clinical perspective of therapeutic immune checkpoint modulation. Cell Commun Signal 2017; 15:5. [PMID: 28100240 PMCID: PMC5244547 DOI: 10.1186/s12964-016-0155-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/06/2016] [Indexed: 12/17/2022] Open
Abstract
In favor of their outgrowth, cancer cells must resist immune surveillance and edit the immune response. Cancer immunoediting is characterized by fundamental changes in the cellular composition and the inflammatory cytokine profiles in the microenvironment of the primary tumor and metastatic niches, with an ever increasing complexity of interactions between tumor cells and the immune system. Recent data suggest that genetic instability and immunoediting are not necessarily disparate processes. Increasing mutational load may be associated with multiple neoepitopes expressed by the tumor cells and thus increased chances for the immune system to recognize and combat these cells. At the same time the immune system is more and more suppressed and exhausted by this process. Consequently, immune checkpoint modulation may have the potential to be most successful in genetically highly altered and usually extremely unfavorable types of cancer. Moreover, the fact that epitopes recognized by the immune system are preferentially encoded by passenger gene mutations opens windows of synergy in targeting cancer-specific signaling pathways by small molecules simultaneously with antibodies modifying T-cell activation or exhaustion. This review covers some aspects of the current understanding of the immunological basis necessary to understand the rapidly developing therapeutic endeavours in cancer treatment, the clinical achievements made, and raises some burning questions for translational research in this field.
Collapse
Affiliation(s)
- Richard Greil
- Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Disease and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, A-5020, Salzburg, Austria. .,Salzburg Cancer Research Institute (SCRI) - Laboratory for Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria. .,Arbeitsgemeinschaft Medikamentöse Tumortherapie (AGMT) Study Group, Salzburg, Austria. .,Cancer Cluster Salzburg (CCS), Salzburg, Austria.
| | - Evelyn Hutterer
- Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Disease and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, A-5020, Salzburg, Austria.,Salzburg Cancer Research Institute (SCRI) - Laboratory for Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria.,Cancer Cluster Salzburg (CCS), Salzburg, Austria
| | - Tanja Nicole Hartmann
- Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Disease and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, A-5020, Salzburg, Austria.,Salzburg Cancer Research Institute (SCRI) - Laboratory for Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria.,Cancer Cluster Salzburg (CCS), Salzburg, Austria
| | - Lisa Pleyer
- Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Disease and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, A-5020, Salzburg, Austria.,Salzburg Cancer Research Institute (SCRI) - Laboratory for Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria.,Arbeitsgemeinschaft Medikamentöse Tumortherapie (AGMT) Study Group, Salzburg, Austria.,Cancer Cluster Salzburg (CCS), Salzburg, Austria
| |
Collapse
|
491
|
Haebe S, Weigert O. It's a long way to the top (if you want to personalize immunotherapy). J Immunother Cancer 2017; 5:6. [PMID: 28116089 PMCID: PMC5240409 DOI: 10.1186/s40425-016-0207-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/28/2016] [Indexed: 01/21/2023] Open
Abstract
Harnessing the immune system to attack tumor cells by targeting tumor-associated or –preferably– tumor-specific antigens has emerged as a promising but challenging treatment option for malignant lymphomas. Follicular lymphoma is among the most common lymphomas worldwide and remains incurable for most patients. Considered to be an immunogenic disease it represents an interesting disease entity for various immunotherapeutic approaches. In an article published in the May issue of Clinical Cancer Research, Nielsen and colleagues provided important proof-of-principle data on the immunogenicity of follicular lymphoma that might represent a first step towards personalized adoptive immunotherapies in this disease. The authors combined targeted next-generation sequencing and in silico analyses to explore the concept of somatic neoepitope prediction. Neoantigen-specific CD8+ T-cells could be identified in a small subset of patients selected for in vitro immunogenicity experiments, however at remarkably low frequencies and in only a few patients at single time-points. Of note, the immunogenic neoepitopes were derived from mutant CREBBP and MEF2B, two genes that have previously been shown to be functionally and prognostically relevant in this disease. In this commentary we discuss the promises but also the challenges of how to translate these findings into clinical practice.
