551
|
Recommendations for managing PD-1 blockade in the context of allogeneic HCT in Hodgkin lymphoma: taming a necessary evil. Blood 2018; 132:9-16. [DOI: 10.1182/blood-2018-02-811174] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022] Open
Abstract
Abstract
PD-1 blockade is an effective therapy in relapsed/refractory (R/R) classical Hodgkin Lymphoma (cHL) who have relapsed after or are ineligible for autologous hematopoietic cell transplantation (HCT). Although single-agent anti-PD-1 monoclonal antibodies (mAb’s) are associated with high response rates and durable remissions, available results to date suggest that a large majority of patients will eventually progress on therapy. Many of these patients are potential candidates for allogeneic HCT (allo-HCT) after receiving anti-PD-1 mAb’s, and allo-HCT remains for now the only treatment with demonstrated curative potential in this setting. However, initial reports suggested that allo-HCT in this setting may be associated with increased risk of early transplant-related toxicity, likely driven by lingering effects of PD-1 blockade. Furthermore, many patients with R/R cHL who undergo allo-HCT will relapse after transplantation, most often with limited treatment options. Here again, PD-1 blockade appears to yield high response rates, but with an increased risk of attendant immune toxicity. Many questions remain regarding the use of PD-1 blockade before or after allo-HCT, especially in relation to the feasibility, outcome, optimal timing, and method of allo-HCT after PD-1 blockade. Despite the scarcity of prospective data, these questions are unavoidable and must be tackled by clinicians in the routine care of patients with advanced cHL. We provide consensus recommendations of a working group based on available data and experience, in an effort to help guide treatment decisions until more definitive data are obtained.
Collapse
|
552
|
Abstract
PURPOSE OF REVIEW This review discusses novel immunotherapeutic approaches to treat Hodgkin lymphoma (HL), specifically PD-1 inhibitors and cellular immunotherapy. RECENT FINDINGS PD-1 inhibitors have shown promising results in the treatment of relapsed or refractory HL, leading to FDA approval of nivolumab and pembrolizumab, although complete remissions are rare. Chimeric antigen receptor T cells directed against CD30 have been investigated with preliminary clinical trials showing minimal toxicities and some responses in heavily pre-treated patients with HL. HL is unique as it consists of a small percentage of malignant cells (Hodgkin Reed Sternberg cells) surrounded by an inflammatory microenvironment which promotes tumor growth and suppresses immune responses, making it an ideal target for immunotherapeutic approaches, such as PD-1 inhibitors and cellular immunotherapy. Current research is focused on overcoming barriers to efficacy via rational combinations that overcome resistance to therapy.
Collapse
Affiliation(s)
- Natalie S Grover
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27516, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27516, USA.
| |
Collapse
|
553
|
Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol 2018; 62:29-39. [PMID: 29990692 DOI: 10.1016/j.intimp.2018.06.001] [Citation(s) in RCA: 845] [Impact Index Per Article: 120.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022]
Abstract
Although T lymphocytes have long been appreciated for their role in the immunosurveillance of cancer, it has been the realization that cancer cells may ultimately escape a response from tumor-reactive T cells that has ignited efforts to enhance the efficacy of anti-tumor immune responses. Recent advances in our understanding of T cell immunobiology have been particularly instrumental in informing therapeutic strategies to overcome mechanisms of tumor immune escape, and immune checkpoint blockade has emerged as one of the most promising therapeutic options for patients in the history of cancer treatment. Designed to interfere with inhibitory pathways that naturally constrain T cell reactivity, immune checkpoint blockade releases inherent limits on the activation and maintenance of T cell effector function. In the context of cancer, where negative T cell regulatory pathways are often overactive, immune checkpoint blockade has proven to be an effective strategy for enhancing the effector activity and clinical impact of anti-tumor T cells. Checkpoint inhibitors targeting CTLA-4, PD-1, and PD-L1 have yielded unprecedented and durable responses in a significant percentage of cancer patients in recent years, leading to U.S. FDA approval of six checkpoint inhibitors for numerous cancer indications since 2011. In this review, we highlight the clinical success of these FDA-approved immune checkpoint inhibitors and discuss current challenges and future strategies that must be considered going forward to maximize the efficacy of immune checkpoint blockade therapy for cancer.
Collapse
|
554
|
Xu PP, Sun C, Cao X, Zhao X, Dai HJ, Lu S, Guo JJ, Fu SJ, Liu YX, Li SC, Chen M, McCord R, Venstrom J, Szafer-Glusman E, Punnoose E, Kiermaier A, Cheng G, Zhao WL. Immune Characteristics of Chinese Diffuse Large B-Cell Lymphoma Patients: Implications for Cancer Immunotherapies. EBioMedicine 2018; 33:94-104. [PMID: 29936139 PMCID: PMC6085499 DOI: 10.1016/j.ebiom.2018.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/28/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022] Open
Abstract
Immunotherapeutic agents have demonstrated encouraging signs of clinical utility in non-Hodgkin lymphoma. The goal of this study is to analyze the immune characteristics of Chinese patients with diffuse large B-cell lymphoma (DLBCL) to inform the development of immunotherapies in this patient population. Tumor samples from 211 DLBCL patients were analyzed for cell of origin (COO) and immune characteristics using the NanoString platform as well as MYC protein expression through immunohistochemistry. Lower incidence of the germinal center B-cell (GCB) subtype (93/211, 44.1%) was observed in this cohort. Compared to the GCB subtype, the activated B-cell (ABC) subtype was associated with significantly increased expression of multiple pro-inflammatory gene signatures and decreased expression of anti-inflammatory gene signatures. Instead of affecting the pro-inflammatory genes, MYC protein overexpression showed a negative correlation with the expression of T-cell receptor (TCR) and T regulatory genes as well as the OX40 gene. Regardless of COO, higher PD-L1 or IDO1 gene expression correlated with increased expression of T effector and Interferon-γ gene signatures while the expression of multiple oncogenes including ACTR3B, ERBB2, AKT2 and SMARCD1 was down-regulated. Our findings may thus be helpful in guiding further development of immunotherapies for the different subsets of Chinese DLBCL patients.
Collapse
Affiliation(s)
- Peng-Peng Xu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun Sun
- Oncology Biomarker Development, Genentech Inc., Shanghai, China
| | - Xu Cao
- Oncology Biomarker Development, Genentech Inc., Shanghai, China
| | - Xia Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hang-Jun Dai
- Roche Product Development in Asia Pacific, Roche (China) Holding, Ltd., Shanghai, China
| | - Shan Lu
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - Jian-Jun Guo
- Oncology Biomarker Development, Genentech Inc., Shanghai, China
| | - Shi-Jing Fu
- Oncology Biomarker Development, Genentech Inc., Shanghai, China
| | - Yu-Xia Liu
- Oncology Biomarker Development, Genentech Inc., Shanghai, China
| | - Su-Chun Li
- Roche Product Development in Asia Pacific, Roche (China) Holding, Ltd., Shanghai, China
| | - Meng Chen
- Roche Product Development in Asia Pacific, Roche (China) Holding, Ltd., Shanghai, China
| | - Ron McCord
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - Jeff Venstrom
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | | | - Elizabeth Punnoose
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - Astrid Kiermaier
- Oncology Biomarker Development, Genentech Inc., Basel, Switzerland
| | - Gang Cheng
- Oncology Biomarker Development, Genentech Inc., Shanghai, China.
| | - Wei-Li Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
555
|
Atanackovic D, Luetkens T. Biomarkers for checkpoint inhibition in hematologic malignancies. Semin Cancer Biol 2018; 52:198-206. [PMID: 29775689 DOI: 10.1016/j.semcancer.2018.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 01/27/2023]
Abstract
In the past few years we have seen remarkable paradigm shifts in the treatment of many solid tumors due to the introduction of inhibitors targeting immune checkpoints such as PD-1/PD-L1 and CTLA-4. Recent results indicate that checkpoint inhibition also represents a very promising approach for certain types of hematologic malignancies. Unfortunately, treatment with checkpoint inhibitors is also associated with substantial toxicities and high costs and only a subset of patients appears to derive clinical benefit from these treatments. This demonstrates the urgent need for biomarkers for the identification of patient populations that are likely to respond to this type of therapy and/or have fewer side effects. Here, we have reviewed available information on the prognostic and predictive value of biomarkers for anti-CTLA-4 and anti-PD-1/PD-L1 as the most commonly used checkpoint inhibitors. There are currently no reliable biomarkers capable of predicting responses to anti-CTLA-4 agents, such as ipilimumab, in hematologic malignancies. Certain polymorphisms in the CTLA-4 gene, however, seem to have an impact on the patients' outcome, especially in the case of chronic lymphocytic leukemia (CLL). There is now sufficient data supporting PD-L1 expression levels in the tumor tissue as an independent prognostic factor in B cell lymphomas such as diffuse large B-cell lymphoma (DLBCL). Overexpression of PD-L1 in the tumor tissue and elevated serum levels of soluble PD-L1 may also represent adverse prognostic factors in certain subtypes of T cell lymphomas. Finally, expression levels of PD-L1 also seem to predict responses to anti-PD-1/PD-L1 approaches in patients with Hodgkin lymphoma. Future studies will have to further delineate the prognostic/predictive role of PD-L1 expression as a biomarker in hematologic malignancies and may be able to identify confounding variables, which will hopefully to some extent be generalizable to other types of anti-tumor immunotherapies.
