401
|
Abstract
Lymphoid malignancies typically promote an infiltrate of immune cells at sites involved by the disease. While some of the immune cells present in lymphoma have effector function, the immune system is unable to eradicate the malignant clone. Therapies that optimize immune function therefore have the potential to improve the outcome of lymphoma patients. In this Review, we discuss immunologic approaches that directly target the malignant cell as well as approaches to optimize both the innate and adaptive immune response to the tumor. While many of these therapies have shown single-agent activity, the future will clearly require thoughtful combinations of these approaches.
Collapse
|
402
|
Kansara RR, Speziali C. Immunotherapy in hematologic malignancies. ACTA ACUST UNITED AC 2020; 27:S124-S131. [PMID: 32368182 DOI: 10.3747/co.27.5117] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The management of hematologic malignancies has traditionally relied on chemotherapy regimens, many of which are still in use today. However, with advancements in the knowledge of tumour pathophysiology, therapies are continually evolving. Monoclonal antibodies against specific targets on tumour cells are now widely used to treat hematologic malignancies, either in combination with chemotherapy or as single agents. Rituximab, a monoclonal antibody against the CD20 antigen, is a good example of successful monoclonal antibody therapy that has improved outcomes for patients with B cell non-Hodgkin lymphomas. Monoclonal antibodies are now being used against the immune checkpoints that function to inhibit T cell activation and subsequent tumour eradication by those cytotoxic T cells. Such therapies enhance T cell-mediated tumour eradication and are widely successful in treating patients with solid tumours such as malignant melanoma. Now, they are slowly finding their place in the management of hematologic neoplasms. Even though, currently, immune checkpoint inhibitors are used for relapsed or refractory hematologic neoplasms, trials are ongoing to evaluate their role in frontline treatment. Our review focuses on the current use of immunotherapies in various hematologic malignancies.
Collapse
Affiliation(s)
- R R Kansara
- Section of Medical Oncology/Hematology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB
| | - C Speziali
- Section of Medical Oncology/Hematology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB
| |
Collapse
|
403
|
Penter L, Wu CJ. Personal tumor antigens in blood malignancies: genomics-directed identification and targeting. J Clin Invest 2020; 130:1595-1607. [PMID: 31985488 PMCID: PMC7108890 DOI: 10.1172/jci129209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hematological malignancies have long been at the forefront of the development of novel immune-based treatment strategies. The earliest successful efforts originated from the extensive body of work in the field of allogeneic hematopoietic stem cell transplantation. These efforts laid the foundation for the recent exciting era of cancer immunotherapy, which includes immune checkpoint blockade, personal neoantigen vaccines, and adoptive T cell transfer. At the heart of the specificity of these novel strategies is the recognition of target antigens presented by malignant cells to T cells. Here, we review the advances in systematic identification of minor histocompatibility antigens and neoantigens arising from personal somatic alterations or recurrent driver mutations. These exciting efforts pave the path for the implementation of personalized combinatorial cancer therapy.
Collapse
Affiliation(s)
- Livius Penter
- Department of Hematology, Oncology, and Tumor Immunology, Charité – Universitätsmedizin Berlin (CVK), Berlin, Germany
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
404
|
Yang J, Dong L, Yang S, Han X, Han Y, Jiang S, Yao J, Zhang Z, Zhang S, Liu P, Qin Y, Wu H, Feng H, Yao S, Sun Y, Song H, Shi Y. Safety and clinical efficacy of toripalimab, a PD-1 mAb, in patients with advanced or recurrent malignancies in a phase I study. Eur J Cancer 2020; 130:182-192. [PMID: 32224416 DOI: 10.1016/j.ejca.2020.01.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE This is a first-in-human phase I study investigating the safety and efficacy of toripalimab, a humanized monoclonal antibody against the programmed cell death-1 (PD-1) receptor, in Chinese patients with advanced or recurrent malignant tumor refractory to standard treatment. PATIENTS AND METHODS During dose escalation, patients received a single-dose intravenous infusion of toripalimab for 56 days followed by multidose infusions every two weeks. The planned dosing groups were 1, 3 and 10 mg/kg. During dose expansion, patients received toripalimab every two weeks. Clinical response was evaluated by investigators every 6 weeks. RESULTS Thirty-three patients were enrolled, including 12 patients with alveolar soft part sarcoma (ASPS), seven with non-small-cell lung cancer and 11 with lymphoma. Patients were heavily pretreated with a median of 3 prior lines of systemic treatments. Toripalimab was well tolerated without dose-limiting toxicity. All patients experienced treatment-related adverse events. Grade 3 and above treatment-related adverse events occurred in six (18.2%) patients. Among 22 solid tumors, the objective response rate (ORR) was 22.7% per RECIST v1.1. The ORR was 90.9% in 11 lymphoma patients per IWG 2007. The median duration of response was 21.5 months. The median progression-free survival was 5.7 months for solid tumors and 8.3 months for lymphomas. The median OS was not reached for all patients and the lymphoma subgroups. The median OS was 34.7 months for patients with ASPS. CONCLUSION Toripalimab was well tolerated up to 10 mg/kg Q2W without dose-limiting toxicity and showed promising and durable antitumour activities in patients with ASPS and lymphoma, who were heavily pretreated. CLINICAL TRIAL INFORMATION ClinicalTrials.gov Identifier: NCT02836834.
Collapse
Affiliation(s)
- Jianliang Yang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Lihou Dong
- Institute of Lifeomics, Academy of Military Medical Sciences, National Engineering Research Center for Protein Drugs, Beijing, China
| | - Sheng Yang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Xiaohong Han
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Ying Han
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Shiyu Jiang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Jiarui Yao
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Zhishang Zhang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Shuxiang Zhang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Peng Liu
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Yan Qin
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Hai Wu
- Shanghai Junshi Bioscience Co., Ltd, Shanghai, China
| | - Hui Feng
- Shanghai Junshi Bioscience Co., Ltd, Shanghai, China
| | - Sheng Yao
- Shanghai Junshi Bioscience Co., Ltd, Shanghai, China
| | - Yan Sun
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Haifeng Song
- Institute of Lifeomics, Academy of Military Medical Sciences, National Engineering Research Center for Protein Drugs, Beijing, China
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China.
| |
Collapse
|
405
|
The microenvironmental niche in classic Hodgkin lymphoma is enriched for CTLA-4-positive T cells that are PD-1-negative. Blood 2020; 134:2059-2069. [PMID: 31697809 DOI: 10.1182/blood.2019002206] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022] Open
Abstract
Classic Hodgkin lymphoma (cHL) is a tumor composed of rare, atypical, germinal center-derived B cells (Hodgkin Reed-Sternberg [HRS] cells) embedded within a robust but ineffective inflammatory milieu. The cHL tumor microenvironment (TME) is compartmentalized into "niches" rich in programmed cell death-1 ligand (PD-L1)-positive HRS cells and tumor-associated macrophages (TAMs), which associate with PD-1-positive T cells to suppress antitumor immunity via PD-L1/PD-1 signaling. Despite the exquisite sensitivity of cHL to PD-1 checkpoint blockade, most patients eventually relapse and need therapeutic alternatives. Using multiplex immunofluorescence microscopy with digital image analysis, we found that cHL is highly enriched for non-T-regulatory, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4)-positive T cells (compared with reactive lymphoid tissues) that outnumber PD-1-positive and lymphocyte-activating gene-3 (LAG-3)-positive T cells. In addition, T cells touching HRS cells are more frequently positive for CTLA-4 than for PD-1 or LAG-3. We further found that HRS cells, and a subset of TAMs, are positive for the CTLA-4 ligand CD86 and that the fractions of T cells and TAMs that are CTLA-4-positive and CD86-positive, respectively, are greater within a 75 μm HRS cell niche relative to areas outside this region (CTLA-4, 38% vs 18% [P = .0001]; CD86, 38% vs 24% [P = .0007]). Importantly, CTLA-4-positive cells are present, and focally contact HRS cells, in recurrent cHL tumors following a variety of therapies, including PD-1 blockade. These results implicate CTLA-4:CD86 interactions as a component of the immunologically privileged niche surrounding HRS cells and raise the possibility that patients with cHL refractory to PD-1 blockade may benefit from CTLA-4 blockade.
Collapse
|
406
|
Peringeth G, Torka P, Wong J, Hernandez-Ilizaliturri FJ. Successful Treatment of Paraneoplastic Cholestasis in Relapsed/Refractory Hodgkin Lymphoma With Bridging Therapy and Checkpoint Blockade. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e316-e319. [PMID: 32179034 DOI: 10.1016/j.clml.2020.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/05/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Gopisree Peringeth
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Pallawi Torka
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Jerry Wong
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | |
Collapse
|
407
|
Song W, Zhang M. Use of CAR-T cell therapy, PD-1 blockade, and their combination for the treatment of hematological malignancies. Clin Immunol 2020; 214:108382. [PMID: 32169439 DOI: 10.1016/j.clim.2020.108382] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/05/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022]
Abstract
With the successful treatment of B-cell lymphomas using rituximab, a monoclonal antibody targeting CD20, novel immunotherapies have developed rapidly in recent years. Immune checkpoint blockade and chimeric antigen receptor-T (CAR-T) cell therapy, which are antibody-based therapy and cell-based therapy, respectively, show promising efficacy and have been approved by the Food and Drug Administration for treating hematological malignancies. However, considering severe side effects and short-term clinical remission, the combination of CAR-T cell therapy and programmed cell-death protein-1 (PD-1) blockade has been applied to enhance therapeutic efficacy in preclinical models and clinical trials. Herein, we review the mechanism of the two therapies, show their toxicities and clinical use respectively, address their combined application, and discuss the scope of further investigations of this mechanism-based combination therapy.
Collapse
Affiliation(s)
- Wenting Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou City, Henan Province, China; Academy of Medical Sciences of Zhengzhou University, 450052 Zhengzhou City, Henan Province, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou City, Henan Province, China.
| |
Collapse
|
408
|
Bentolila G, Pavlovsky A. Relapse or refractory Hodgkin lymphoma: determining risk of relapse or progression after autologous stem-cell transplantation. Leuk Lymphoma 2020; 61:1548-1554. [PMID: 32148142 DOI: 10.1080/10428194.2020.1732959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The treatment of classic Hodgkin lymphoma (HL) is a success in onco-hematology. Despite the high cure rate of HL with initial therapy, 5-10% of patients are primary refractory and 10-20% will eventually relapse. The standard treatment for these patients is salvage chemotherapy and autologous stem cell transplantation (ASCT). Only about half of these patients will benefit from this procedure. The prognosis of relapsed refractory (rr) HL has improved with the introduction of effective drugs. With these options available, identification of reliable risk factors is important to guide treatment over the course of disease. Different variables including performance status, anemia, B symptoms, laboratory abnormalities, treatment intensity before ASCT, response to therapy, and duration of remission, have been analyzed to determine risk for progression-free survival (PFS) and overall survival (OS) after ASCT. This review will discuss the publications analyzing these factors, the validated risk scores useful to identify patients at high risk of progression after ASCT, and will describe future perspectives.