Collapse
Affiliation(s)
- Sarah Haebe
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Max-Lebsche Platz 30, 81377 Munich, Germany
| | - Oliver Weigert
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Max-Lebsche Platz 30, 81377 Munich, Germany ; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
492
|
Safety and efficacy of allogeneic hematopoietic stem cell transplant after PD-1 blockade in relapsed/refractory lymphoma. Blood 2017; 129:1380-1388. [PMID: 28073785 DOI: 10.1182/blood-2016-09-738385] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/29/2016] [Indexed: 12/24/2022] Open
Abstract
Anti-programmed cell death protein 1 (PD-1) monoclonal antibodies are being increasingly tested in patients with advanced lymphoma. Following treatment, many of those patients are likely to be candidates for allogeneic hematopoietic stem cell transplant (HSCT). However, the safety and efficacy of HSCT may be affected by prior PD-1 blockade. We conducted an international retrospective analysis of 39 patients with lymphoma who received prior treatment with a PD-1 inhibitor, at a median time of 62 days (7-260) before HSCT. After a median follow-up of 12 months, the 1-year cumulative incidences of grade 2-4 and grade 3-4 acute graft-versus-host disease (GVHD) were 44% and 23%, respectively, whereas the 1-year incidence of chronic GVHD was 41%. There were 4 treatment-related deaths (1 from hepatic sinusoidal obstruction syndrome, 3 from early acute GVHD). In addition, 7 patients developed a noninfectious febrile syndrome shortly after transplant requiring prolonged courses of steroids. One-year overall and progression-free survival rates were 89% (95% confidence interval [CI], 74-96) and 76% (95% CI, 56-87), respectively. One-year cumulative incidences of relapse and nonrelapse mortality were 14% (95% CI, 4-29) and 11% (95% CI, 3-23), respectively. Circulating lymphocyte subsets were analyzed in 17 patients. Compared with controls, patients previously treated with PD-1 blockade had significantly decreased PD-1+ T cells and decreased ratios of T-regulatory cells to conventional CD4 and CD8 T cells. In conclusion, HSCT after PD-1 blockade appears feasible with a low rate of relapse. However, there may be an increased risk of early immune toxicity, which could reflect long-lasting immune alterations triggered by prior PD-1 blockade.
Collapse
|
493
|
Tumor regression concomitant with steroid-refractory GvHD highlights the pitfalls of PD-1 blockade following allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2017; 52:759-761. [PMID: 28067871 DOI: 10.1038/bmt.2016.346] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
494
|
Dynamic versus static biomarkers in cancer immune checkpoint blockade: unravelling complexity. Nat Rev Drug Discov 2017; 16:264-272. [PMID: 28057932 DOI: 10.1038/nrd.2016.233] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recently, there has been a coordinated effort from academic institutions and the pharmaceutical industry to identify biomarkers that can predict responses to immune checkpoint blockade in cancer. Several biomarkers have been identified; however, none has reliably predicted response in a sufficiently rigorous manner for routine use. Here, we argue that the therapeutic response to immune checkpoint blockade is a critical state transition of a complex system. Such systems are highly sensitive to initial conditions, and critical transitions are notoriously difficult to predict far in advance. Nevertheless, warning signals can be detected closer to the tipping point. Advances in mathematics and network biology are starting to make it possible to identify such warning signals. We propose that these dynamic biomarkers could prove to be useful in distinguishing responding from non-responding patients, as well as facilitate the identification of new therapeutic targets for combination therapy.
Collapse
|
495
|
Programmed cell death-1 pathway inhibition in myeloid malignancies: implications for myeloproliferative neoplasms. Ann Hematol 2017; 96:919-927. [DOI: 10.1007/s00277-016-2915-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/25/2016] [Indexed: 01/22/2023]
|
496
|
Braschi-Amirfarzan M, Tirumani SH, Hodi FS, Nishino M. Immune-Checkpoint Inhibitors in the Era of Precision Medicine: What Radiologists Should Know. Korean J Radiol 2017; 18:42-53. [PMID: 28096717 PMCID: PMC5240494 DOI: 10.3348/kjr.2017.18.1.42] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/11/2016] [Indexed: 12/15/2022] Open
Abstract
Over the past five years immune-checkpoint inhibitors have dramatically changed the therapeutic landscape of advanced solid and hematologic malignancies. The currently approved immune-checkpoint inhibitors include antibodies to cytotoxic T-lymphocyte antigen-4, programmed cell death (PD-1), and programmed cell death ligand (PD-L1 and PD-L2). Response to immune-checkpoint inhibitors is evaluated on imaging using the immune-related response criteria. Activation of immune system results in a unique toxicity profile termed immune-related adverse events. This article will review the molecular mechanism, clinical applications, imaging of immune-related response patterns and adverse events associated with immune-checkpoint inhibitors.
Collapse
Affiliation(s)
- Marta Braschi-Amirfarzan
- Department of Radiology, Brigham and Women's Hospital and Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Sree Harsha Tirumani
- Department of Radiology, Brigham and Women's Hospital and Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Frank Stephen Hodi
- Department of Medical Oncology and Medicine, Dana Farber Cancer Institue, Boston, MA 02215, USA
| | - Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital and Dana Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
497
|
Sun R, Wang J, Young KH. Oncogenic Signaling Pathways and Pathway-Based Therapeutic Biomarkers in Lymphoid Malignancies. Crit Rev Oncog 2017; 22:527-557. [PMID: 29604930 PMCID: PMC5961736 DOI: 10.1615/critrevoncog.2017020816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lymphoma is characterized by heterogeneous biology, pathologic features, and clinical outcome. This has been proven by accumulating pathologic and molecular evidence attributed to underlying aberrant alterations at genetic, epigenetic, transcriptional, protein, microenvironmental levels, and dysregulated oncogenic signaling pathways. In the era of precision medicine, targeting oncogenic pathways to design drugs and to optimize treatment regimens for the lymphoma patients is feasible and clinically significant. As such, further understanding of the biology and the mechanisms behind lymphoma development and identification of oncogenic pathway activation and pathway-based biomarkers to better design precise therapies are challenging but hopeful. Furthermore, pathway-based targeted therapies in combination with traditional chemotherapy, single specific targeted antibody therapy, and immunotherapy might raise the hope for the patients with lymphoma, especially for relapsed and refractory lymphoma patients.