Collapse
Affiliation(s)
- Djordje Atanackovic
- Multiple Myeloma Program & Cancer Immunology, Division of Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, United States.
| | - Tim Luetkens
- Multiple Myeloma Program & Cancer Immunology, Division of Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, United States
| |
Collapse
|
556
|
Meti N, Esfahani K, Johnson NA. The Role of Immune Checkpoint Inhibitors in Classical Hodgkin Lymphoma. Cancers (Basel) 2018; 10:cancers10060204. [PMID: 29914088 PMCID: PMC6025119 DOI: 10.3390/cancers10060204] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/04/2018] [Accepted: 06/12/2018] [Indexed: 01/06/2023] Open
Abstract
Hodgkin Lymphoma (HL) is a unique disease entity both in its pathology and the young patient population that it primarily affects. Although cure rates are high, survivorship can be linked with significant recent long-term morbidity associated with both chemotherapy and radiotherapy. The most significant advances have been with the use of the anti-CD30-drug conjugated antibody brentuximab vedotin (BV) and inhibitors of program death 1 (PD-1). HL is genetically wired to up-regulate program death ligand 1 (PD-L1) in >95% of cases, creating a state of so-called “T cell exhaustion”, which can be reversed with immune checkpoint-inhibitor blockade. The overall and complete response rates to PD-1 inhibitors in patients with relapsed or refractory HL are 70% and 20%, respectively, with a long median duration of response of ~16 months. In fact, PD-1 inhibitors can benefit a wide spectrum of relapsed HL patients, including some who have “progressive disease” by strict response criteria. We review the biology of HL, with a focus on the immune micro-environment and mechanisms of immune evasion. We also provide the rationale supporting the use of PD-1 inhibitors in HL and highlight some of the challenges of monitoring disease response in patients treated with this immunotherapy.
Collapse
Affiliation(s)
- Nicholas Meti
- Department of Medicine, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada.
| | - Khashayar Esfahani
- Department of Medicine, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada.
| | - Nathalie A Johnson
- Department of Medicine, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada.
| |
Collapse
|
557
|
Affiliation(s)
| | - Jayesh Desai
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia
- Medical Oncology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
558
|
Tsirigotis P, Vassilakopoulos T, Batsis I, Bousiou Z, Gkirkas K, Sakellari I, Kaloyannidis P, Roussou P, Pangalis GA, Moschogiannis M, Vassilopoulos G, Repousis P, Megalakaki A, Michalis E, Kalpadakis C, Papadaki HA, Kotsianidis I, Hatzimichael E, Spyridonidis A, Anargyrou K, Poulakidas E, Giannoullia P, Apostolidis I, Stamouli M, Konstantopoulos K, Pappa V, Panayiotidis P, Harhalakis N, Anagnostopoulos A, Angelopoulou M. Positive impact of brentuximab vedotin on overall survival of patients with classical Hodgkin lymphoma who relapse or progress after autologous stem cell transplantation: A nationwide analysis. Hematol Oncol 2018; 36:645-650. [PMID: 29882363 DOI: 10.1002/hon.2521] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 11/12/2022]
Abstract
The outcome of patients with relapsed/refractory classical Hodgkin lymphoma (R/R cHL) after autologous stem cell transplantation (auto-SCT) is poor. Recently, the anti-CD30 monoclonal antibody-drug conjugate, brentuximab vedotin (BV), has shown remarkable activity in the setting of R/R cHL. In the pivotal phase II study, BV produced an overall response rate of 75% and a median progression-free survival of 6.7 months. Although these results have been reproduced by large registry studies, the impact of BV on the overall survival (OS) of patients with R/R cHL has not been addressed so far. The aim of this study was to examine the impact of BV on OS in the setting of post auto-SCT R/R cHL. Analysis was performed in a group of patients with R/R cHL after a previous auto-SCT reported in the Greek registry during the last 2 decades. By using a multivariate model and censoring patients at the time of subsequent allo-SCT or treatment with immune checkpoint inhibitors, we showed that treatment with BV in the posttransplant relapse setting has a positive impact on the outcome and results in significant improvement of OS. To our knowledge, this the first published study, addressing the impact of BV on the OS in the setting of posttransplant relapse.
Collapse
Affiliation(s)
- Panagiotis Tsirigotis
- Second Dept of Internal Medicine Propaedeutic, ATTIKO General University Hospital, Athens, Greece
| | | | - Ioannis Batsis
- Dept of Hematology and BMT, PAPANIKOLAOU Hospital, Thessaloniki, Greece
| | - Zoi Bousiou
- Dept of Hematology and BMT, PAPANIKOLAOU Hospital, Thessaloniki, Greece
| | - Konstantinos Gkirkas
- Second Dept of Internal Medicine Propaedeutic, ATTIKO General University Hospital, Athens, Greece
| | - Ioanna Sakellari
- Dept of Hematology and BMT, PAPANIKOLAOU Hospital, Thessaloniki, Greece
| | | | | | | | | | | | | | | | | | | | - Helen A Papadaki
- Dept of Hematology, University Hospital of Crete, Heraklion, Greece
| | | | | | | | | | - Elias Poulakidas
- Dept of Hematology, 401 Military Hospital of Athens, Athens, Greece
| | | | | | - Maria Stamouli
- Second Dept of Internal Medicine Propaedeutic, ATTIKO General University Hospital, Athens, Greece
| | | | - Vassiliki Pappa
- Second Dept of Internal Medicine Propaedeutic, ATTIKO General University Hospital, Athens, Greece
| | | | | | | | | |
Collapse
|
559
|
Gong J, Le TQ, Massarelli E, Hendifar AE, Tuli R. Radiation therapy and PD-1/PD-L1 blockade: the clinical development of an evolving anticancer combination. J Immunother Cancer 2018; 6:46. [PMID: 29866197 PMCID: PMC5987486 DOI: 10.1186/s40425-018-0361-7] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 05/16/2018] [Indexed: 02/06/2023] Open
Abstract
Several inhibitors of programmed cell death-1 (PD-1) and programmed death ligand-1 (PD-L1) have been approved as a form of immunotherapy for multiple cancers. Ionizing radiation therapy (RT) has been shown to enhance the priming and effector phases of the antitumor T-cell response rendering it an attractive therapy to combine with PD-1/PD-L1 inhibitors. Preclinical data support the rational combination of the 2 modalities and has paved way for the clinical development of the combination across a spectrum of cancers. In this review, we highlight the preclinical and clinical development of combined RT and PD-1/PD-L1 blockade to date. In addition to a comprehensive evaluation of available safety and efficacy data, we discuss important points of consideration in clinical trial design for this promising combination.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Thang Q Le
- Division of Angiography and Interventional Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Erminia Massarelli
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Andrew E Hendifar
- Division of Medical Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Richard Tuli
- Departments of Radiation Oncology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1023, Los Angeles, CA, 90048, USA.
| |
Collapse
|
560
|
Toxicities associated with immunotherapies for hematologic malignancies. Best Pract Res Clin Haematol 2018; 31:158-165. [DOI: 10.1016/j.beha.2018.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 03/12/2018] [Indexed: 12/20/2022]
|
561
|
Abdel-Rahman O, Eltobgy M, Oweira H, Giryes A, Tekbas A, Decker M. Immune-related musculoskeletal toxicities among cancer patients treated with immune checkpoint inhibitors: a systematic review. Immunotherapy 2018; 9:1175-1183. [PMID: 29067884 DOI: 10.2217/imt-2017-0108] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM Immune-related musculoskeletal toxicities are uncommon but potentially serious adverse events; and they may accompany the use of immune checkpoint inhibitors (ICIs). The objective of this systematic review is to assess the patterns of these musculoskeletal toxicities. METHODS & RESULTS PubMed database has been searched till May 2017. Clinical studies and case reports reporting the occurrence of immune-related musculoskeletal toxicities (other than arthralgia and myalgia) in cancer patients treated with ICIs were included. Eight trials with 2263 participants were included. Likewise, nine case reports reporting the outcomes of 12 patients were included. CONCLUSION Immune-related arthritis and myositis occur uncommonly in cancer patients treated with ICIs. Further studies are required to better describe the pathogenesis as well as the time course of these events.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Lotfy Elsayed Street, Cairo, 11566, Egypt
| | | | - Hani Oweira
- Oncology Department, Swiss Cancer Institute, Cham, Switzerland 3OncoCentrum Zurich, Gastrointestinal Tumor Center Zurich (GITZ), Zurich, Switzerland.,Surgical Center Zurich - Hirslanden Hospital Zurich, Zurich, Switzerland.,Surgery Department, University of Jena, Jena, Germany
| | - Anwar Giryes
- Oncology Department, Swiss Cancer Institute, Cham, Switzerland 3OncoCentrum Zurich, Gastrointestinal Tumor Center Zurich (GITZ), Zurich, Switzerland
| | - Aysun Tekbas
- Surgery Department, University of Jena, Jena, Germany
| | - Michael Decker
- Oncology Department, Zentrum für Integrative Onkologie, 8001 Zurich, Switzerland
| |
Collapse
|
562
|
Carella AM, Corradini P, Mussetti A, Ricardi U, Vitolo U, Viviani S. Treatment of classical Hodgkin lymphoma in the era of brentuximab vedotin and immune checkpoint inhibitors. Ann Hematol 2018; 97:1301-1315. [PMID: 29802458 DOI: 10.1007/s00277-018-3366-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 05/06/2018] [Indexed: 12/19/2022]
Abstract
The majority of Hodgkin lymphoma patients are now cured with conventional first-line therapy; however, 10-15% of early-stage disease and less than 30% of advanced-stage patients are refractory(rare) or relapsed. Salvage second-line therapy combined with high-dose therapy and autologous stem-cell transplantation can cure 40-50% of patients. Recently novel agents (Brentuximab Vedotin and Immune Checkpoint inhibitors) have demonstrated evidence of therapeutic activity and are potential bridge to an allogeneic stem-cell transplantation. The review is aimed to present not only salvage strategies; indeed, the paper contains paragraphs about therapy and new treatment options at diagnosis.