Collapse
Affiliation(s)
| | - Astrid Pavlovsky
- FUNDALEU: Fundacion contra la Leucemia, Buenos Aires, Argentina.,Centro de Hematologia Pavlovsky, Buenos Aires, Argentina.,GATLA: Grupo Argentino de Tratamiento de Leucemia Aguda, Buenos Aires, Argentina
| |
Collapse
|
409
|
Hradska K, Kascak M, Hajek R, Jelinek T. Identifying and treating candidates for checkpoint inhibitor therapies in multiple myeloma and lymphoma. Expert Rev Hematol 2020; 13:375-392. [PMID: 32116068 DOI: 10.1080/17474086.2020.1733405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: One of the hallmarks of cancerogenesis is the ability of tumor cells to evade the immune system. They can achieve it by abusing inhibitory immune checkpoint pathways, which, under normal circumstances, maintain peripheral tolerance during infection. Immune checkpoint inhibitors, especially anti-PD-1/PD-L1 monoclonal antibodies, currently represent a widely discussed treatment option not only in solid oncology, but in hematology-oncology as well.Areas covered: The manuscript is focused on clinical research concerning PD-1/PD-L1 blockade in lymphoma and multiple myeloma in order to identify the patients who would profit the most from this treatment modality. The authors reviewed articles on the topic on PubMed and relevant clinical trials on clinicaltrials.gov before October 2019.Expert opinion: So far, nivolumab and pembrolizumab have been approved for treating patients with relapsed/refractory classical Hodgkin lymphoma and primary mediastinal B cell lymphoma. Nevertheless, monotherapy alone is not curative and a combinational approach is needed. Modern treatment strategies and combinations are comprehensively summarized in this manuscript. There is no approved immune checkpoint inhibitor for the multiple myeloma indication. Although the combination of PD-1/PD-L1 inhibitors with immunomodulatory agents initially seemed promising, unexpected immune related toxicities have stopped any further development. Novel strategies and more potent combinations in myeloma and lymphoma are further discussed in the manuscript.
Collapse
Affiliation(s)
- Katarina Hradska
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Michal Kascak
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Roman Hajek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Tomas Jelinek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.,Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.,Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
410
|
Otsuka Y, Nishikori M, Arima H, Izumi K, Kitawaki T, Hishizawa M, Takaori-Kondo A. EZH2 inhibitors restore epigenetically silenced CD58 expression in B-cell lymphomas. Mol Immunol 2020; 119:35-45. [DOI: 10.1016/j.molimm.2020.01.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/08/2020] [Accepted: 01/11/2020] [Indexed: 12/31/2022]
|
411
|
Kelly WJ, Gilbert MR. Glucocorticoids and immune checkpoint inhibitors in glioblastoma. J Neurooncol 2020; 151:13-20. [PMID: 32108294 DOI: 10.1007/s11060-020-03439-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/14/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Immunotherapy, activation of the immune system to target tumor cells, represents a paradigm shift in the treatment of cancer. Immune checkpoint therapies, which target immunomodulatory molecules expressed on T-lymphocytes, have demonstrated improved survival in a variety of malignancies. However, benefit in glioblastoma, the most common and devastating malignant brain tumor, remains to be seen. With several recent clinical trials failing to show efficacy of immunotherapy, concerns have been raised regarding the impact of glucocorticoid use in this patient population that may impair the ability for immune checkpoint inhibitors to affect a response. METHODS For this article we examined the mechanism by which immune checkpoint inhibitors activate, and glucocorticoids impair, T-lymphocyte function. RESULTS In this context, we review the clinical data of immune checkpoint inhibitors in glioblastoma as well as the impact glucocorticoids have on immune checkpoint inhibitor efficacy. Finally, we highlight key questions that remain in the field, and the potential benefit of further research for central nervous system tumors. CONCLUSION More information on the extent, character and duration of glucocorticoids on patients treated with PD-(L)1 will better inform both clinical management and novel therapeutic development of immunotherapy in patients with CNS malignancies.
Collapse
Affiliation(s)
- William J Kelly
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Building 82, Room 235, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Building 82, Room 235, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA.
| |
Collapse
|
412
|
Normalization Cancer Immunotherapy for Melanoma. J Invest Dermatol 2020; 140:1134-1142. [PMID: 32092349 DOI: 10.1016/j.jid.2020.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/26/2019] [Accepted: 02/05/2020] [Indexed: 12/28/2022]
Abstract
Today, we are witnessing a revolution in the treatment of cancer using immunotherapy. In the past decade, work from many laboratories and clinicians has unequivocally demonstrated that the immune system can eradicate established cancers and enhance patient survival. However, immunotherapies have distinct tumor response-to-toxicity profiles owing to distinct mechanisms of action. We have previously termed immunotherapies that activate a general systemic immune response as enhancement cancer immunotherapy and those that target a specific dysfunctional immune response, especially within the tumor microenvironment, as normalization cancer immunotherapy. In this perspective, we provide a framework for normalization cancer immunotherapy in the context of melanoma.
Collapse
|
413
|
Vassilakopoulos TP, Asimakopoulos JV, Konstantopoulos K, Angelopoulou MK. Optimizing outcomes in relapsed/refractory Hodgkin lymphoma: a review of current and forthcoming therapeutic strategies. Ther Adv Hematol 2020; 11:2040620720902911. [PMID: 32110285 PMCID: PMC7026824 DOI: 10.1177/2040620720902911] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 12/18/2019] [Indexed: 12/21/2022] Open
Abstract
The outcome of patients with relapsed/refractory classical Hodgkin lymphoma (rr-cHL) has improved considerably in recent years owing to the approval of highly active novel agents such as brentuximab vedotin and Programmed Death-1 (PD-1) inhibitors. Although no randomized trials have been conducted to provide formal proof, it is almost undisputable that the survival of these patients has been prolonged. As autologous stem-cell transplantation (SCT) remains the standard of care for second-line therapy of most patients with rr-cHL, optimization of second-line regimens with the use of brentuximab vedotin, or, in the future, checkpoint inhibitors, is promising to increase both the eligibility rate for transplant and the final outcome. The need for subsequent therapy, and especially allogeneic SCT, can be reduced with brentuximab vedotin consolidation for 1 year, while pembrolizumab is also being tested in this setting. Several other drug categories appear to be active in rr-cHL, but their development has been delayed by the appearance of brentuximab vedotin, nivolumab and pembrolizumab, which have dominated the field of rr-cHL treatment in the last 5 years. Combinations of active drugs in chemo-free approaches may further increase efficacy and hopefully reduce toxicity in rr-cHL, but are still under development.
Collapse
Affiliation(s)
- Theodoros P Vassilakopoulos
- Department of Haematology and Bone Marrow Transplantation, National and Kapodistrian University of Athens, School of Medicine, Laikon General Hospital, 17 Ag. Thoma Str., Goudi, Athens, 11527, Greece
| | - John V Asimakopoulos
- Department of Haematology and Bone Marrow Transplantation, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | - Kostas Konstantopoulos
- Department of Haematology and Bone Marrow Transplantation, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | - Maria K Angelopoulou
- Department of Haematology and Bone Marrow Transplantation, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| |
Collapse
|
414
|
Potential Survival Benefit for Patients Receiving Allogeneic Hematopoietic Stem Cell Transplantation after Nivolumab Therapy for Relapse/Refractory Hodgkin Lymphoma: Real-Life Experience in Spain. Biol Blood Marrow Transplant 2020; 26:1534-1542. [PMID: 32068094 DOI: 10.1016/j.bbmt.2020.02.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 02/03/2023]
Abstract
Clinical trials have shown that nivolumab has remarkable activity against relapsed/refractory classical Hodgkin lymphoma (cHL). However, the role of allogeneic hematopoietic stem cell transplantation (allo-HSCT) as consolidation therapy in these patients remains controversial. We performed a retrospective analysis of data from 74 patients treated with nivolumab. The overall response rate was 58% (including 30.6% with complete responses). Treatment-related adverse events were reported in 56.8% of patients (grade ≥3 in 9.4%). The main reasons for nivolumab discontinuation were referral for transplantation (41.7% patients) and disease progression (37.5%). The 2-year overall survival (OS) rate was 52% for the entire series. Ultimately, 39 patients underwent allo-HSCT. The cumulative incidence of grade II-IV acute graft-versus-host disease was 33.3% (grade III-IV in 2 patients). The cumulative incidence of nonrelapse mortality was 13.2%. Among the patients who responded to nivolumab, the 2-year OS and progression-free survival (PFS) were higher in patients who underwent consolidation with allo-HSCT (77.5% versus 42.6% [P = .126] and 73.9% versus 27.2% [P = .025], respectively). Thus, the efficacy and safety of nivolumab were comparable to values reported in previous clinical trials. The percentage of patients who bridged to transplantation was high, indicating a preference for Spanish physicians. These results suggest that consolidation allo-HSCT increases OS and PFS.