Collapse
Affiliation(s)
- Ruifang Sun
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
- Tumor Biobank, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Jinfen Wang
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Ken H. Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
498
|
Jiménez-Ubieto A, Rodriguez A, Martinez Sánchez P, Gómez A, Rodriguez Y, Carreño-Tarragona G, Martinez-López J, Grande C. Fatal graft-versus-host disease after allogeneic stem cell transplantation in a patient recently exposed to nivolumab. J Oncol Pharm Pract 2017; 25:1078155217743069. [PMID: 29207936 DOI: 10.1177/1078155217743069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation and checkpoint blockade therapy are immune-based salvage therapies for Hodgkin's lymphoma; however, the use of programmed death 1 blocking agents in the allogeneic stem cell transplantation setting could augment the incidence of steroid refractory graft-versus-host disease. Few studies suggest that that nivolumab is safe in patients previously treated with an allogeneic stem cell transplantation. Likewise, there are very limited data on the use of nivolumab before allogeneic stem cell transplantation. Here, we report a case of fatal graft-versus-host disease in a patient who underwent allogeneic stem cell transplantation 26 days after the last administration of nivolumab. Careful monitoring and close clinical assessment of atypical presentation for graft-versus-host disease in these patients, interval of time from nivolumab administration to allogeneic stem cell transplantation, drug dosage adjustments or more effective allo prophilaxys should been evaluated in prospective clinical trial.
Collapse
Affiliation(s)
- Ana Jiménez-Ubieto
- 1 Department of Hematology, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| | - Antonia Rodriguez
- 1 Department of Hematology, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| | | | - Adolfo Gómez
- 2 Department of Nuclear Medicine, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| | - Yolanda Rodriguez
- 3 Department of Pathological Anatomy, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| | | | | | - Carlos Grande
- 1 Department of Hematology, 16473 Hospital Universitario 12 de Octubre , Madrid, Spain
| |
Collapse
|
499
|
Hude I, Sasse S, Engert A, Bröckelmann PJ. The emerging role of immune checkpoint inhibition in malignant lymphoma. Haematologica 2017; 102:30-42. [PMID: 27884973 PMCID: PMC5210230 DOI: 10.3324/haematol.2016.150656] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/19/2016] [Indexed: 12/19/2022] Open
Abstract
To evade elimination by the host immune system, tumor cells commonly exploit physiological immune checkpoint pathways, restraining efficient anti-tumor immune cell function. Growing understanding of the complex dialog between tumor cells and their microenvironment contributed to the development of immune checkpoint inhibitors. This innovative strategy has demonstrated paradigm-shifting clinical activity in various malignancies. Antibodies targeting programmed death 1 and cytotoxic T-lymphocyte-associated protein-4 are also being investigated in lymphoid malignancies with varying levels of activity and a favorable toxicity profile. To date, evaluated only in the setting of relapsed or refractory disease, anti-programmed death 1 antibodies such as nivolumab and pembrolizumab show encouraging response rates particularly in classical Hodgkin lymphoma but also in follicular lymphoma and diffuse-large B-cell lymphoma. As the first immune checkpoint inhibitor in lymphoma, nivolumab was approved for the treatment of relapsed or refractory classical Hodgkin lymphoma by the Food and Drug Administration in May 2016. In this review, we assess the role of the pathways involved and potential rationale of checkpoint inhibition in various lymphoid malignancies. In addition to data from current clinical trials, immune-related side effects, potential limitations and future perspectives including promising combinatory approaches with immune checkpoint inhibition are discussed.
Collapse
Affiliation(s)
- Ida Hude
- Department of Internal Medicine, Division of Hematology, University Hospital Center Zagreb, Croatia
| | - Stephanie Sasse
- Department I of Internal Medicine and German Hodgkin Study Group (GHSG), University Hospital of Cologne, Germany
| | - Andreas Engert
- Department I of Internal Medicine and German Hodgkin Study Group (GHSG), University Hospital of Cologne, Germany
| | - Paul J Bröckelmann
- Department I of Internal Medicine and German Hodgkin Study Group (GHSG), University Hospital of Cologne, Germany
| |
Collapse
|
500
|
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
|