Collapse
Affiliation(s)
- A M Carella
- Division of Hematology and BMT Unit, Ospedale Policlinico San Martino, Genoa, Italy.
| | - P Corradini
- Division of Hematology and BMT Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - A Mussetti
- Division of Hematology and BMT Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - U Ricardi
- Department of Oncology, Università di Torino, Turin, Italy
| | - U Vitolo
- AOU Citta' della Salute e della Scienza, Turin, Italy
| | - S Viviani
- Division of Hematology and BMT Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
563
|
Gökbuget N, Canaani J, Nagler A, Bishop M, Kröger N, Avigan D. Prevention and treatment of relapse after stem cell transplantation with immunotherapy. Bone Marrow Transplant 2018; 53:664-672. [DOI: 10.1038/s41409-018-0232-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/29/2018] [Accepted: 04/16/2018] [Indexed: 12/11/2022]
|
564
|
Sureda A, Dreger P, Bishop MR, Kroger N, Porter DL. Prevention and treatment of relapse after stem cell transplantation in lymphoid malignancies. Bone Marrow Transplant 2018; 54:17-25. [PMID: 29795433 DOI: 10.1038/s41409-018-0214-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 04/05/2018] [Accepted: 04/16/2018] [Indexed: 12/11/2022]
Abstract
Relapse is now the major cause of treatment failure after allogeneic HSCT (alloHSCT). Many novel strategies to address this critical issue are now being developed and tested. At the 3rd International Workshop on Biology, Prevention, and Treatment of Relapse held in Hamburg, Germany in November 2016, international experts presented and discussed recent developments in the field. Some approaches may be applicable to a wide range of patients after transplant, whereas some may be very disease-specific. We present a report from the session dedicated to issues related to prevention and treatment of relapse of lymphoid malignancies after alloHSCT. This session included detailed reviews as well as forward-looking commentaries that focused on Hodgkin lymphoma, chronic lymphocytic leukemia and mantle cell lymphoma, diffuse large cell and follicular lymphoma, and multiple myeloma.
Collapse
Affiliation(s)
- Anna Sureda
- Hematology Department Institut Català d'Oncologia - Hospitale Barcelona, Barcelona, Spain
| | - Peter Dreger
- Department Medicine V, University of Heidelberg, Heidelberg, Germany.,European Society for Blood and Marrow Transplantation (EBMT), Leiden, The Netherlands
| | - Michael R Bishop
- Hematopoietic Cellular Therapy Program Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Nicolaus Kroger
- Department of Stem Cell Transplantation, University Medical Center, Hamburg, Germany
| | - David L Porter
- Division of Hematology Oncology, Blood and Marrow Transplant Program, University of Pennsylvania and Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
565
|
Strati P, Patel S, Nastoupil L, Fanale MA, Bollard CM, Lin AY, Gordon LI. Beyond Chemotherapy: Checkpoint Inhibition and Cell-Based Therapy in Non-Hodgkin Lymphoma. Am Soc Clin Oncol Educ Book 2018; 38:592-603. [PMID: 30231316 DOI: 10.1200/edbk_200549] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Immune-based treatment strategies, such as checkpoint inhibition and chimeric antigen receptor (CAR) T cells, have started a new frontier for treatment in non-Hodgkin lymphoma (NHL). Checkpoint inhibition has been most successful in Hodgkin lymphoma, where higher expression of PD-L1 is correlated with better overall response rate. Combinations of checkpoint inhibition with various chemotherapy or biologics are in clinical trials, with initially promising results and manageable safety profiles. CAR T-cell therapies that target CD19 are a promising and attractive therapy for B-cell NHLs, with a product approved by the US Food and Drug Administration in 2017. Changes in the target, hinge, or costimulatory domain can dramatically alter the persistence and efficacy of the CAR T cells. The ZUMA trials from Kite used CD19-(CD28z) CAR T cells, whereas the TRANSCEND studies from Juno and the JULIET studies from Novartis used CD19-(4-1BBz) CARs. Despite the recent successes with CAR T-cell clinical trials, major concerns associated with this therapy include cytokine release syndrome, potential neurotoxicities, B-cell aplasia, loss of tumor antigen leading to relapse, and cost and accessibility of the treatment. Although first-generation CAR T-cell therapies have failed in solid malignancies, newer second- and third-generation CAR T cells that target antigens other than CD19 (such as mesothelin or B-cell maturation antigen) are being studied in clinical trials for treatment of lung cancer or multiple myeloma. Overall, immune-based treatment strategies have given oncologists and patients hope when there used to be none, as well as a new basket of tools yet to come with further research and development.
Collapse
Affiliation(s)
- Paolo Strati
- From the Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Children's National Health System and The George Washington University, Washington, DC; Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Shabnum Patel
- From the Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Children's National Health System and The George Washington University, Washington, DC; Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Loretta Nastoupil
- From the Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Children's National Health System and The George Washington University, Washington, DC; Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Michelle A Fanale
- From the Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Children's National Health System and The George Washington University, Washington, DC; Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Catherine M Bollard
- From the Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Children's National Health System and The George Washington University, Washington, DC; Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Adam Y Lin
- From the Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Children's National Health System and The George Washington University, Washington, DC; Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Leo I Gordon
- From the Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Children's National Health System and The George Washington University, Washington, DC; Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| |
Collapse
|
566
|
Keeping S, Wu E, Chan K, Mojebi A, Ferrante SA, Balakumaran A. Pembrolizumab versus the standard of care for relapsed and refractory classical Hodgkin's lymphoma progressing after brentuximab vedotin: an indirect treatment comparison. Expert Rev Hematol 2018; 11:503-511. [PMID: 29764245 DOI: 10.1080/17474086.2018.1475226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
BACKGROUND There is significant unmet need among patients with relapsed and refractory classical Hodgkin's lymphoma (RRcHL) who have failed multiple lines of therapy, including brentuximab vedotin (BV). Pembrolizumab, an immune checkpoint inhibitor, is one possible treatment solution for this population. RESEARCH METHODS The objective of this study was to compare progression-free survival (PFS) with standard of care (SOC) versus pembrolizumab in previously BV treated RRcHL patients. A systematic literature review identified one observational study of SOC that was suitable for comparison with KEYNOTE-087, the principal trial of pembrolizumab in this population. Both naïve and population-adjusted (using outcomes regression) pairwise indirect comparisons were conducted. The primary analysis included all patients who had failed BV, with a secondary analysis conducted including only those known to have failed BV that was part of definitive treatment. RESULTS In the primary analysis, SOC was inferior to pembrolizumab in both the unadjusted comparison (HR 5.00 [95% confidence interval (CI) 3.56-7.01]) and the adjusted comparison (HR 6.35 [95% CI 4.04-9.98]). These HRs increased to 5.16 (95% CI 3.61-7.38) and 6.56 (95% CI 4.01-10.72), respectively, in the secondary analysis. CONCLUSION Pembrolizumab offers a significant improvement in PFS compared to SOC in this population.
Collapse
Affiliation(s)
- Sam Keeping
- a Evidence Synthesis and Decision Modeling , Precision Xtract , Vancouver , BC , Canada
| | - Elise Wu
- b Center for Observational & Real World Evidence , Merck & Co, Inc , Kenilworth , NJ , USA
| | - Keith Chan
- a Evidence Synthesis and Decision Modeling , Precision Xtract , Vancouver , BC , Canada
| | - Ali Mojebi
- a Evidence Synthesis and Decision Modeling , Precision Xtract , Vancouver , BC , Canada
| | - Shannon Allen Ferrante
- b Center for Observational & Real World Evidence , Merck & Co, Inc , Kenilworth , NJ , USA
| | - Arun Balakumaran
- b Center for Observational & Real World Evidence , Merck & Co, Inc , Kenilworth , NJ , USA
| |
Collapse
|
567
|
Spinner MA, Advani RH, Connors JM, Azzi J, Diefenbach C. New Treatment Algorithms in Hodgkin Lymphoma: Too Much or Too Little? Am Soc Clin Oncol Educ Book 2018; 38:626-636. [PMID: 30231319 DOI: 10.1200/edbk_200679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Hodgkin lymphoma treatment continues to evolve as new means of assessing response to treatment, new appreciation of important risk factors, and more effective therapeutic agents become available. Treatment algorithms integrating functional imaging now provide the opportunity to modify therapy during its delivery, allowing adjustment of duration and intensity of chemotherapy and rationale identification of patients who may benefit from the addition of therapeutic irradiation. Novel agents, including the antibody drug conjugate brentuximab vedotin and checkpoint inhibitors such as nivolumab and pembrolizumab can improve the effectiveness of treatment while keeping toxicity within acceptable limits. Carefully designed clinical trials permit the identification of superior approaches in which efficacy is enhanced and toxicity minimized. Clinicians treating patients with Hodgkin lymphoma now have access to novel treatment approaches, which will require detailed assessment of each patient and careful discussion of the goals and risks of treatment at the time of planning primary treatment, again during delivery of that treatment as data indicating ongoing effectiveness become available, at the conclusion of initial intervention, and, when the need arises, at the time of recurrence of disease.
Collapse
Affiliation(s)
- Michael A Spinner
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Ranjana H Advani
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Joseph M Connors
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Jacques Azzi
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| | - Catherine Diefenbach
- From the Department of Medicine, Division of Oncology, Stanford University, Stanford, CA; BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada; Perlmutter Cancer Center at NYU Langone Health, New York, NY
| |
Collapse
|
568
|
Meister A, Hentrich M, Wyen C, Hübel K. Malignant lymphoma in the HIV-positive patient. Eur J Haematol 2018; 101:119-126. [PMID: 29663523 DOI: 10.1111/ejh.13082] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2018] [Indexed: 12/27/2022]
Abstract
The introduction of combination antiretroviral therapy (cART) drastically improved performance status, immune function, and life expectancy of HIV-infected individuals. In addition, incidence of opportunistic infections and of AIDS-defining malignancies declined. Nevertheless, aggressive non-Hodgkin's lymphoma still remains the leading cause of AIDS-related deaths. The availability of cART, however, significantly improved the therapeutic options for HIV-positive patients with lymphomas. Diffuse large B-cell lymphoma, Burkitt's lymphoma, or Hodgkin lymphoma has increasingly become curable diseases. In light of these favorable developments in the treatment of HIV and HIV-associated lymphomas, reduction in treatment-associated toxicities and further improvement of outcome of patients with advanced immune suppression are major requirements for future clinical trials. This review summarizes the current treatment landscape and gives an overview on future needs in HIV-positive patients with lymphoma.