Collapse
|
415
|
Zhang J, Fan L, Li JY. [Research progress of treatment in Hodgkin lymphoma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 40:969-972. [PMID: 31856452 PMCID: PMC7342374 DOI: 10.3760/cma.j.issn.0253-2727.2019.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- J Zhang
- Department of Hematology, the First Affiliated Hospital of NanJing Medical University, JiangSu Province Hospital, Nanjing 210029, China
| | | | | |
Collapse
|
416
|
Ozturk V, Yikilmaz AS, Kilicarslan A, Bakanay SM, Akinci S, Dilek İ. The Triple Positivity for EBV, PD-1, and PD-L1 Identifies a Very High Risk Classical Hodgkin Lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e375-e381. [PMID: 32295735 DOI: 10.1016/j.clml.2019.11.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/10/2019] [Accepted: 11/28/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND The programmed death receptor (PD-1) and ligand (PD-L1) pathway act by suppressing the antitumor response in chronic Hodgkin lymphoma (cHL). In this study, we aimed to investigate the effect of PD-1, PD-L1, and Epstein-Barr virus (EBV) positivity on prognosis at the initial diagnosis of cHL. MATERIAL AND METHODS Thirty-six patients with cHL were retrospectively analyzed. PD-L1 staining was performed for RS cells and tumor microenvironment in the biopsy materials of cases. The presence of EBV was investigated by EBER (EBV-encoded RNA) method in tumor cell. P < .05 was accepted as significant. RESULTS The presence of advanced-stage disease, B symptoms, intermediate or high-risk international prognostic index (IPS), and extranodal involvement were found to be related to both PD-L1 positivity and EBV positivity in RS cells. PD-L1 positivity in RS cells was also associated with EBV positivity. There were 6 (16.7%) triple-positive (EBV+, RS-PD-L1+, mic-PD-1+) patients. All of these patients had advanced-stage disease, B symptoms at the time of diagnosis, and intermediate-high IPS score, and 4 of 6 patients had extranodal involvement. This group also had significantly shortened overall survival compared with others (38.4 months vs. 67.9 months P = .024). CONCLUSION Our data suggest that there is correlation between PD-L1 positivity and EBV positivity in tumor RS cells that are also associated with extranodal involvement, intermediate and high IPS score, presence of B symptoms, and advanced-stage disease. In addition, we identified a group of triple-positive (EBV+, RS-PD-L1+, mic-PD-1+) cHL patients who have a very high-risk disease.
Collapse
Affiliation(s)
- Vedia Ozturk
- Department of Pathology, Yıldırım Beyazıt University, Ankara, Turkey
| | | | - Aydan Kilicarslan
- Department of Pathology, Yıldırım Beyazıt University, Ankara, Turkey
| | - Sule Mine Bakanay
- Department of Hematology, Yıldırım Beyazıt University, Ankara, Turkey
| | - Sema Akinci
- Department of Hematology, Yıldırım Beyazıt University, Ankara, Turkey
| | - İmdat Dilek
- Department of Hematology, Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
417
|
Abstract
PURPOSE OF REVIEW The landscape of relapsed or refractory (R/R) Hodgkin lymphoma (HL) treatment has changed significantly since the FDA approval of brentuximab vedotin in 2011. In this review, we summarize the recent advances in the therapy for R/R classical Hodgkin lymphoma (cHL). RECENT FINDINGS Immunotherapies with pembrolizumab, nivolumab, and ipilimumab, and chimeric antigen receptor (CAR) T cell therapies have shown promising results in early phase trials. Other novel agents under investigation include targeted therapies with histone deacetylase inhibitors, Janus kinase 2 inhibitors, and immunomodulators. While further studies with larger populations and longer follow-up times are needed to determine the safe and effective combinations, these novel approaches represent a growing list of treatment options that are on the horizon to improve the cure rate and increase duration of remission for R/R HL patients.
Collapse
Affiliation(s)
- Yun Choi
- NYU Langone Hematology & Medical Oncology Fellowship Program, 550 First Avenue, New York, NY, 10016, USA
| | - Catherine S Diefenbach
- Division of Hematology and Medical Oncology, Perlmutter Cancer Center at NYU Langone Health, New York University School of Medicine, Langone Medical Center, 240 East 38th Street, 19th Floor, New York, NY, 10016, USA.
| |
Collapse
|
418
|
Xie Y, Wang X, Leng X, Zheng W, Ping L, Zhang C, Liu W, Deng L, Wu M, Song Y, Zhu J. High-dose chemotherapy followed by autologous stem cell transplantation for patients with refractory/relapsed classical Hodgkin lymphoma: a single center experience from China. Ann Hematol 2020; 99:549-555. [PMID: 31980860 DOI: 10.1007/s00277-019-03812-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/26/2019] [Indexed: 01/20/2023]
Abstract
To evaluate the outcomes of refractory/relapsed cHL patients after high-dose chemotherapy (HDCT) and autologous stem cell transplantation (ASCT) in Beijing Cancer hospital and to identify the prognostic risk factors. We retrospectively analyzed 115 relapsed/refractory cHL patients who accepted HDCT and ASCT in our cancer center and had complete follow-up data from April 2000 to May 2017. Ages of these 115 patients at ASCT ranged from 14 to 63 (median age 28). Forty-four (38.3%) patients achieved CR and 50 (43.5%) patients achieved PR before ASCT. Thirty-seven (48.7%) patients of those 76 patients who did PET-CT before ASCT had negative PET-CT scans. The median follow-up time was 72 months. A total of 23 patients died in our study. The 5-year OS and PFS rates of all patients after ASCT were 78.7% and 53%, respectively. The 5-year OS rates after ASCT of patients who were in CR or PR or less than PR status before ASCT were 92.8%, 68.2%, and 76.2%, respectively (log-rank = 2.913, p = 0.233). And their 5-year PFS rates after ASCT were 69.2%, 54.2%, and 18.5%, respectively (log-rank = 13.615, p = 0.001). Univariate analysis revealed that ECOG (p = 0.010; hazard ratio = 1.578), disease status before ASCT (CR: p = 0.001; hazard ratio = 0.227) and after ASCT (CR: p < 0.001; hazard ratio = 0.154), and PET-CT results after ASCT (p = 0.023; hazard ratio = 0.438) significantly impact patients' PFS while number of pretransplant salvage chemotherapy (p = 0.037; hazard ratio = 2.521), radiotherapy (p = 0.046; hazard ratio = 0.423), and disease status after ASCT (CR: p = 0.010; hazard ratio = 0.197) significantly affected patients' OS. Multivariate analysis shown only disease status before ASCT (p = 0.002) had significant impact on PFS and disease status after ASCT (p = 0.021) had significant impact on OS. R/R HL patients can still obtain long-term PFS after HDCT and ASCT and disease status before ASCT was the most significant prognostic factor for PFS.
Collapse
Affiliation(s)
- Yan Xie
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaopei Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin Leng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wen Zheng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lingyan Ping
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chen Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Weiping Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lijuan Deng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yuqin Song
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ju Zhu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
419
|
Anti-CD30 chimeric antigen receptor T cell therapy for relapsed/refractory CD30 + lymphoma patients. Blood Cancer J 2020; 10:8. [PMID: 31974371 PMCID: PMC6978321 DOI: 10.1038/s41408-020-0274-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/20/2019] [Accepted: 01/10/2020] [Indexed: 11/26/2022] Open
|
420
|
|
421
|
Cost-effectiveness of first-line treatment options for patients with advanced-stage Hodgkin lymphoma: a modelling study. LANCET HAEMATOLOGY 2020; 7:e146-e156. [PMID: 31948928 DOI: 10.1016/s2352-3026(19)30218-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/28/2019] [Accepted: 09/11/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Several strategies are available for the initial treatment of advanced-stage Hodgkin lymphoma, but the optimal strategy in terms of cost-effectiveness is unclear. The aim of this study was to compare the quality-adjusted effectiveness and costs of five modern treatment options for transplantation-eligible patients with newly diagnosed advanced-stage Hodgkin lymphoma. METHODS A Markov decision-analytic model was developed using a 20-year time horizon. Five of the most common treatment approaches were selected based on clinical experience and expert opinion: (1) six cycles of doxorubicin, bleomycin, vinblastine, dacarbazine (ABVD), including data from the HD2000 trial, Viviani and colleagues, and EORTC trial; (2) six cycles of bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone (BEACOPP; from the HD15 trial or PET-adapted as in the HD18 trial, two initial cycles of BEACOPP followed by four additional cycles for patients with a positive PET and either two or four additional cycles of BEACOPP for patients with a negative PET); (3) PET-adapted escalation (as in the RATHL trial, two cycles of standard ABVD chemotherapy followed by an additional four cycles of ABVD or AVD in PET-negative patients and four cycles of BEACOPP in PET-positive patients); (4) six cycles of brentuximab vedotin, doxorubicin, vinblastine, dacarbazine (A-AVD) or ABVD as in the Echelon-1 trial; and (5) PET-adapted de-escalation (as in the AHL2011 trial, two cycles of BEACOPP followed by PET2 scan; PET-positive patients received two additional BEACOPP cycles and PET-negative patients received two cycles of ABVD; at PET4, PET-negative patients completed two further cycles of either ABVD or BEACOPP depending on what they received after PET2, and PET-positive patients received salvage therapy). Note that all uses of BEACOPP in these strategies were BEACOPPescalated. The randomised groups of interest from these studies comprised 4255 patients enrolled between April, 2000, and January, 2016. Baseline probability estimates and utilities were derived from the included trials in addition to a systematic review of published studies. A Canadian public health payer's perspective was considered (CAN$1=US$0·74) and adjusted for inflation for 2018. All costs and benefits were discounted by 1·5% per year because life-years now are more valuable than future potential life-years. FINDINGS Probabilistic analyses (10 000 simulations) showed that, for a willingness-to-pay threshold of CAN$50 000, a PET-adapted de-escalation strategy based on AHL2011 was more cost-effective 87% of the time. This strategy had the highest number of life-years (14·6 years [95% CI 13·7-15·1]) and quality-adjusted life years (13·2 years [95% CI 10·2-14·4]), and the lowest direct costs ($53 129 [95% CI 31 914-94 446]) compared with the other treatment regimens. Sensitivity analyses showed that the model was robust to key variables, including probability of treatment-related mortality, relapse, frequency of secondary malignancy, death from secondary malignancy, and probability of infertility after BEACOPP. INTERPRETATION Our results suggest that, when considering cost, effectiveness, and short and long-term toxicities, the preferred treatment strategy for patients with newly diagnosed advanced-stage Hodgkin lymphoma is the PET-adapted de-escalation regimen starting with BEACOPP and de-escalating to ABVD as appropriate. Although our findings do not provide an absolute best treatment approach for clinicians to follow for all patients, they can contribute to shared decision making between patients and treating physicians. FUNDING None.
Collapse
|
422
|
Khurana A, Armand P, Ansell SM. Checkpoint inhibition therapy as possible frontline therapy for Hodgkin lymphoma. Leuk Lymphoma 2020; 61:1063-1074. [PMID: 31914840 DOI: 10.1080/10428194.2019.1709832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Classic Hodgkin lymphoma (cHL) is a unique lymphoid malignancy with an immunosuppressed tumor microenvironment. Increased PD-L1 expression on the malignant Hodgkin Reed Sternberg (HRS) cells due to genetic amplification at chromosome 9p24.1 is likely one of the primary mechanisms for this unique biology. For this reason, immune checkpoint inhibitors targeting PD-1/PD-L1 interaction have proven to be uniquely successful in relapsed/refractory cHL. While the response rates are in the 70-80% range and are often durable, most patients still relapse. Combination strategies with conventional chemotherapy, novel drugs such as antibody-drug conjugates (ADCs), and other immune therapies are ongoing. Many unanswered questions about checkpoint inhibitors remain, such as defining the best modality for evaluation of response, confirming a strategy of modifying therapy based on the response, validating response endpoints specific to immune therapies and, identifying predictive biomarkers for response. As we evaluate the use of checkpoint inhibitors in the frontline setting for cHL, we need a critical approach to evaluate the benefits and challenges that ensue.
Collapse
Affiliation(s)
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | |
Collapse
|
423
|
Beck A, Dumontet C, Joubert N. [Antibody-drug conjugates in oncology. New strategies in development]. Med Sci (Paris) 2020; 35:1043-1053. [PMID: 31903916 DOI: 10.1051/medsci/2019228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
An Antibody-Drug Conjugate (armed antibody) is a vectorized chemotherapy that results from the grafting of a cytotoxic agent on a monoclonal antibody via a judiciously designed spacer arm. ADCs have made considerable progress in 10 years. In 2009, only gemtuzumab ozogamicin (Mylotarg®) was used clinically. In 2019, 4 other ADCs have been approved and more than 80 others are in active clinical trials. The second part of this review will focus on new emerging strategies to address ADCs drawbacks and attempt to broaden their therapeutic window. Finally, combinations with conventional chemotherapy or checkpoint inhibitors will be discussed, in the pursuit to make Antibody-Drug Conjugates the embodiment of Paul Ehrlich's dream of the magic bullet.
Collapse
Affiliation(s)
- Alain Beck
- Institut de Recherche Pierre Fabre, Centre d'Immunologie Pierre Fabre, 5 Avenue Napoléon III, 74160 Saint Julien-en-Genevois, France
| | - Charles Dumontet
- Cancer Research Center of Lyon (CRCL), Inserm 1052/CNRS, 69000 Lyon, France - Université de Lyon, 69000 Lyon, France - Hospices Civils de Lyon, 69000 Lyon, France
| | - Nicolas Joubert
- GICC EA7501, Université de Tours, équipe IMT, 31 avenue Monge, 37200 Tours, France
| |
Collapse
|
424
|
Outcome after autologous stem cell transplantation in primary refractory or relapsed Hodgkin lymphoma-a long-term follow-up single center experience. Ann Hematol 2020; 99:265-276. [PMID: 31897675 DOI: 10.1007/s00277-019-03900-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Autologous stem cell transplantation (autoSCT) can achieve long-term remission in primary refractory or relapsed Hodgkin lymphoma (r/r HL); however, still up to 50% of patients relapse after autoSCT. In this retrospective analysis, we investigated the impact of autologous stem cell transplantation in a consecutive, unselected cohort of primary refractory and relapsed Hodgkin lymphoma patients (n = 66) with the majority of patients treated in the pre-brentuximab vedotin and immune checkpoint inhibitor era. In our cohort, a 5-year overall survival (OS) from autoSCT of 59.5% and a 5-year progression-free survival (PFS) after autoSCT of 46.1% was achieved. Multivariate analysis revealed primary refractory disease and early relapse (< 12 months) after initial therapy as well as the presence of B symptoms at relapse as independent risk factors associated with a higher risk for relapse and an inferior PFS and OS. Several other clinical factors, including the presence of extranodal disease at relapse and failure to achieve a complete response to salvage chemotherapy, were associated with a trend towards an inferior survival. Patients relapsing after autoSCT had a particularly poor outcome, regardless of eligibility to undergo allogeneic stem cell transplantation (alloSCT). We further evaluated recently published prognostic models for r/r HL patients undergoing autoSCT and could validate several risk scores in our independent "real world" cohort.
Collapse
|
425
|
Hodgkin lymphoma: a review of pathological features and recent advances in pathogenesis. Pathology 2020; 52:154-165. [DOI: 10.1016/j.pathol.2019.09.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 02/08/2023]
|
426
|
Lopci E, Meignan M. Current Evidence on PET Response Assessment to Immunotherapy in Lymphomas. PET Clin 2020; 15:23-34. [DOI: 10.1016/j.cpet.2019.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
427
|
Kiesewetter B, Cherny NI, Boissel N, Cerisoli F, Dafni U, de Vries EGE, Ghia P, Gökbuget N, González-Calle V, Huntly B, Jäger U, Latino NJ, Douillard JY, Malcovati L, Mateos MV, Ossenkoppele GJ, Porkka K, Raderer M, Ribera JM, Scarfò L, Wester R, Zygoura P, Sonneveld P. EHA evaluation of the ESMO-Magnitude of Clinical Benefit Scale version 1.1 (ESMO-MCBS v1.1) for haematological malignancies. ESMO Open 2020; 5:e000611. [PMID: 31958292 PMCID: PMC7003483 DOI: 10.1136/esmoopen-2019-000611] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Value frameworks in oncology have not been validated for the assessment of treatments in haematological malignancies, but to avoid overlaps and duplications it appears reasonable to build up experience on existing value frameworks, such as the European Society for Medical Oncology-Magnitude of Clinical Benefit Scale (ESMO-MCBS). METHODS Here we present the results of the first feasibility testing of the ESMO-MCBS v1.1 for haematological malignancies based on the grading of 80 contemporary studies for acute leukaemia, chronic leukaemia, lymphoma, myeloma and myelodysplastic syndromes. The aims were (1) to evaluate the scorability of data, (2) to evaluate the reasonableness of the generated grades for clinical benefit using the current version and (3) to identify shortcomings in the ESMO-MCBS v1.1 that require amendments to improve the efficacy and validity of the scale in grading new treatments in the management of haematological malignancies. RESULTS In general, the ESMO-MCBS v1.1 was found to be widely applicable to studies in haematological malignancies, generating scores that were judged as reasonable by European Hematology Association (EHA) experts. A small number of studies could either not be graded or were not appropriately graded. The reasons, related to the differences between haematological and solid tumour malignancies, are identified and described. CONCLUSIONS Based on the findings of this study, ESMO and EHA are committed to develop a version of the ESMO-MCBS that is validated for haematological malignancies. This development process will incorporate all of the usual stringencies for accountability of reasonableness that have characterised the development of the ESMO-MCBS including field testing, statistical modelling, evaluation for reasonableness and openness to appeal and revision. Applying such a scale will support future public policy decision-making regarding the value of new treatments for haematological malignancies and will provide insights that could be helpful in the design of future clinical trials.
Collapse
Affiliation(s)
- Barbara Kiesewetter
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Nathan I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Nicolas Boissel
- Department of Hematology, Hospital Saint-Louis, Paris, Île-de-France, France
- Adolescent and Young Adult Hematology Unit, Diderot University Paris Faculty of Medicine, Paris, Île-de-France, France
| | - Francesco Cerisoli
- European Hematology Association, Den Haag, Zuid-Holland, The Netherlands
| | - Urania Dafni
- Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
- Frontier Science Foundation-Hellas, Frontier Science Foundation-Hellas, Athens, Greece
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Paolo Ghia
- Strategic Research Program on CLL, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milano, Lombardia, Italy
- Universita Vita e Salute San Raffaele, Milano, Lombardia, Italy
| | - Nicola Gökbuget
- Department of Hematology/Oncology, Goethe University, Frankfurt am Main, Hessen, Germany
| | - Verónica González-Calle
- Department of Hematology and Instituto de Investigación Biomédica de Salamanca-IBSAL, University Hospital of Salamanca, Salamanca, Castilla y León, Spain
| | - Brian Huntly
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Ulrich Jäger
- Department of Medicine I, Clinical Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | | | | | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia, Pavia, Lombardia, Italy
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Lombardia, Italy
| | - María-Victoria Mateos
- Department of Hematology and Instituto de Investigación Biomédica de Salamanca-IBSAL, University Hospital of Salamanca, Salamanca, Castilla y León, Spain
| | - Gert J Ossenkoppele
- Department of Hematology, VU University Medical Centre Amsterdam, Amsterdam, Noord-Holland, The Netherlands
| | - Kimmo Porkka
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Markus Raderer
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Josep-Maria Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Barcelona, Catalunya, Spain
| | - Lydia Scarfò
- Strategic Research Program on CLL, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milano, Lombardia, Italy
- Universita Vita e Salute San Raffaele, Milano, Lombardia, Italy
| | - Ruth Wester
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Zuid-Holland, The Netherlands
| | - Panagiota Zygoura
- Frontier Science Foundation-Hellas, Frontier Science Foundation-Hellas, Athens, Greece
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Zuid-Holland, The Netherlands
- Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, Zuid-Holland, The Netherlands
| |
Collapse
|
428
|
Bisso A, Sabò A, Amati B. MYC in Germinal Center-derived lymphomas: Mechanisms and therapeutic opportunities. Immunol Rev 2019; 288:178-197. [PMID: 30874346 DOI: 10.1111/imr.12734] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/11/2018] [Indexed: 12/13/2022]
Abstract
The rearrangement of immunoglobulin loci during the germinal center reaction is associated with an increased risk of chromosomal translocations that activate oncogenes such as MYC, BCL2 or BCL6, thus contributing to the development of B-cell lymphomas. MYC and BCL2 activation are initiating events in Burkitt's (BL) and Follicular Lymphoma (FL), respectively, but can occur at later stages in other subtypes such as Diffuse Large-B Cell Lymphoma (DLBCL). MYC can also be activated during the progression of FL to the transformed stage. Thus, either DLBCL or FL can give rise to aggressive double-hit lymphomas (DHL) with concurrent activation of MYC and BCL2. Research over the last three decades has improved our understanding of the functions of these oncogenes and the basis for their cooperative action in lymphomagenesis. MYC, in particular, is a transcription factor that contributes to cell activation, growth and proliferation, while concomitantly sensitizing cells to apoptosis, the latter being blocked by BCL2. Here, we review our current knowledge about the role of MYC in germinal center B-cells and lymphomas, discuss MYC-induced dependencies that can sensitize cancer cells to select pharmacological inhibitors, and illustrate their therapeutic potential in aggressive lymphomas-and in particular in DHL, in combination with BCL2 inhibitors.