Collapse
Affiliation(s)
- Anne Meister
- Department of Medicine I, University Hospital of Cologne, Cologne, Germany
| | - Marcus Hentrich
- Department of Medicine III, Red Cross Hospital Munich, Munich, Germany
| | - Christoph Wyen
- Department of Medicine I, University Hospital of Cologne, Cologne, Germany.,Praxis am Ebertplatz, Cologne, Germany
| | - Kai Hübel
- Department of Medicine I, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
569
|
Peng L, Wu YL. Immunotherapy in the Asiatic population: any differences from Caucasian population? J Thorac Dis 2018; 10:S1482-S1493. [PMID: 29951300 DOI: 10.21037/jtd.2018.05.106] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As "Immunotherapy age" is coming, immune checkpoint inhibitors (CPI) therapies have shown favorable clinical benefits and low toxicity profiles in patients with advanced non-small cell lung cancer (NSCLC). While it is a pity that there is a little clear clinical trials evidence about immunotherapy among Asian population. Moreover, since there is an ethnic difference for targeted therapy, what about immunotherapy? Which factors may associate with ethnic differences from Caucasian population to Asiatic population? In this review, we supposed that the characteristics of the much higher proportion of EGFR mutation, hepatitis B virus infection and unexpected immune-related toxicity among Asian patients should be considered.
Collapse
Affiliation(s)
- Lunxi Peng
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| |
Collapse
|
570
|
Ansell SM. Hodgkin lymphoma: 2018 update on diagnosis, risk-stratification, and management. Am J Hematol 2018; 93:704-715. [PMID: 29634090 DOI: 10.1002/ajh.25071] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 02/20/2018] [Indexed: 12/12/2022]
Abstract
DISEASE OVERVIEW Hodgkin lymphoma (HL) is an uncommon B-cell lymphoid malignancy affecting 8500 new patients annually and representing approximately 10.2% of all lymphomas in the United States. DIAGNOSIS HL is composed of two distinct disease entities: classical HL and nodular lymphocyte predominant HL. Nodular sclerosis, mixed cellularity, lymphocyte depletion, and lymphocyte-rich HL are subgroups of classical HL. RISK STRATIFICATION An accurate assessment of the stage of disease in patients with HL is critical for the selection of the appropriate therapy. Prognostic models that identify patients at low or high risk for recurrence, as well as the response to therapy as determined by positron emission tomography scan, are used to optimize therapy. RISK-ADAPTED THERAPY Initial therapy for HL patients is based on the histology of the disease, the anatomical stage and the presence of poor prognostic features. Patients with early stage disease are typically treated with combined modality strategies utilizing abbreviated courses of combination chemotherapy followed by involved-field radiation therapy, while those with advanced stage disease receive a longer course of chemotherapy often without radiation therapy. Newer agents including brentuximab vedotin are now being incorporated into frontline therapy and these new combinations are becoming a standard of care. MANAGEMENT OF RELAPSED/REFRACTORY DISEASE High-dose chemotherapy (HDCT) followed by an autologous stem cell transplant (ASCT) is the standard of care for most patients who relapse following initial therapy. For patients who fail HDCT with ASCT, brentuximab vedotin, PD-1 blockade, nonmyeloablative allogeneic transplant or participation in a clinical trial should be considered.
Collapse
|
571
|
Wang Y, Nowakowski GS, Wang ML, Ansell SM. Advances in CD30- and PD-1-targeted therapies for classical Hodgkin lymphoma. J Hematol Oncol 2018; 11:57. [PMID: 29685160 PMCID: PMC5914042 DOI: 10.1186/s13045-018-0601-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/13/2018] [Indexed: 11/10/2022] Open
Abstract
CD30 and programmed cell death protein 1 (PD-1) are two ideal therapeutic targets in classical Hodgkin lymphoma (cHL). The CD30 antibody-drug conjugate (ADC) brentuximab vedotin and the PD-1 antibodies nivolumab and pembrolizumab are highly efficacious in treating relapsed and/or refractory cHL. Ongoing studies are evaluating their efficacy in earlier lines of therapy and have demonstrated encouraging results. These agents are expected to further change the landscape of cHL management. Increased cure rates and reduced long-term toxicity from traditional chemotherapy and radiotherapy are likely with the emergence of these novel targeted therapies.
Collapse
Affiliation(s)
- Yucai Wang
- Division of Hematology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | | | - Michael L Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen M Ansell
- Division of Hematology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
572
|
Wang GX, Kurra V, Gainor JF, Sullivan RJ, Flaherty KT, Lee SI, Fintelmann FJ. Immune Checkpoint Inhibitor Cancer Therapy: Spectrum of Imaging Findings. Radiographics 2018; 37:2132-2144. [PMID: 29131763 DOI: 10.1148/rg.2017170085] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Immune checkpoint inhibitors are a new class of cancer therapeutics that have demonstrated striking successes in a rapid series of clinical trials. Consequently, these drugs have dramatically increased in clinical use since being first approved for advanced melanoma in 2011. Current indications in addition to melanoma are non-small cell lung cancer, head and neck squamous cell carcinoma, renal cell carcinoma, urothelial carcinoma, and classical Hodgkin lymphoma. A small subset of patients treated with immune checkpoint inhibitors undergoes an atypical treatment response pattern termed pseudoprogression: New or enlarging lesions appear after initiation of therapy, thereby mimicking tumor progression, followed by an eventual decrease in total tumor burden. Traditional response standards applied at the time of initial increase in tumor burden can falsely designate this as treatment failure and could lead to inappropriate termination of therapy. Currently, when new or enlarging lesions are observed with immune checkpoint inhibitors, only follow-up imaging can help distinguish patients with pseudoprogression from the large majority in whom this observation represents true treatment failure. Furthermore, the unique mechanism of immune checkpoint inhibitors can cause a distinct set of adverse events related to autoimmunity, which can be severe or life threatening. Given the central role of imaging in cancer care, radiologists must be knowledgeable about immune checkpoint inhibitors to correctly assess treatment response and expeditiously diagnose treatment-related complications. The authors review the molecular mechanisms and clinical applications of immune checkpoint inhibitors, the current strategy to distinguish pseudoprogression from progression, and the imaging appearances of common immune-related adverse events. ©RSNA, 2017.
Collapse
Affiliation(s)
- Gary X Wang
- From the Departments of Radiology (G.X.W., V.K., S.I.L., F.J.F.) and Medicine (J.F.G., R.J.S., K.T.F.), Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Founders 210, Boston, MA 02114
| | - Vikram Kurra
- From the Departments of Radiology (G.X.W., V.K., S.I.L., F.J.F.) and Medicine (J.F.G., R.J.S., K.T.F.), Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Founders 210, Boston, MA 02114
| | - Justin F Gainor
- From the Departments of Radiology (G.X.W., V.K., S.I.L., F.J.F.) and Medicine (J.F.G., R.J.S., K.T.F.), Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Founders 210, Boston, MA 02114
| | - Ryan J Sullivan
- From the Departments of Radiology (G.X.W., V.K., S.I.L., F.J.F.) and Medicine (J.F.G., R.J.S., K.T.F.), Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Founders 210, Boston, MA 02114
| | - Keith T Flaherty
- From the Departments of Radiology (G.X.W., V.K., S.I.L., F.J.F.) and Medicine (J.F.G., R.J.S., K.T.F.), Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Founders 210, Boston, MA 02114
| | - Susanna I Lee
- From the Departments of Radiology (G.X.W., V.K., S.I.L., F.J.F.) and Medicine (J.F.G., R.J.S., K.T.F.), Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Founders 210, Boston, MA 02114
| | - Florian J Fintelmann
- From the Departments of Radiology (G.X.W., V.K., S.I.L., F.J.F.) and Medicine (J.F.G., R.J.S., K.T.F.), Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Founders 210, Boston, MA 02114
| |
Collapse
|
573
|
Vari F, Arpon D, Keane C, Hertzberg MS, Talaulikar D, Jain S, Cui Q, Han E, Tobin J, Bird R, Cross D, Hernandez A, Gould C, Birch S, Gandhi MK. Immune evasion via PD-1/PD-L1 on NK cells and monocyte/macrophages is more prominent in Hodgkin lymphoma than DLBCL. Blood 2018; 131:1809-1819. [PMID: 29449276 PMCID: PMC5922274 DOI: 10.1182/blood-2017-07-796342] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 02/09/2018] [Indexed: 12/21/2022] Open
Abstract
Much focus has been on the interaction of programmed cell death ligand 1 (PD-L1) on malignant B cells with programmed cell death 1 (PD-1) on effector T cells in inhibiting antilymphoma immunity. We sought to establish the contribution of natural killer (NK) cells and inhibitory CD163+ monocytes/macrophages in Hodgkin lymphoma (cHL) and diffuse large B-cell lymphoma (DLBCL). Levels of PD-1 on NK cells were elevated in cHL relative to DLBCL. Notably, CD3-CD56hiCD16-ve NK cells had substantially higher PD-1 expression relative to CD3-CD56dimCD16+ cells and were expanded in blood and tissue, more marked in patients with cHL than patients with DLBCL. There was also a raised population of PD-L1-expressing CD163+ monocytes that was more marked in patients with cHL compared with patients with DLBCL. The phenotype of NK cells and monocytes reverted back to normal once therapy (ABVD [doxorubicin 25 mg/m2, bleomycin 10 000 IU/m2, vinblastine 6 mg/m2, dacarbazine 375 mg/m2, all given days 1 and 15, repeated every 28 days] or R-CHOP [rituximab 375 mg/m2, cyclophosphamide 750 mg/m2 IV, doxorubicin 50 mg/m2 IV, vincristine 1.4 mg/m2 (2 mg maximum) IV, prednisone 100 mg/day by mouth days 1-5, pegfilgrastim 6 mg subcutaneously day 4, on a 14-day cycle]) had commenced. Tumor-associated macrophages (TAMs) expressed high levels of PD-L1/PD-L2 within diseased lymph nodes. Consistent with this, CD163/PD-L1/PD-L2 gene expression was also elevated in cHL relative to DLBCL tissues. An in vitro functional model of TAM-like monocytes suppressed activation of PD-1hi NK cells, which was reversed by PD-1 blockade. In line with these findings, depletion of circulating monocytes from the blood of pretherapy patients with cHL and patients with DLBCL enhanced CD3-CD56hiCD16-ve NK-cell activation. We describe a hitherto unrecognized immune evasion strategy mediated via skewing toward an exhausted PD-1-enriched CD3-CD56hiCD16-ve NK-cell phenotype. In addition to direct inhibition of NK cells by the malignant B cell, suppression of NK cells can occur indirectly by PD-L1/PD-L2-expressing TAMs. The mechanism is more prominent in cHL than DLBCL, which may contribute to the clinical sensitivity of cHL to PD-1 blockade.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal, Murine-Derived/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- B7-H1 Antigen/immunology