Collapse
Affiliation(s)
- Andrea Bisso
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Arianna Sabò
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Bruno Amati
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
429
|
Rigacci L, Puccini B, Broccoli A, Dona M, Gotti M, Evangelista A, Santoro A, Bonfichi M, Re A, Spina M, Botto B, Pulsoni A, Pagani C, Stelitano C, Salvi F, Nassi L, Mannelli L, Kovalchuk S, Gioia D, Zinzani PL. Clinical characteristics of interim-PET negative patients with a positive end PET from the prospective HD08-01 FIL study. Ann Hematol 2019; 99:283-291. [PMID: 31872361 DOI: 10.1007/s00277-019-03889-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 12/06/2019] [Indexed: 11/28/2022]
Abstract
FDG-positron emission tomography (PET) performed early during therapy in advanced Hodgkin lymphoma patients has been confirmed as being important for progression-free survival. A group of patients with a negative interim-PET (i-PET) showed a positive end induction PET (e-PET). The aim of this study was to evaluate the clinical characteristics of patients with a positive e-PET as a secondary end point of the HD0801 study. A total of 519 patients with advanced-stage de novo Hodgkin lymphoma received initial treatment and underwent an i-PET. Patients with negative results continued the standard treatment. i-PET negative patients were then evaluated for response with an e-PET and those patients found to have a positive one were also then given a salvage therapy. Among 409 i-PET negative, 16 interrupted the therapy, 393 patients were evaluated with an e-PET, and 39 were positive. Sixteen out of 39 underwent a diagnostic biopsy and 15 were confirmed as HD. Seventeen out of 39 e-PET were reviewed according to the Deauville Score and, in sixteen, it was confirmed positive (10 DS 5, 6 DS 4). With the exception of high LDH value at diagnosis (p = 0.01; HR 95% CI 1.18-4.89), no clinical characteristics were significantly different in comparison with e-PET negative patients. Positive e-PET after a negative i-PET has a worse outcome when compared with i-PET positive patients salvaged with therapy intensification. It was not possible to identify clinical characteristics associated with a positive e-PET.
Collapse
Affiliation(s)
- Luigi Rigacci
- Department of Hematology, AOU Careggi, Florence, Italy. .,Hematology Unit and Bone Marrow Transplant Unit, AO San Camillo Forlanini, Rome, Italy.
| | | | - Alessandro Broccoli
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Manjola Dona
- Department of Nuclear Medicine, Ospedale Santo Stefano, Prato, Italy
| | - Manuel Gotti
- Policlinico San Matteo Pavia Fondazione, IRCCS, Pavia, Italy
| | - Andrea Evangelista
- Unit of Cancer Epidemiology, AO Città della Salute e della Scienza di Torino and FIL Secretary, Turin, Italy
| | | | | | - Alessandro Re
- Department of Hematology, Spedali Civili, Brescia, Italy
| | | | - Barbara Botto
- Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | | | - Chiara Pagani
- Department of Hematology, Spedali Civili, Brescia, Italy
| | | | - Flavia Salvi
- Division of Hematology, A.O. SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Luca Nassi
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Lara Mannelli
- Department of Hematology, AOU Careggi, Florence, Italy
| | | | - Daniela Gioia
- Unit of Cancer Epidemiology, AO Città della Salute e della Scienza di Torino and FIL Secretary, Turin, Italy
| | - Pier Luigi Zinzani
- Institute of Hematology "L. e A. Seràgnoli", University of Bologna, Bologna, Italy
| |
Collapse
|
430
|
Aoki T, Chong LC, Takata K, Milne K, Hav M, Colombo A, Chavez EA, Nissen M, Wang X, Miyata-Takata T, Lam V, Viganò E, Woolcock BW, Telenius A, Li MY, Healy S, Ghesquiere C, Kos D, Goodyear T, Veldman J, Zhang AW, Kim J, Saberi S, Ding J, Farinha P, Weng AP, Savage KJ, Scott DW, Krystal G, Nelson BH, Mottok A, Merchant A, Shah SP, Steidl C. Single-Cell Transcriptome Analysis Reveals Disease-Defining T-cell Subsets in the Tumor Microenvironment of Classic Hodgkin Lymphoma. Cancer Discov 2019; 10:406-421. [PMID: 31857391 DOI: 10.1158/2159-8290.cd-19-0680] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/13/2019] [Accepted: 12/13/2019] [Indexed: 12/22/2022]
Abstract
Hodgkin lymphoma is characterized by an extensively dominant tumor microenvironment (TME) composed of different types of noncancerous immune cells with rare malignant cells. Characterization of the cellular components and their spatial relationship is crucial to understanding cross-talk and therapeutic targeting in the TME. We performed single-cell RNA sequencing of more than 127,000 cells from 22 Hodgkin lymphoma tissue specimens and 5 reactive lymph nodes, profiling for the first time the phenotype of the Hodgkin lymphoma-specific immune microenvironment at single-cell resolution. Single-cell expression profiling identified a novel Hodgkin lymphoma-associated subset of T cells with prominent expression of the inhibitory receptor LAG3, and functional analyses established this LAG3+ T-cell population as a mediator of immunosuppression. Multiplexed spatial assessment of immune cells in the microenvironment also revealed increased LAG3+ T cells in the direct vicinity of MHC class II-deficient tumor cells. Our findings provide novel insights into TME biology and suggest new approaches to immune-checkpoint targeting in Hodgkin lymphoma. SIGNIFICANCE: We provide detailed functional and spatial characteristics of immune cells in classic Hodgkin lymphoma at single-cell resolution. Specifically, we identified a regulatory T-cell-like immunosuppressive subset of LAG3+ T cells contributing to the immune-escape phenotype. Our insights aid in the development of novel biomarkers and combination treatment strategies targeting immune checkpoints.See related commentary by Fisher and Oh, p. 342.This article is highlighted in the In This Issue feature, p. 327.
Collapse
Affiliation(s)
- Tomohiro Aoki
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lauren C Chong
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Katsuyoshi Takata
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Katy Milne
- Deeley Research Centre, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Monirath Hav
- Cedars-Sinai Medical Center, Los Angeles, California
| | | | - Elizabeth A Chavez
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Michael Nissen
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Xuehai Wang
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Tomoko Miyata-Takata
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Vivian Lam
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Elena Viganò
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce W Woolcock
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Adèle Telenius
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Michael Y Li
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shannon Healy
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Chanel Ghesquiere
- Deeley Research Centre, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Daniel Kos
- Deeley Research Centre, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Talia Goodyear
- Deeley Research Centre, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Johanna Veldman
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Allen W Zhang
- Department of Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jubin Kim
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Saeed Saberi
- Department of Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Jiarui Ding
- Department of Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Pedro Farinha
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Andrew P Weng
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Kerry J Savage
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - David W Scott
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Gerald Krystal
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Brad H Nelson
- Deeley Research Centre, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anja Mottok
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada.,Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Akil Merchant
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Sohrab P Shah
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christian Steidl
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada. .,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
431
|
Safety and Efficacy in Relapsed or Refractory Classic Hodgkin's Lymphoma Treated with PD-1 Inhibitors: A Meta-Analysis of 9 Prospective Clinical Trials. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9283860. [PMID: 31950058 PMCID: PMC6948280 DOI: 10.1155/2019/9283860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/01/2019] [Accepted: 11/14/2019] [Indexed: 01/03/2023]
Abstract
Background Classic Hodgkin's lymphoma (cHL) is characterized by the unique biology in which rare Hodgkin-Reed-Sternberg cells propagate an immunosuppressive microenvironment. Checkpoint inhibitors that target the interaction of PD-1 immune checkpoint receptors have demonstrated remarkable activities in various cancers, such as cHL. This study aims to evaluate the safety and efficacy of PD-1 inhibitors in treating relapsed or refractory cHL (rrHL). Methods We searched PubMed, MEDLINE, Embase, Cochrane Central Register of Controlled Trials, China National Knowledge Infrastructure, Wanfang, Chinese Biological Medical Literature, and Abstracts of Conference proceedings of annual meetings without any language restrictions to limit language bias (up to January 2019) for prospective clinical trials that evaluate PD-1 inhibitors in treating relapsed or refractory cHL. Results A total of 9 prospective clinical trials with 731 patients were included in the meta-analysis. The pooled risks of all-grade and grade ≥3 adverse events (AEs) were 0.86 (95% CI: 0.66–0.98) and 0.21 (95% CI: 0.17–0.24), respectively. The pooled response, complete response, partial response, and stable disease rates were 0.74 (95% CI: 0.70–0.79), 0.24 (95% CI: 0.18–0.34), 0.48 (95% CI: 0.41–0.55), and 0.15 (95% CI: 0.12–0.17), respectively. The pooled 6-month progression-free survival and 1-year overall survival rates were 0.76 (95% CI: 0.72–0.79) and 0.93 (95% CI: 0.90–0.96), correspondingly. Conclusions Our meta-analysis suggested that anti-PD1 monoclonal antibodies improve the outcomes of response and survival rates with tolerable AEs in cHL. However, evidence of immune checkpoint inhibitors for patients with cHL remained insufficient. Well-designed randomized controlled trials or at least nonrandomized trials with a control group should be conducted to confirm the findings of this meta-analysis.
Collapse
|
432
|
Abstract
Immunotherapy is distinct from traditional chemotherapy in that it acts on immune cells rather than cancer cells themselves. Monoclonal antibodies targeting immune checkpoints on T cells - CTLA-4 and PD-1 - and PD-L1 on the cells of immune microenvironment are now approved for clinical use in several solid tumors and hematological malignancies. This article provides a general overview of the use of checkpoint inhibitors in hematologic malignancies with a special focus in acute myeloid leukemia.
Collapse
Affiliation(s)
- Arnab Ghosh
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Pere Barba
- Hematology Department, Vall d'Hebron University Hospital-Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
433
|
Huntington SF. Cure at what (systemic) financial cost? Integrating novel therapies into first-line Hodgkin lymphoma treatment. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:252-259. [PMID: 31808838 PMCID: PMC6913455 DOI: 10.1182/hematology.2019000030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Classic Hodgkin lymphoma (cHL) stands out as success story in the field of medical oncology, with multiagent chemotherapy with or without radiation leading to durable remission for most patients. Large-scale clinical trials during the past 40 years have sought to minimize toxicities while maintaining strong efficacy, including efforts to reduce the size of radiation fields, minimize alkylator chemotherapy, reduce the number of chemotherapy cycles, and omit radiation in select populations. The last decade has also ushered in novel therapies, including brentuximab vedotin (BV), that have improved clinical outcomes for patients with cHL resistant to standard cytotoxic therapies. More recently, a large randomized trial compared BV plus chemotherapy with chemotherapy alone for first-line treatment of advanced stage cHL. With ∼24 months of available follow-up, the BV containing regimen was found to be associated with a reduction in the risk of progression, death, or incomplete response to first-line treatment (modified progression-free survival). Whether this early signal of improved efficacy is worth the additional acute toxicities and added drug-related expenses associated with incorporating BV into first-line treatment remains controversial. This chapter provides historical background; reviews the cost-effectiveness of available cHL therapies; and summarizes potential ways to balance innovation, affordability, and patient access to novel therapeutics.
Collapse
Affiliation(s)
- Scott F. Huntington
- Department of Internal Medicine, Section of Hematology, Yale University, New Haven, CT
| |
Collapse
|
434
|
Allen PB, LaCasce AS. Evidence-Based Minireview: What is the optimal timing of anti-PD-1 antibodies in relapsed classical Hodgkin lymphoma? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:249-251. [PMID: 31808863 PMCID: PMC6913434 DOI: 10.1182/hematology.2019000077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A 26-year-old woman was initially diagnosed with stage III classical Hodgkin lymphoma (HL) treated with doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) for 6 cycles and relapsed 9 months after completing therapy. She was treated with salvage chemotherapy followed by an autologous transplantation and 1 year of brentuximab vedotin (BV) maintenance therapy. She now presents 1 year later with relapsed disease above and below the diaphragm. What treatment would you recommend for this patient?
Collapse
Affiliation(s)
- Pamela Blair Allen
- Department of Hematology and Oncology, Winship Cancer Institute of Emory University, Atlanta, GA; and
| | - Ann S LaCasce
- Department of Hematology and Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
435
|
Merryman RW, LaCasce A. Novel agents and immune invasion in Hodgkin lymphoma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:243-248. [PMID: 31808827 PMCID: PMC6913426 DOI: 10.1182/hematology.2019000029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The approval of brentuximab vedotin (BV) and the PD-1 inhibitors nivolumab and pembrolizumab has dramatically improved outcomes for patients with relapsed or refractory (R/R) classic Hodgkin lymphoma (HL). With the goal of increasing long-term disease control rates and decreasing late toxicities, these agents are currently being tested in earlier phases of treatment in combination with chemotherapy agents. In the R/R setting, our expanding understanding of HL's various mechanisms of immune evasion and treatment resistance has spurred a growing number of rationally designed combination trials. Beyond BV and PD-1 blockade, other novel therapies have demonstrated encouraging preliminary results, including targeted agents, like the CD25 antibody-drug conjugate ADCT-301, and cellular therapies, including CD30 chimeric antigen receptor T cells and Epstein-Barr virus (EBV)-directed cytotoxic T cells. These trials, coupled with the rapid development of prognostic and predictive biomarkers, should drive additional breakthroughs that promise safer and more effective therapies for patients with HL in the future.
Collapse
Affiliation(s)
- Reid W Merryman
- Department of Hematologic Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ann LaCasce
- Department of Hematologic Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
436
|
Özdemir BC, Bohanes P, Bisig B, Missiaglia E, Tsantoulis P, Coukos G, Montemurro M, Homicsko K, Michielin O. Deep Response to Anti-PD-1 Therapy of Metastatic Neurofibromatosis Type 1-Associated Malignant Peripheral Nerve Sheath Tumor With CD274/PD-L1 Amplification. JCO Precis Oncol 2019; 3:1-6. [DOI: 10.1200/po.18.00375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Berna C. Özdemir
- Lausanne University Hospital, Lausanne, Switzerland
- International Cancer Prevention Institute, Lausanne, Switzerland
| | | | | | | | | | - George Coukos
- Lausanne University Hospital, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Krisztian Homicsko
- Lausanne University Hospital, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Olivier Michielin
- Lausanne University Hospital, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
437
|
Lu J, Li L, Lan Y, Liang Y, Meng H. Immune checkpoint inhibitor-associated pituitary-adrenal dysfunction: A systematic review and meta-analysis. Cancer Med 2019; 8:7503-7515. [PMID: 31679184 PMCID: PMC6912062 DOI: 10.1002/cam4.2661] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/25/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
With the growing use of immune checkpoint inhibitors (ICIs), case reports of rare yet life-threatening pituitary-adrenal dysfunctions, particularly for hypopituitarism, are increasingly being published. In this analysis, we focus on these events by including the most recent publications and reports from early phase I/II and phase III clinical trials and comparing the incidence and risks across different ICI regimens. PubMed, Embase, and the Cochrane Library were systematically searched from inception to April 2019 for clinical trials that reported on pituitary-adrenal dysfunction. The rates of events, odds ratios (ORs), and 95% confidence intervals (CIs) were obtained using random effects meta-analysis. The analyses included data from 160 trials involving 40 432 participants. The rate was 2.43% (95% CI, 1.73%-3.22%) for all-grade adrenal insufficiency and 3.25% (95% CI, 2.15%-4.51%) for hypophysitis. Compared with the placebo or other therapeutic regimens, ICI agents were associated with a higher incidence of serious-grade adrenal insufficiency (OR 3.19, 95% CI, 1.84 to 5.54) and hypophysitis (OR 4.77, 95% CI, 2.60 to 8.78). Among 71 serious-grade hypopituitarism instances in 12 336 patients, there was a significant association between ICIs and hypopituitarism (OR 3.62, 95% CI, 1.86 to 7.03). Substantial heterogeneity was noted across the studies for the rates of these events, which in part was attributable to the different types of ICIs and varied phases of the clinical trials. Although the rates of these events were low, the risk was increased following ICI-based treatment, particularly for CTLA-4 inhibitors, which were associated with a higher incidence of pituitary-adrenal dysfunction than PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Jingli Lu
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| | - Lulu Li
- Department of PharmacyWuhan No.1 HospitalWuhanHubeiChina
| | - Yan Lan
- Department of PharmacyHuangshi Center HospitalHuangshiHubeiChina
| | - Yan Liang
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| | - Haiyang Meng
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
438
|
Tan AC, Emmett L, Lo S, Liu V, Kapoor R, Carlino MS, Guminski AD, Long GV, Menzies AM. FDG-PET response and outcome from anti-PD-1 therapy in metastatic melanoma. Ann Oncol 2019; 29:2115-2120. [PMID: 30137228 DOI: 10.1093/annonc/mdy330] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background Immune checkpoint inhibitor therapy has resulted in impressive and durable clinical activity for many cancers including melanoma; however, there remain few reliable predictors for long-term response. This study investigated whether [18F]2-fluoro-2-deoxy-D-glucose (FDG-PET) imaging may better predict long-term outcomes compared with standard computed tomography (CT) response criteria. Patients and methods Retrospective analysis of metastatic melanoma patients treated with anti-PD-1-based immunotherapy with baseline and 1-year FDG-PET and CT imaging at Melanoma Institute Australia. One-year response was determined using RECIST for CT and EORTC criteria for PET, coded as complete response (CR or CMR), partial response (PR or PMR), stable disease (SD or SMD) or progressive disease (PD or PMD). Progression-free survival (PFS) was determined from the 1-year landmark. Results Patients (n = 104) were evaluated with median follow-up 30.1 months and 98% remain alive. Most received anti-PD-1 as monotherapy (67%) or combined with ipilimumab (31%). At 1 year, 28% had CR, 66% had PR and 6% had SD on CT, while 75% had CMR, 16% PMR and 9% SMD/PMD on PET. CMR was observed in 68% of patients with PR on CT. RECIST PFS post 1-year landmark was similar in patients with CR versus PR/SD, but improved in patients with CMR versus non-CMR {median not reached [NR] versus 12.8 month; hazard ratio [HR] 0.06 [95% confidence interval (CI) 0.02-0.23]; P < 0.01}. In patients with PR on CT, PFS was improved in patients with PR + CMR versus PR + non-CMR (median NR versus 12.8 months; HR 0.07 [95% CI 0.02-0.27]; P < 0.01). In the 78 CMR patients, 78% had discontinued treatment and 96% had ongoing response. Conclusions Whilst only a small proportion of patients have a CR at 1 year, most patients with a PR have CMR on PET. Almost all patients with CMR at 1 year have ongoing response to therapy thereafter. PET may have utility in predicting long-term benefit and help guide discontinuation of therapy.
Collapse
Affiliation(s)
- A C Tan
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia
| | - L Emmett
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Nuclear Medicine, St Vincent's Hospital, Sydney, Australia; The University of New South Wales, Sydney, Australia
| | - S Lo
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia
| | - V Liu
- Department of Nuclear Medicine, St Vincent's Hospital, Sydney, Australia
| | - R Kapoor
- Department of Radiology, Royal Prince Alfred Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia
| | - M S Carlino
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, Australia
| | - A D Guminski
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia
| | - G V Long
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia
| | - A M Menzies
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia.
| |
Collapse
|
439
|
Immune and Inflammatory Cells of the Tumor Microenvironment Represent Novel Therapeutic Targets in Classical Hodgkin Lymphoma. Int J Mol Sci 2019; 20:ijms20215503. [PMID: 31694167 PMCID: PMC6862619 DOI: 10.3390/ijms20215503] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023] Open
Abstract
Classical Hodgkin Lymphoma (cHL) is a B-cell malignancy that, typically, responds well to standard therapies. However, patients who relapse after standard regimens or are refractory to induction therapy have a dismal outcome. The implementation of novel therapies such as the anti-CD30 monoclonal antibody Brentuximab Vedotin and immune checkpoint inhibitors has provided curative options for many of these patients. Nonetheless, responses are rarely durable, emphasizing the need for new agents. cHL is characterized by a unique microenvironment in which cellular and humoral components interact to promote tumor survival and dissemination. Knowledge of the complex composition of cHL microenvironment is constantly evolving; in particular, there is growing interest in certain cell subsets such as tumor-associated macrophages, myeloid-derived suppressor cells and neutrophils, all of which have a relevant role in the pathogenesis of the disease. The unique biology of the cHL microenvironment has provided opportunities to develop new drugs, many of which are currently being tested in preclinical and clinical settings. In this review, we will summarize novel insights in the crosstalk between tumor cells and non-malignant inflammatory cells. In addition, we will discuss the relevance of tumor-microenvironment interactions as potential therapeutic targets.
Collapse
|
440
|
Chow JC, Ngan RK, Cheung KM, Cho WC. Immunotherapeutic approaches in nasopharyngeal carcinoma. Expert Opin Biol Ther 2019; 19:1165-1172. [PMID: 31361154 DOI: 10.1080/14712598.2019.1650910] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022]
Abstract
Introduction: Nasopharyngeal carcinoma (NPC) is endemic in Southern China and Southeast Asia. Epstein-Barr virus (EBV) represents a unique etiological culprit in the poorly differentiated nonkeratinizing and undifferentiated subtypes. EBV antigens are expressed on tumor cells albeit in a restricted manner. Treatment options for recurrent or metastatic disease are limited. Nevertheless, emerging data from immunotherapy studies in NPC have shed light into their potential antitumor efficacy. Areas covered: This article reviews existing clinical evidence for different immunotherapeutic approaches for NPC, including adoptive cellular therapy, therapeutic cancer vaccines, and immune checkpoint inhibitors. Expert opinion: There is a growing understanding on EBV virology and the immune evasion mechanisms in NPC. Immunotherapeutic strategies leveraging these properties have shown encouraging efficacy and safety results in early-phase clinical studies. Moving forward, areas to be addressed include appropriate patient selection, optimal incorporation into standard treatment paradigms, biomarker identification, as well as the development of scalable and reproducible immune product generation processes.