- Bleomycin/administration & dosage
- Cyclophosphamide/administration & dosage
- Dacarbazine/administration & dosage
- Doxorubicin/administration & dosage
- Female
- Hodgkin Disease/drug therapy
- Hodgkin Disease/immunology
- Hodgkin Disease/pathology
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Macrophages/immunology
- Macrophages/pathology
- Male
- Models, Immunological
- Monocytes/immunology
- Monocytes/pathology
- Neoplasm Proteins/immunology
- Prednisone/administration & dosage
- Programmed Cell Death 1 Receptor/immunology
- Rituximab
- Tumor Escape
- Vinblastine/administration & dosage
- Vincristine/administration & dosage
Collapse
Affiliation(s)
- Frank Vari
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - David Arpon
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Colm Keane
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | | | - Dipti Talaulikar
- Canberra Hospital, Canberra, ACT, Australia
- Australian National University Medical School, Acton, ACT, Australia; and
| | | | - Qingyan Cui
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Erica Han
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Josh Tobin
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Robert Bird
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Donna Cross
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Annette Hernandez
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Clare Gould
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Simone Birch
- Department of Pathology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Maher K Gandhi
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| |
Collapse
|
574
|
Watanabe T, Okuda K, Murase T, Moriyama S, Haneda H, Kawano O, Yokota K, Sakane T, Oda R, Inagaki H, Nakanishi R. Four immunohistochemical assays to measure the PD-L1 expression in malignant pleural mesothelioma. Oncotarget 2018; 9:20769-20780. [PMID: 29755688 PMCID: PMC5945532 DOI: 10.18632/oncotarget.25100] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/24/2018] [Indexed: 12/29/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 pathway are expected to be a novel therapy for combating future increases in numbers of malignant pleural mesothelioma (MPM) patients. However, the PD-L1 expression, which is a predictor of the response to ICIs, is unclear in MPM. We studied the PD-L1 expression using four immunohistochemical assays (SP142, SP263, 28-8 and 22C3) in 32 MPM patients. The PD-L1 expression in tumor cells and immune cells was evaluated to clarify the rate of PD-L1 expression and the concordance among the four assays in MPM. The positivity rate of PD-L1 expression was 53.1% for SP142, 28.1% for SP263, 53.1% for 28-8, and 56.3% for 22C3. Nine cases were positive and 10 were negative for all assays. Discordance among the four assays was found in 13 cases. The concordance rates between SP142 and 22C3 and between 28-8 and 22C3 were the highest (84.4%). The concordance rates between SP263 and the other three assays were low (71.9% to 75.0%). The PD-L1 expression in MPM was almost equivalent for three of the assays. Given the cut-off values set in our study, these findings suggested that these assays, except for SP263, can be used for accurate PD-L1 immunostaining in MPM.
Collapse
Affiliation(s)
- Takuya Watanabe
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Katsuhiro Okuda
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Takayuki Murase
- Department of Pathology and Molecular Diagnostics, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Satoru Moriyama
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiroshi Haneda
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Osamu Kawano
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Keisuke Yokota
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Tadashi Sakane
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Risa Oda
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiroshi Inagaki
- Department of Pathology and Molecular Diagnostics, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Ryoichi Nakanishi
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| |
Collapse
|
575
|
Sequential immunotherapy in a patient with primary refractory Hodgkin lymphoma and novel mutations. Oncotarget 2018; 9:20928-20940. [PMID: 29755699 PMCID: PMC5945535 DOI: 10.18632/oncotarget.25037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 03/15/2018] [Indexed: 12/16/2022] Open
Abstract
Primary resistant Hodgkin lymphoma is an aggressive disease with few treatment options and short survival. Neoplastic cells of classical Hodgkin lymphoma are heavily dependent on microenvironmental stimuli, regularly express PD-L1, and a relevant proportion of relapsed patients is sensitive to blocking of the PD1/PD-L1 axis. However, response duration is limited and further treatment options are unknown but urgently needed. We report a case of a patient without relevant response to five subsequent chemotherapy regimens who immediately and dramatically responded to an anti-PD1 mab. During the following two years she responded to the anti-CTLA-4 mab ipilimumab, the Jak2 inhibitor ruxolitinib, and a combination of lenalidomide plus cyclophosphamide given in subsequent relapses. A thorough genomic analysis demonstrated seven genomic alterations with six of them not previously described in this disease (i.e. BRIP1 G212fs*62, KRAS L19F, KDM5A R1239W, MYC A59T, ARIDA1A E1683fs*15 and TP53 277Y). Three alterations were considered actionable and one of them drugable. The number of mutations increased over time and the BRIP1 mutation was found to be a germline mutation.
Collapse
|
576
|
Balancing risk and benefit in early-stage classical Hodgkin lymphoma. Blood 2018; 131:1666-1678. [DOI: 10.1182/blood-2017-10-772665] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 02/07/2018] [Indexed: 01/23/2023] Open
Abstract
Abstract
With defined chemotherapy and radiotherapy (RT) and risk-adapted treatment, early-stage classical Hodgkin lymphoma (HL) has become curable in a majority of patients. Hence, a major current goal is to reduce treatment-related toxicity while maintaining long-term disease control. Patients with early-stage favorable disease (ie, limited stage without risk factors [RFs]) are frequently treated with 2 cycles of doxorubicin, bleomycin, vinblastine, and dacarbazine (2×ABVD) followed by 20-Gy involved-field or involved-site RT (IF/ISRT). In patients with early-stage unfavorable disease (ie, limited stage with RFs), 4 cycles of chemotherapy are usually consolidated with 30-Gy IF/ISRT. Compared with 4×ABVD, 2 cycles of bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone (2×BEACOPPescalated) followed by 2×ABVD improved 5-year progression-free survival (PFS), with similar 5-year overall survival. Recently, treatment strategies based on [18F]fluorodeoxyglucose positron emission tomography (PET) response were evaluated. In early-stage unfavorable HL, a majority of patients achieved a negative interim PET after 2×ABVD and an excellent outcome after 4×ABVD, whereas in those with a positive interim PET, 2×BEACOPPescalated improved 5-year PFS. Furthermore, a PET-guided RT approach was evaluated to decrease long-term toxicity. Although both the RAPID and H10 trials reported poorer disease control without RT, PET-guided omission of RT can constitute a valid therapeutic option in patients with an increased risk of RT-associated toxicity (eg, because of sex, age, or disease localization). Implementation of drugs such as the anti-CD30 antibody-drug conjugate brentuximab vedotin or the anti–programmed death 1 antibodies nivolumab or pembrolizumab might allow further reduction of overall mortality and improve quality of life in affected patients.
Collapse
|
577
|
Mehta-Shah N, Bartlett NL. Management of relapsed/refractory classical Hodgkin lymphoma in transplant-ineligible patients. Blood 2018; 131:1698-1703. [PMID: 29500171 PMCID: PMC6536701 DOI: 10.1182/blood-2017-09-772681] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/07/2017] [Indexed: 11/20/2022] Open
Abstract
Addition of brentuximab vedotin, a CD30-targeted antibody-drug conjugate, and the programmed death 1 (PD-1) inhibitors nivolumab and pembrolizumab to the armamentarium for transplant-ineligible relapsed/refractory classical Hodgkin lymphoma has resulted in improved outcomes, including the potential for cure in a small minority of patients. For patients who have failed prior transplant or are unsuitable for dose-intense approaches based on age or comorbidities, an individualized approach with sequential use of single agents such as brentuximab vedotin, PD-1 inhibitors, everolimus, lenalidomide, or conventional agents such as gemcitabine or vinorelbine may result in prolonged survival with a minimal or modest effect on quality of life. Participation in clinical trials evaluating new approaches such as combination immune checkpoint inhibition, novel antibody-drug conjugates, or cellular therapies such as Epstein-Barr virus-directed cytotoxic T lymphocytes and chimeric antigen receptor T cells offer additional options for eligible patients.