Collapse
Affiliation(s)
- James Ch Chow
- Department of Clinical Oncology, Queen Elizabeth Hospital , Hong Kong SAR , China
| | - Roger Kc Ngan
- Department of Clinical Oncology, Gleneagles Hong Kong Hospital , Hong Kong SAR, China
| | - K M Cheung
- Department of Clinical Oncology, Queen Elizabeth Hospital , Hong Kong SAR , China
| | - William Cs Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital , Hong Kong SAR , China
| |
Collapse
|
441
|
Targeting BCL-W and BCL-XL as a therapeutic strategy for Hodgkin lymphoma. Leukemia 2019; 34:947-952. [PMID: 31636344 PMCID: PMC7056566 DOI: 10.1038/s41375-019-0611-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/20/2019] [Accepted: 08/28/2019] [Indexed: 01/18/2023]
|
442
|
Chen A, Mokrane FZ, Schwartz LH, Morschhauser F, Stamatoullas A, Schiano de Colella JM, Vercellino L, Casasnovas O, Chauchet A, Delmer A, Nicolas-Virelizier E, Ghesquières H, Moles-Moreau MP, Schmitt A, Dulery R, Bouabdallah K, Borel C, Touati M, Deau-Fischer B, Peyrade F, Seban RD, Manson G, Armand P, Houot R, Dercle L. Early 18F-FDG PET/CT Response Predicts Survival in Relapsed or Refractory Hodgkin Lymphoma Treated with Nivolumab. J Nucl Med 2019; 61:649-654. [PMID: 31628220 DOI: 10.2967/jnumed.119.232827] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/25/2019] [Indexed: 11/16/2022] Open
Abstract
Monoclonal antibodies (mAbs) against programmed cell death 1 (PD-1), such as nivolumab and pembrolizumab, are associated with high response rates in patients with relapsed or refractory classic Hodgkin lymphoma (HL). To date, no prognostic factor for overall survival (OS) has been established with these agents in HL. We examined whether the first early response assessment evaluated using 18F-FDG PET/CT may be associated with OS in this setting. Methods: This retrospective study included 45 patients from 34 institutions. In a masked, centralized review, 3 independent radiologists classified PET/CT scans obtained at a median of 2.0 mo (interquartile range, 1.7-3.7 mo) after nivolumab initiation using existing criteria (i.e., 2014 Lugano classification and 2016 LYRIC). Patients were classified according to 4 possible response categories: complete metabolic response (CMR), partial metabolic response (PMR), no metabolic response (NMR), or progressive metabolic disease (PMD). Because the OS of patients with NMR and PMR was similar, they were grouped together. OS was estimated using the Kaplan-Meier method and compared between groups using log-rank testing. Results: Eleven patients (24%) died after a median follow-up of 21.2 mo. The classification was identical between Lugano and LYRIC because all 16 progression events classified as indeterminate response per LYRIC were confirmed on subsequent evaluations. Both Lugano and LYRIC classified patients as CMR in 13 cases (29%), PMD in 16 (36%), NMR in 4 (9%), and PMR in 12 (27%). The 2-y OS probability was significantly different in patients with PMD (0.53; 95% confidence interval [95%CI], 0.32-0.87), NMR or PMR (0.80; 95%CI, 0.63-1.00), and CMR (1.00; 95%CI, 1.00-1.00) in the overall population (P = 0.02, 45 patients), as well as according to a landmark analysis at 3 mo (P = 0.05, 32 patients). Conclusion: In relapsed or refractory HL patients treated with anti-PD-1 mAbs, the first early PET/CT assessment using either Lugano or LYRIC predicted OS and allowed early risk stratification, suggesting that PET/CT might be used to develop risk-adapted strategies.
Collapse
Affiliation(s)
- Aiping Chen
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, New York.,Department of Radiology, First Affiliated Hospital of NanJing Medical University, Nanjing, China
| | - Fatima-Zohra Mokrane
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, New York.,Radiology Department. Rangueil University Hospital, Toulouse, France
| | - Lawrence H Schwartz
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, New York
| | - Franck Morschhauser
- EA 7365-GRITA (Groupe de Recherche sur les Formes Injectables et les Technologies Associées), University Lille, CHU Lille, Lille, France
| | | | | | | | | | - Adrien Chauchet
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Alain Delmer
- Department of Hematology, University Hospital of Reims, Reims, France
| | | | | | | | - Anna Schmitt
- Department of Hematology, Bergonie Institute, Bordeaux, France
| | - Rémy Dulery
- Department of Hematology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - Krimo Bouabdallah
- Department of Hematology, University Hospital of Bordeaux, Bordeaux, France
| | - Cecile Borel
- Department of Hematology, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Mohamed Touati
- Department of Hematology, University Hospital of Limoges, Limoges, France
| | | | | | | | - Guillaume Manson
- Department of Hematology, University Hospital of Rennes, Rennes, France.,U1236, INSERM, Rennes, France
| | - Philippe Armand
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; and
| | - Roch Houot
- Department of Hematology, University Hospital of Rennes, Rennes, France.,U1236, INSERM, Rennes, France
| | - Laurent Dercle
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, New York .,UMR1015, INSERM, Institut Gustave Roussy, Université Paris Saclay, Villejuif, France
| |
Collapse
|
443
|
Hotchkiss RS, Colston E, Yende S, Crouser ED, Martin GS, Albertson T, Bartz RR, Brakenridge SC, Delano MJ, Park PK, Donnino MW, Tidswell M, Mayr FB, Angus DC, Coopersmith CM, Moldawer LL, Catlett IM, Girgis IG, Ye J, Grasela DM. Immune checkpoint inhibition in sepsis: a Phase 1b randomized study to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of nivolumab. Intensive Care Med 2019; 45:1360-1371. [PMID: 31576433 DOI: 10.1007/s00134-019-05704-z] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Sepsis-associated immunosuppression increases hospital-acquired infection and viral reactivation risk. A key underlying mechanism is programmed cell death protein-1 (PD-1)-mediated T-cell function impairment. This is one of the first clinical safety and pharmacokinetics (PK) assessments of the anti-PD-1 antibody nivolumab and its effect on immune biomarkers in sepsis. METHODS Randomized, double-blind, parallel-group, Phase 1b study in 31 adults at 10 US hospital ICUs with sepsis diagnosed ≥ 24 h before study treatment, ≥ 1 organ dysfunction, and absolute lymphocyte count ≤ 1.1 × 103 cells/μL. Participants received one nivolumab dose [480 mg (n = 15) or 960 mg (n = 16)]; follow-up was 90 days. Primary endpoints were safety and PK parameters. RESULTS Twelve deaths occurred [n = 6 per study arm; 40% (480 mg) and 37.5% (960 mg)]. Serious AEs occurred in eight participants [n = 1, 6.7% (480 mg); n = 7, 43.8% (960 mg)]. AEs considered by the investigator to be possibly drug-related and immune-mediated occurred in five participants [n = 2, 13.3% (480 mg); n = 3, 18.8% (960 mg)]. Mean ± SD terminal half-life was 14.7 ± 5.3 (480 mg) and 15.8 ± 7.9 (960 mg) days. All participants maintained > 90% receptor occupancy (RO) 28 days post-infusion. Median (Q1, Q3) mHLA-DR levels increased to 11,531 (6528, 19,495) and 11,449 (6225, 16,698) mAbs/cell in the 480- and 960-mg arms by day 14, respectively. Pro-inflammatory cytokine levels did not increase. CONCLUSIONS In this sepsis population, nivolumab administration did not result in unexpected safety findings or indicate any 'cytokine storm'. The PK profile maintained RO > 90% for ≥ 28 days. Further efficacy and safety studies are warranted. TRIAL REGISTRATION NUMBER (CLINICALTRIALS.GOV): NCT02960854.
Collapse
Affiliation(s)
- Richard S Hotchkiss
- Departments of Anesthesiology, Medicine, and Surgery, Washington University School of Medicine, St Louis, 660 South Euclid Avenue, St Louis, MO, 63110-1093, USA.
| | - Elizabeth Colston
- Innovative Medicines Development, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Sachin Yende
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.,The CRISMA Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Greg S Martin
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Timothy Albertson
- Department of Internal Medicine, University of California, Davis, Davis, CA, USA.,Northern California Veterans Administration Health Care System, Mather, CA, USA
| | - Raquel R Bartz
- Department of Anesthesiology and Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Scott C Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Matthew J Delano
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Pauline K Park
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Michael W Donnino
- Department of Emergency Medicine and Department of Medicine (Division of Pulmonary and Critical Care), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mark Tidswell
- Division of Pulmonary and Critical Care Medicine, Baystate Medical Center, Springfield, MA, USA
| | - Florian B Mayr
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.,The CRISMA Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Derek C Angus
- The CRISMA Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ian M Catlett
- Innovative Medicines Development, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Ihab G Girgis
- Innovative Medicines Development, Bristol-Myers Squibb, Princeton, NJ, USA
| | - June Ye
- Innovative Medicines Development, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Dennis M Grasela
- Innovative Medicines Development, Bristol-Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
444
|
Momotow J, Goergen H, Behringer K, Bröckelmann PJ, Borchmann S, Tresckow BV, Kobe C, Engert A, Sasse S. Nivolumab in Relapsed/Refractory Classical Hodgkin Lymphoma - Extended Follow-up of 30 Patients Treated Within the CheckMate 205 Trial in a Single-Center. Hemasphere 2019; 3:e293. [PMID: 31942546 PMCID: PMC6919467 DOI: 10.1097/hs9.0000000000000293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/14/2019] [Accepted: 08/15/2019] [Indexed: 11/25/2022] Open
Affiliation(s)
- Jesko Momotow
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne
- German Hodgkin Study Group (GHSG)
| | - Helen Goergen
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne
- German Hodgkin Study Group (GHSG)
| | - Karolin Behringer
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne
- German Hodgkin Study Group (GHSG)
| | - Paul J. Bröckelmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne
- German Hodgkin Study Group (GHSG)
| | - Sven Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne
- German Hodgkin Study Group (GHSG)
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Center for Molecular Medicine, University of Cologne
- Faculty of Medicine, Else Kröner Forschungskolleg Clonal Evolution in Cancer, University of Cologne
| | - Bastian v. Tresckow
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne
- German Hodgkin Study Group (GHSG)
| | - Carsten Kobe
- German Hodgkin Study Group (GHSG)
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
| | - Andreas Engert
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne
- German Hodgkin Study Group (GHSG)
| | - Stephanie Sasse
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne
- German Hodgkin Study Group (GHSG)
| |
Collapse
|
445
|
Shahid K, Khalife M, Dabney R, Phan AT. Immunotherapy and targeted therapy-the new roadmap in cancer treatment. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:595. [PMID: 31807576 DOI: 10.21037/atm.2019.05.58] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Immunotherapy is the new frontier in cancer medicine. This manuscript summarizes historical aspect of immunotherapy, particularly its pathway to drug approval as the main therapeutic modality used in clinical medicine. We will discuss the role of immunotherapy in treating various types of cancers and how the treatment landscape once dominated by chemotherapy is rapidly changing.