Collapse
Affiliation(s)
- Neha Mehta-Shah
- Division of Medical Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO
| | - Nancy L Bartlett
- Division of Medical Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
578
|
Lim SH, Johnson PWM. Optimizing therapy in advanced-stage Hodgkin lymphoma. Blood 2018; 131:1679-1688. [PMID: 29500173 DOI: 10.1182/blood-2017-09-772640] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/25/2017] [Indexed: 02/07/2023] Open
Abstract
The treatment of Hodgkin lymphoma has evolved continuously since the introduction of extended-field radiotherapy in the 1960s to involved-field and then involved-node radiotherapy, multiagent chemotherapy, combined chemoradiotherapy, risk-adapted and response-adapted modulation, and, most recently, introduction of antibody-drug conjugates and immune checkpoint-blocking antibodies. These changes have translated into progressively increasing cure rates, so that 10-year survival figures now exceed 80%, compared with <50% 40 years ago. The challenge now is how to improve upon success while maintaining, or if possible improving, the quality of life for survivors. Steering between undertreatment, with the risk of avoidable recurrences, and overtreatment, with the risk of unnecessary toxicity, remains complex because control of the lymphoma and the probability of survival are no longer closely linked. This requires trials with long follow-up and continuous reappraisal of the interaction between the illness; the method used to define risk, and the type of treatment involved. One important factor in this is age: outcomes in older patients have not improved at the same rate as those in the population under 60 years of age, reflecting the need for different approaches. Recently, treatment has moved from being primarily risk-based, using baseline characteristics such as anatomical stage and severity of the illness, to a more dynamic approach that takes account of the response to therapy, using functional imaging to make an early appraisal, with the option to modulate subsequent treatment. The results of several trials indicate that this has advantages, but a combination of risk- and response-adaptation is probably ideal.
Collapse
Affiliation(s)
- Sean H Lim
- Antibody and Vaccine Group and
- Cancer Sciences Unit, Cancer Research UK Centre, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| | - Peter W M Johnson
- Cancer Sciences Unit, Cancer Research UK Centre, Southampton General Hospital, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
579
|
Witkowska M, Smolewski P. Immune Checkpoint Inhibitors to Treat Malignant Lymphomas. J Immunol Res 2018; 2018:1982423. [PMID: 29850620 PMCID: PMC5925139 DOI: 10.1155/2018/1982423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 02/05/2018] [Indexed: 12/31/2022] Open
Abstract
Genetic and/or epigenetic changes provide antigen-derived diversity in neoplastic cells. Beside, these cells do not initiate immune response of host organisms. A variety of factors are responsible for the resistant to treatment, including individual variations in patients and somatic cell genetic differences in tumors, even those from the same tissue of origin. Immune system is controlled by several controlling mechanisms. Recently, a significant progress in hematologic treatment has been made; however, majority of diseases still remain incurable. Immunotherapy with checkpoint inhibitors has emerged as promising modality of antitumor treatment, showing marked response to several antigens, including cytotoxic T lymphocyte-associate protein-4 (CTLA-4) or programmed cell death 1 receptor (PD-1). In this review, we demonstrate actual knowledge on immune checkpoint function and its impact on development of new modality of antineoplastic treatment, using, for example, anti-CTLA-4 or PD-1/PD1 ligand (PD-L1) monoclonal antibodies in malignant lymphomas.
Collapse
Affiliation(s)
- Magdalena Witkowska
- Department of Experimental Hematology, Medical University of Lodz, Lodz, Poland
| | - Piotr Smolewski
- Department of Experimental Hematology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
580
|
Bollard CM, Tripic T, Cruz CR, Dotti G, Gottschalk S, Torrano V, Dakhova O, Carrum G, Ramos CA, Liu H, Wu MF, Marcogliese AN, Barese C, Zu Y, Lee DY, O’Connor O, Gee AP, Brenner MK, Heslop HE, Rooney CM. Tumor-Specific T-Cells Engineered to Overcome Tumor Immune Evasion Induce Clinical Responses in Patients With Relapsed Hodgkin Lymphoma. J Clin Oncol 2018; 36:1128-1139. [PMID: 29315015 PMCID: PMC5891126 DOI: 10.1200/jco.2017.74.3179] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Purpose Transforming growth factor-β (TGF-β) production in the tumor microenvironment is a potent and ubiquitous tumor immune evasion mechanism that inhibits the expansion and function of tumor-directed responses; therefore, we conducted a clinical study to discover the effects of the forced expression of a dominant-negative TGF-β receptor type 2 (DNRII) on the safety, survival, and activity of infused tumor-directed T cells. Materials and Methods In a dose escalation study, eight patients with Epstein Barr virus-positive Hodgkin lymphoma received two to 12 doses of between 2 × 107 and 1.5 × 108 cells/m2 of DNRII-expressing T cells with specificity for the Epstein Barr virus-derived tumor antigens, latent membrane protein (LMP)-1 and LMP-2 (DNRII-LSTs). Lymphodepleting chemotherapy was not used before infusion. Results DNRII-LSTs were resistant to otherwise inhibitory concentrations of TGF-β in vitro and retained their tumor antigen-specific activity. After infusion, the signal from transgenic T cells in peripheral blood increased up to 100-fold as measured by quantitative polymerase chain reaction for the transgene, with a corresponding increase in the frequency of functional LMP-specific T cells. Expansion was not associated with any acute or long-term toxicity. DNRII-LSTs persisted for up to ≥ 4 years. Four of the seven evaluable patients with active disease achieved clinical responses that were complete and ongoing in two patients at > 4 years, including in one patient who achieved only a partial response to unmodified tumor-directed T cells. Conclusion TGF-β-resistant tumor-specific T cells safely expand and persist in patients with Hodgkin lymphoma without lymphodepleting chemotherapy before infusion. DNRII-LSTs can induce complete responses even in patients with resistant disease. Expression of DNRII may be useful for the many other tumors that exploit this potent immune evasion mechanism.
Collapse
Affiliation(s)
- Catherine M. Bollard
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Tamara Tripic
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Conrad Russell Cruz
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Gianpietro Dotti
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Stephen Gottschalk
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Vicky Torrano
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Olga Dakhova
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - George Carrum
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Carlos A. Ramos
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Hao Liu
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Meng-Fen Wu
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Andrea N. Marcogliese
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Cecilia Barese
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Youli Zu
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Daniel Y. Lee
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Owen O’Connor
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Adrian P. Gee
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Malcolm K. Brenner
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Helen E. Heslop
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| | - Cliona M. Rooney
- Catherine M. Bollard, Tamara Tripic, Gianpietro Dotti, Stephen Gottschalk, Vicky Torrano, Olga Dakhova, George Carrum, Carlos A. Ramos, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Houston Methodist Hospital, and Texas Children's Hospital; Catherine M. Bollard, Gianpietro Dotti, Stephen Gottschalk, George Carrum, Carlos A. Ramos, Hao Liu, Meng-Fen Wu, Andrea N. Marcogliese, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine, Houston, TX; Catherine M. Bollard, Conrad Russell Cruz, and Cecilia Barese, Children’s National Health System, Washington, DC; Youli Zu and Daniel Y. Lee, Weill Medical College of Cornell University; and Owen O’Connor, Columbia University College of Physicians and Surgeons, The New York Presbyterian Hospital, New York, NY
| |
Collapse
|
581
|
Omuro A, Vlahovic G, Lim M, Sahebjam S, Baehring J, Cloughesy T, Voloschin A, Ramkissoon SH, Ligon KL, Latek R, Zwirtes R, Strauss L, Paliwal P, Harbison CT, Reardon DA, Sampson JH. Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neuro Oncol 2018; 20:674-686. [PMID: 29106665 PMCID: PMC5892140 DOI: 10.1093/neuonc/nox208] [Citation(s) in RCA: 360] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Immunotherapies have demonstrated efficacy across a diverse set of tumors supporting further evaluation in glioblastoma. The objective of this study was to evaluate the safety/tolerability and describe immune-mediated effects of nivolumab ± ipilimumab in patients with recurrent glioblastoma. Exploratory efficacy outcomes are also reported. Methods Patients were randomized to receive nivolumab 3 mg/kg every 2 weeks (Q2W; NIVO3) or nivolumab 1 mg/kg + ipilimumab 3 mg/kg every 3 weeks (Q3W) for 4 doses, then nivolumab 3 mg/kg Q2W (NIVO1+IPI3). An alternative regimen of nivolumab 3 mg/kg + ipilimumab 1 mg/kg Q3W for 4 doses, then nivolumab 3 mg/kg Q2W (NIVO3+IPI1) was investigated in a nonrandomized arm. Results Forty patients were enrolled (NIVO3, n = 10; NIVO1+IPI3, n = 10; NIVO3+IPI1, n = 20). The most common treatment-related adverse events (AEs) were fatigue (NIVO3, 30%; NIVO1+IPI3, 80%; NIVO3+IPI1, 55%) and diarrhea (10%, 70%, 30%, respectively). AEs leading to discontinuation occurred in 10% (NIVO3), 30% (NIVO1+IPI3), and 20% (NIVO3+IPI1) of patients. Three patients achieved a partial response (NIVO3, n = 1; NIVO3+IPI1, n = 2) and 8 had stable disease for ≥12 weeks (NIVO3, n = 2; NIVO1+IPI3, n = 2; NIVO3+IPI1, n = 4 [Response Assessment in Neuro-Oncology criteria]). Most patients (68%) had tumor-cell programmed death ligand-1 expression ≥1%. Immune-mediated effects mimicking radiographic progression occurred in 2 patients. Conclusions Nivolumab monotherapy was better tolerated than nivolumab + ipilimumab; the tolerability of the combination was influenced by ipilimumab dose. These safety and exploratory findings merit further investigation of immunotherapies in glioblastoma.
Collapse
Affiliation(s)
- Antonio Omuro
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Michael Lim
- The Johns Hopkins Hospital, Baltimore, Maryland
| | - Solmaz Sahebjam
- Moffitt Cancer Center, University of South Florida, Tampa, Florida
| | | | | | | | - Shakti H Ramkissoon
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Keith L Ligon
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | - David A Reardon
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
582
|
Gastrointestinal toxicity of immune checkpoint inhibitors: from mechanisms to management. Nat Rev Gastroenterol Hepatol 2018; 15:222-234. [PMID: 29512649 DOI: 10.1038/nrgastro.2018.14] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitor therapies are a novel group of monoclonal antibodies with proven effectiveness in a wide range of malignancies, including melanoma, renal cell carcinoma, non-small-cell lung cancer, urothelial carcinoma and Hodgkin lymphoma. Their use in a range of other indications, such as gastrointestinal and head and neck cancer, is currently under investigation. The number of agents included in this drug group is increasing, as is their use. Although they have the potential to improve the treatment of advanced malignancies, they are also associated with a substantial risk of immune-related adverse events. The incidence of gastrointestinal toxicity associated with their use is second only in frequency to dermatological toxicity. Thus, gastroenterologists can expect to be increasingly frequently consulted by oncologists as part of a multidisciplinary approach to managing toxicity. Here, we describe this novel group of agents and their mechanisms of action. We review the manifestations of gastrointestinal toxicity associated with their use so that it can be recognized early and diagnosed accurately. We also discuss the proposed mechanisms underlying this toxicity and describe an algorithmic and, wherever possible, evidence-based approach to its management.
Collapse
|
583
|
Shindiapina P, Alinari L. Pembrolizumab and its role in relapsed/refractory classical Hodgkin's lymphoma: evidence to date and clinical utility. Ther Adv Hematol 2018; 9:89-105. [PMID: 29623180 PMCID: PMC5881987 DOI: 10.1177/2040620718761777] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/06/2018] [Indexed: 12/28/2022] Open
Abstract
Immune evasion is a critical mechanism of malignant cell survival, and relies in part on molecular signaling through the programmed cell death 1 (PD-1)/PD-1 ligand (PD-L1) axis that contributes to T cell exhaustion. Immune modulatory therapy with monoclonal antibodies against PD-1 designed to enhance antitumor immune response have shown promise in the treatment of advanced solid tumors and hematologic malignancies. Classical Hodgkin's lymphoma (cHL), a unique B cell malignancy characterized by an extensive but ineffective immune cell infiltrate surrounding a small number of tumor cells, has shown significant response to anti-PD-1 directed therapy. The anti-PD-1 monoclonal antibodies nivolumab and pembrolizumab have shown similarly remarkable activity in relapsed/refractory cHL and have been approved by the Food and Drug Administration for treatment of this disease. In this article we review the rationale of targeting the PD-1/PD-L1 axis in cHL and the pharmacology of pembrolizumab, and summarize the data on activity and safety profile of this agent in the treatment of relapsed/refractory cHL. We also discuss the potential benefits and pitfalls of using PD-1 blockade in the setting of allogeneic stem-cell transplantation, and summarize ongoing prospective trials of single-agent pembrolizumab and combination strategies as well as future directions.
Collapse
Affiliation(s)
- Polina Shindiapina
- Department of Internal Medicine, Division of Hematology, Arthur G James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lapo Alinari
- Division of Hematology, Department of Internal Medicine, Ohio State University Wexner Medical Center, 410 West 12th Avenue, 481A Wiseman Hall, Columbus, Ohio, 43210, USA
| |
Collapse
|
584
|
Jakobsen NA, Vyas P. From genomics to targeted treatment in haematological malignancies: a focus on acute myeloid leukaemia. Clin Med (Lond) 2018; 18. [PMID: 29700093 PMCID: PMC6334029 DOI: 10.7861/clinmedicine.18-2s-s47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The haematological malignancies are a heterogeneous group of neoplastic disorders, which lead to almost 10,000 deaths annually in the UK. Over the past 2 decades, there has been significant progress in our understanding of the pathological mechanisms underlying these cancers, accompanied by improvements in outcomes for some patients. In particular, advances in next-generation sequencing now make it possible to define the genetic lesions present in each patient, which has led to improved disease classification, risk stratification and identification of new therapeutic targets. Here we discuss recent advances in the genomic classification and targeted treatment of haematological malignancies, focusing on acute myeloid leukaemia. Multiple novel drug classes are now on the horizon, including agents that target overactive signalling pathways, differentiation therapies and immunotherapies. By combining molecular diagnostics with targeted therapy, the management of these diseases is set to change radically over the coming years.
Collapse
Affiliation(s)
- Niels Asger Jakobsen
- AWeatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Paresh Vyas
- BMRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK and Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK,Address for correspondence: Professor Paresh Vyas, MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK.
| |
Collapse
|
585
|
Jakobsen NA, Vyas P. From genomics to targeted treatment in haematological malignancies: a focus on acute myeloid leukaemia. Clin Med (Lond) 2018; 18:s47-s53. [PMID: 29700093 PMCID: PMC6334029 DOI: 10.7861/clinmedicine.18-2-s47] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The haematological malignancies are a heterogeneous group of neoplastic disorders, which lead to almost 10,000 deaths annually in the UK. Over the past 2 decades, there has been significant progress in our understanding of the pathological mechanisms underlying these cancers, accompanied by improvements in outcomes for some patients. In particular, advances in next-generation sequencing now make it possible to define the genetic lesions present in each patient, which has led to improved disease classification, risk stratification and identification of new therapeutic targets. Here we discuss recent advances in the genomic classification and targeted treatment of haematological malignancies, focusing on acute myeloid leukaemia. Multiple novel drug classes are now on the horizon, including agents that target overactive signalling pathways, differentiation therapies and immunotherapies. By combining molecular diagnostics with targeted therapy, the management of these diseases is set to change radically over the coming years.
Collapse
Affiliation(s)
- Niels Asger Jakobsen
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Paresh Vyas
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK and Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
586
|
Bair SM, Mato A, Svoboda J. Immunotherapy for the Treatment of Hodgkin Lymphoma: An Evolving Paradigm. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:380-391. [PMID: 29685424 DOI: 10.1016/j.clml.2018.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/18/2018] [Accepted: 03/27/2018] [Indexed: 12/09/2022]
Abstract
Classical Hodgkin lymphoma (cHL) is one of the most common lymphomas in the Western world. Although most patients are cured with standard first-line therapy, up to 20% of patients will have relapsed or refractory disease. Although the conventional approach to treatment has consisted of chemotherapy, radiation, and for those who relapse, autologous or allogeneic transplantation, newer approaches have become available in recent years, including immunoconjugates and checkpoint inhibitors. These approaches have shown significant efficacy in clinical trials and might be associated with fewer long-term toxicities compared with conventional therapies. In this review we discuss the biology of cHL as it pertains to the immunosuppressive tumor microenvironment and then review the existing clinical trial results of several emerging immunotherapies in this context, including immune checkpoint inhibitors and adoptive cellular therapy. Finally, several clinical practice issues pertaining to the use of immunotherapies are discussed.
Collapse
Affiliation(s)
- Steven M Bair
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Anthony Mato
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jakub Svoboda
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
587
|
Armand P, Engert A, Younes A, Fanale M, Santoro A, Zinzani PL, Timmerman JM, Collins GP, Ramchandren R, Cohen JB, De Boer JP, Kuruvilla J, Savage KJ, Trneny M, Shipp MA, Kato K, Sumbul A, Farsaci B, Ansell SM. Nivolumab for Relapsed/Refractory Classic Hodgkin Lymphoma After Failure of Autologous Hematopoietic Cell Transplantation: Extended Follow-Up of the Multicohort Single-Arm Phase II CheckMate 205 Trial. J Clin Oncol 2018; 36:1428-1439. [PMID: 29584546 PMCID: PMC6075855 DOI: 10.1200/jco.2017.76.0793] [Citation(s) in RCA: 519] [Impact Index Per Article: 74.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose Genetic alterations causing overexpression of programmed death-1 ligands are near universal in classic Hodgkin lymphoma (cHL). Nivolumab, a programmed death-1 checkpoint inhibitor, demonstrated efficacy in relapsed/refractory cHL after autologous hematopoietic cell transplantation (auto-HCT) in initial analyses of one of three cohorts from the CheckMate 205 study of nivolumab for cHL. Here, we assess safety and efficacy after extended follow-up of all three cohorts. Methods This multicenter, single-arm, phase II study enrolled patients with relapsed/refractory cHL after auto-HCT treatment failure into cohorts by treatment history: brentuximab vedotin (BV)–naïve (cohort A), BV received after auto-HCT (cohort B), and BV received before and/or after auto-HCT (cohort C). All patients received nivolumab 3 mg/kg every 2 weeks until disease progression/unacceptable toxicity. The primary end point was objective response rate per independent radiology review committee. Results Overall, 243 patients were treated; 63 in cohort A, 80 in cohort B, and 100 in cohort C. After a median follow-up of 18 months, 40% continued to receive treatment. The objective response rate was 69% (95% CI, 63% to 75%) overall and 65% to 73% in each cohort. Overall, the median duration of response was 16.6 months (95% CI, 13.2 to 20.3 months), and median progression-free survival was 14.7 months (95% CI, 11.3 to 18.5 months). Of 70 patients treated past conventional disease progression, 61% of those evaluable had stable or further reduced target tumor burdens. The most common grade 3 to 4 drug-related adverse events were lipase increases (5%), neutropenia (3%), and ALT increases (3%). Twenty-nine deaths occurred; none were considered treatment related. Conclusion With extended follow-up, responses to nivolumab were frequent and durable. Nivolumab seems to be associated with a favorable safety profile and long-term benefits across a broad spectrum of patients with relapsed/refractory cHL.