Collapse
Affiliation(s)
- Kamran Shahid
- Division of Hematology & Oncology, University of Texas Health Science Center at Tyler, UT Health North Campus Tyler, Tyler, TX, USA
| | - Mustafa Khalife
- Division of Hematology & Oncology, University of Texas Health Science Center at Tyler, UT Health North Campus Tyler, Tyler, TX, USA
| | - Raetasha Dabney
- Division of Hematology & Oncology, University of Texas Health Science Center at Tyler, UT Health North Campus Tyler, Tyler, TX, USA
| | - Alexandria T Phan
- Division of Hematology & Oncology, University of Texas Health Science Center at Tyler, UT Health North Campus Tyler, Tyler, TX, USA
| |
Collapse
|
446
|
Schütte J, Reusch J, Khandanpour C, Eisfeld C. Structural Variants as a Basis for Targeted Therapies in Hematological Malignancies. Front Oncol 2019; 9:839. [PMID: 31555592 PMCID: PMC6722867 DOI: 10.3389/fonc.2019.00839] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/14/2019] [Indexed: 11/13/2022] Open
Abstract
Structural variants (SV) are changes in the genomic landscape that can alter gene expression levels and thus lead to disease development. The most common and best studied SVs in hematological malignancies are chromosomal translocations. Here, parts of two genes that are normally on different chromosomes come into close proximity due to a failure in DNA repair. As a consequence, fusion proteins which show a different function and/or cellular localization compared to the two original proteins are expressed, sometimes even at different levels. The identification of chromosomal translocations is often used to identify the specific disease a patient is suffering from. In addition, SVs such as deletions, duplications, inversions and single nucleotide polymorphisms (SNPs) can occur in hematopoietic cells and lead to their malignant transformations. Changes in the 3D genome structure have also recently been shown to impact disease development. In this review, we describe a variety of SVs occurring in different subtypes of hematological malignancies. Currently, most therapeutic approaches target fusion proteins which are the cellular product of chromosomal translocations. However, amplifications and SNPs also play a role in disease progression and can be targeted. We present some examples for different types of structural variants and how they are currently treated.
Collapse
Affiliation(s)
- Judith Schütte
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | - Julia Reusch
- Medizinische Fakultät, Universität Münster, Münster, Germany
| | | | | |
Collapse
|
447
|
Sell S, Ilic Z. Comparison of survivor scores for differentiation therapy of cancer to those for checkpoint inhibition: Half full or half empty. Tumour Biol 2019; 41:1010428319873749. [PMID: 31496424 DOI: 10.1177/1010428319873749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Differentiation therapy is directed to the self-renewing cancer stem cells, as well as their progeny transit amplifying cells, to force them to mature to terminal differentiation. Differentiation therapy is effective in treatment of neuroblastomas and myeloid leukemias. Checkpoint inhibition therapy removes blocks to cancer reactive T-killer cells and allows them to react to malignant cells and limit the growth of cancer. The percentage of patients with a given cancer that responds to either therapy is less than hoped for, and the duration of response is variable. Multiplying the response rate (percentage of patients responding to therapy) by the duration of response may be used to derive a survival score for patients treated with differentiation therapy or checkpoint inhibition. By this criterion, differentiation therapy gives better survival scores than checkpoint inhibition. Yet, checkpoint inhibition is considered a great success, mostly because it may be applied to many different types of cancer, and differentiation therapy is considered relatively ineffective because it is limited to a few specific cancers. On the other hand, the cost of checkpoint inhibition treatment is 10-20 times more per patient than that of differentiation therapy. Hopefully, future combined treatments and advances in both approaches will increase the effectiveness of these cancer treatments.
Collapse
Affiliation(s)
- Stewart Sell
- Division of Translational Medicine, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Albany College of Pharmacy and University at Albany, Albany, NY, USA
| | - Zoran Ilic
- Division of Translational Medicine, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| |
Collapse
|
448
|
Forbes SG, Mistry HE. Immune-Mediated Therapies in Lymphoma. Semin Oncol Nurs 2019; 35:150925. [PMID: 31526549 DOI: 10.1016/j.soncn.2019.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To review types of lymphoma, risk factors, and evaluate novel immune-mediated therapies, including side effects and management of immune-mediated toxicities. DATA SOURCE Published literature, national statistics, and Web sites. CONCLUSION Novel biologic agents are being developed with the potential to improve outcomes. However, these novel agents pose unique and sometimes serious adverse events. IMPLICATIONS FOR NURSING PRACTICE The immune-mediated adverse events require a multidisciplinary approach and early identification. It is imperative providers and nurses are educated on the management of the unique toxicities caused by lymphoma treatment.
Collapse
|
449
|
Song Y, Gao Q, Zhang H, Fan L, Zhou J, Zou D, Li W, Yang H, Liu T, Wang Q, Lv F, Guo H, Yang L, Elstrom R, Huang J, Novotny W, Wei V, Zhu J. Treatment of relapsed or refractory classical Hodgkin lymphoma with the anti-PD-1, tislelizumab: results of a phase 2, single-arm, multicenter study. Leukemia 2019; 34:533-542. [PMID: 31520078 PMCID: PMC7214259 DOI: 10.1038/s41375-019-0545-2] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/01/2019] [Indexed: 02/05/2023]
Abstract
Prognosis is poor for patients with relapsed/refractory (R/R) classical Hodgkin lymphoma (cHL) after failure of or who are ineligible for autologous stem cell transplant. We evaluated the efficacy and safety of tislelizumab, an investigational anti-PD-1 monoclonal antibody, in phase 2, single-arm study in Chinese patients with R/R cHL. The primary endpoint was overall response rate as assessed by an independent review committee, according to the Lugano 2014 Classification. Seventy patients were enrolled in the study and received at least one dose of tislelizumab. After median follow-up of 9.8 months, 61 (87.1%) patients achieved an objective response, with 44 (62.9%) achieving a complete response (CR). The estimated 9-month progression-free survival rate was 74.5%. Most common grade ≥3 adverse events (AEs) were upper respiratory tract infection and pneumonitis. Infusion-related reactions occurred in 27 (38.6%) patients, and 27 patients (38.6%) experienced an immune-related AE, the most common of which was thyroid dysfunction. Eleven (15.7%) patients experienced at least one treatment-emergent AE leading to dose interruption or delay. No deaths occurred due to AEs. Treatment of patients with R/R cHL with tislelizumab was generally well tolerated and resulted in high overall response and CR rates, potentially translating into more durable responses for these patients.
Collapse
Affiliation(s)
- Yuqin Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Quanli Gao
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Huilai Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei Fan
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Wuhan, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wei Li
- Department of Hematology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Haiyan Yang
- Department of Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ting Liu
- Department of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Quanshun Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Fangfang Lv
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haiyi Guo
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Liudi Yang
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | | | | | | | - Vivian Wei
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Jun Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
450
|
Wang J, Fei K, Jing H, Wu Z, Wu W, Zhou S, Ni H, Chen B, Xiong Y, Liu Y, Peng B, Yu D, Jiang H, Liu J. Durable blockade of PD-1 signaling links preclinical efficacy of sintilimab to its clinical benefit. MAbs 2019; 11:1443-1451. [PMID: 31402780 PMCID: PMC6816392 DOI: 10.1080/19420862.2019.1654303] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Blockade of immune checkpoint pathways by programmed cell death protein 1 (PD-1) antibodies has demonstrated broad clinical efficacy against a variety of malignancies. Sintilimab, a highly selective, fully human monoclonal antibody (mAb), blocks the interaction of PD-1 and its ligands and has demonstrated clinical benefit in various clinical studies. Here, we evaluated the affinity of sintilimab to human PD-1 by surface plasmon resonance and mesoscale discovery and evaluated PD-1 receptor occupancy and anti-tumor efficacy of sintilimab in a humanized NOD/Shi-scid-IL2rgamma (null) (NOG) mouse model. We also assessed the receptor occupancy and immunogenicity of sintilimab from clinical studies in humans (9 patients with advanced solid tumor and 381 patients from 4 clinical studies, respectively). Sintilimab bound to human PD-1 with greater affinity than nivolumab (Opdivo®, MDX-1106) and pembrolizumab (Keytruda®, MK-3475). The high affinity of sintilimab is explained by its distinct structural binding mode to PD-1. The pharmacokinetic behavior of sintilimab did not show any significant differences compared to the other two anti-PD-1 mAbs. In the humanized NOG mouse model, sintilimab showed superior PD-1 occupancy on circulating T cells and a stronger anti-tumor effect against NCI-H292 tumors. The strong anti-tumor response correlated with increased interferon-γ-secreting, tumor-specific CD8+ T cells, but not with CD4+ Tregs in tumor tissue. Pharmacodynamics testing indicated a sustained mean occupancy of ≥95% of PD-1 molecules on circulating T cells in patients following sintilimab infusion, regardless of infusion dose. Sintilimab infusion was associated with 0.52% (2/381 patients) of anti-drug antibodies and 0.26% (1/381 patients) neutralizing antibodies. These data validate sintilimab as a novel, safe, and efficacious anti-PD-1 mAb for cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Wang
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Keke Fei
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Hua Jing
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Zhihai Wu
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Weiwei Wu
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Shuaixiang Zhou
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Haiqing Ni
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Bingliang Chen
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Yan Xiong
- Department of Translational Science, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Yanpeng Liu
- Department of Translational Science, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Bo Peng
- Department of Translational Science, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Dechao Yu
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Haiping Jiang
- Department of Medical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou , Zhejiang Province , China
| | - Junjian Liu
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| |
Collapse
|