Collapse
Affiliation(s)
- Philippe Armand
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Andreas Engert
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Anas Younes
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Michelle Fanale
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Armando Santoro
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Pier Luigi Zinzani
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - John M Timmerman
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Graham P Collins
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Radhakrishnan Ramchandren
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Jonathon B Cohen
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Jan Paul De Boer
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - John Kuruvilla
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Kerry J Savage
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Marek Trneny
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Margaret A Shipp
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Kazunobu Kato
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Anne Sumbul
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Benedetto Farsaci
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| | - Stephen M Ansell
- Philippe Armand and Margaret A. Shipp, Dana-Farber Cancer Institute, Boston, MA; Andreas Engert, University Hospital of Cologne, Cologne, Germany; Anas Younes, Memorial Sloan Kettering Cancer Center, New York, NY; Michelle Fanale, University of Texas MD Anderson Cancer Center, Houston, TX; Armando Santoro, Humanitas Cancer Center, Humanitas University, Milan; Pier Luigi Zinzani, Institute of Hematology "L. e A. Seràgnoli," University of Bologna, Bologna, Italy; John M. Timmerman, University of California Los Angeles Medical Center, Los Angeles, CA; Graham P. Collins, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, United Kingdom; Radhakrishnan Ramchandren, Barbara Ann Karmanos Cancer Institute, Detroit, MI; Jonathon B. Cohen, Winship Cancer Institute, Emory University, Atlanta, GA; Jan Paul De Boer, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands, on behalf of Lunenburg Lymphoma Phase I/II Consortium; John Kuruvilla, University of Toronto and Princess Margaret Cancer Centre, Toronto, Ontario; Kerry J. Savage, BC Cancer Agency, Vancouver, British Columbia, Canada; Marek Trneny, Charles University, General Hospital in Prague, Prague, Czech Republic; Kazunobu Kato, Anne Sumbul, and Benedetto Farsaci, Bristol-Myers Squibb, Princeton, NJ; and Stephen M. Ansell, Mayo Clinic, Rochester, MN
| |
Collapse
|
588
|
Wight JC, Hawkes EA, Berlangieri SU, Khor R, Grigg AP. An abscopal effect may augment PD-1 inhibition in refractory classical Hodgkin lymphoma. Leuk Lymphoma 2018; 59:2749-2751. [PMID: 29569979 DOI: 10.1080/10428194.2018.1452217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Joel C Wight
- a Department of Haematology , Olivia Newton John Cancer and Wellness Centre, Austin Health , Heidelberg , Australia
| | - Eliza A Hawkes
- a Department of Haematology , Olivia Newton John Cancer and Wellness Centre, Austin Health , Heidelberg , Australia.,b Faculty of Health Sciences , Olivia Newton John Cancer Research Institute, La Trobe University , Heidelberg , Australia.,c Faculty of Health Sciences , Monash University , Melbourne , Australia.,d Department of Oncology , Eastern Health , Box Hill , Australia
| | - Salvatore U Berlangieri
- a Department of Haematology , Olivia Newton John Cancer and Wellness Centre, Austin Health , Heidelberg , Australia
| | - Richard Khor
- a Department of Haematology , Olivia Newton John Cancer and Wellness Centre, Austin Health , Heidelberg , Australia.,e Sir Peter MacCallum Department of Oncology , University of Melbourne , Melbourne , Australia.,f Faculty of Health Sciences , University of Melbourne , Melbourne , Australia
| | - Andrew P Grigg
- a Department of Haematology , Olivia Newton John Cancer and Wellness Centre, Austin Health , Heidelberg , Australia.,f Faculty of Health Sciences , University of Melbourne , Melbourne , Australia
| |
Collapse
|
589
|
Kallam A, Armitage JO. Current and emerging treatment options for a patient with a second relapse of Hodgkin’s lymphoma. Expert Rev Hematol 2018. [DOI: 10.1080/17474086.2018.1449637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Avyakta Kallam
- Division of Oncology/Hematology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, USA
| | - James O. Armitage
- Division of Oncology/Hematology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
590
|
Sun C, Mezzadra R, Schumacher TN. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018; 48:434-452. [PMID: 29562194 PMCID: PMC7116507 DOI: 10.1016/j.immuni.2018.03.014] [Citation(s) in RCA: 1544] [Impact Index Per Article: 220.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022]
Abstract
Expression of programmed death-ligand 1 (PD-L1) is frequently observed in human cancers. Binding of PD-L1 to its receptor PD-1 on activated T cells inhibits anti-tumor immunity by counteracting T cell-activating signals. Antibody-based PD-1-PD-L1 inhibitors can induce durable tumor remissions in patients with diverse advanced cancers, and thus expression of PD-L1 on tumor cells and other cells in the tumor microenviroment is of major clinical relevance. Here we review the roles of the PD-1-PD-L1 axis in cancer, focusing on recent findings on the mechanisms that regulate PD-L1 expression at the transcriptional, posttranscriptional, and protein level. We place this knowledge in the context of observations in the clinic and discuss how it may inform the design of more precise and effective cancer immune checkpoint therapies.
Collapse
Affiliation(s)
- Chong Sun
- Division of Molecular Oncology & Immunology, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Riccardo Mezzadra
- Division of Molecular Oncology & Immunology, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Ton N Schumacher
- Division of Molecular Oncology & Immunology, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
591
|
Refractory Hodgkin lymphoma: time for targeted therapies? Blood 2018; 131:1156-1157. [DOI: 10.1182/blood-2017-12-824136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
592
|
Herrera AF, Moskowitz AJ, Bartlett NL, Vose JM, Ramchandren R, Feldman TA, LaCasce AS, Ansell SM, Moskowitz CH, Fenton K, Ogden CA, Taft D, Zhang Q, Kato K, Campbell M, Advani RH. Interim results of brentuximab vedotin in combination with nivolumab in patients with relapsed or refractory Hodgkin lymphoma. Blood 2018; 131:1183-1194. [PMID: 29229594 PMCID: PMC5855021 DOI: 10.1182/blood-2017-10-811224] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 11/30/2017] [Indexed: 12/13/2022] Open
Abstract
In this phase 1/2 study, brentuximab vedotin (BV) and nivolumab (Nivo) administered in combination were evaluated as initial salvage therapy in patients with relapsed or refractory (R/R) classical Hodgkin lymphoma (HL). Patients received up to 4 cycles of combination treatment, with BV administered on day 1 and Nivo on day 8 of the first cycle. For cycles 2 to 4, BV and Nivo were both administered on day 1. After study treatment, responses were evaluated by investigators per the 2014 Lugano classification, and patients could proceed to autologous stem cell transplantation (ASCT). Sixty-two patients were enrolled; the complete response rate among all treated patients (n = 61) was 61%, with an objective response rate of 82%. Before ASCT, adverse events (AEs) occurred in 98% of patients, mostly grades 1 and 2. Infusion-related reactions (IRRs) occurred in 44% of patients overall, with 41% of patients experiencing an IRR during at least 1 infusion of BV. Five patients (8%) were treated with systemic steroids for immune-related AEs. A reduction of peripheral T-cell subsets including regulatory T cells was observed after the first dose of BV, and reduced serum levels of thymus- and activation-regulated chemokine concurrent with an increase in proinflammatory cytokines and chemokines were seen after the first BV plus Nivo infusions. The combination of BV plus Nivo was an active and well-tolerated first salvage regimen, potentially providing patients with R/R HL an alternative to traditional chemotherapy. This trial was registered at www.clinicaltrials.gov as #NCT02572167.
Collapse
Affiliation(s)
| | | | - Nancy L Bartlett
- Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, MO
| | - Julie M Vose
- University of Nebraska Medical Center, Omaha, NE
| | | | | | | | | | | | | | | | | | - Qu Zhang
- Seattle Genetics, Inc, Bothell, WA
| | | | | | | |
Collapse
|
593
|
Passweg JR, Baldomero H, Bader P, Basak GW, Bonini C, Duarte R, Dufour C, Kröger N, Kuball J, Lankester A, Montoto S, Nagler A, Snowden JA, Styczynski J, Mohty M. Is the use of unrelated donor transplantation leveling off in Europe? The 2016 European Society for Blood and Marrow Transplant activity survey report. Bone Marrow Transplant 2018. [PMID: 29540849 PMCID: PMC6128821 DOI: 10.1038/s41409-018-0153-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hematopoietic cell transplantation (HCT) is an established procedure for acquired and congenital disorders of the hematopoietic system. In 2016, there was a tendency for continued activity in this field with 43,636 HCT in 39,313 patients [16,507 allogeneic (42%), 22,806 autologous (58%)] reported by 679 centers in 49 countries in 2016. The main indications were myeloid malignancies 9547 (24%; 96% allogeneic), lymphoid malignancies 25,618 (65%; 20% allogeneic), solid tumors 1516 (4%; 2% allogeneic), and non-malignant disorders 2459 (6%; 85% allogeneic). There was a remarkable leveling off in the use of unrelated donor HCT being replaced by haploidentical HCT. Continued growth in allogeneic HCT for marrow failure, AML, and MPN was seen, whereas MDS appears stable. Allogeneic HCT for lymphoid malignancies vary in trend with increases for NHL and decreases for Hodgkin lymphoma and myeloma. Trends in CLL are not clear, with recent increases after a decrease in activity. In autologous HCT, the use in myeloma continues to expand but is stable in Hodgkin lymphoma. There is a notable increase in autologous HCT for autoimmune disease. These data reflect the most recent advances in the field, in which some trends and changes are likely to be related to development of non-transplant technologies.
Collapse
Affiliation(s)
- Jakob R Passweg
- EBMT Activity Survey Office, Hematology, Department of Medicine, University Hospital, Basel, Switzerland.
| | - Helen Baldomero
- EBMT Activity Survey Office, Hematology, Department of Medicine, University Hospital, Basel, Switzerland
| | - Peter Bader
- Universitätsklinikum Frankfurt, Goethe-Universität, Frankfurt am Main, Germany
| | - Grzegorz W Basak
- Department of Hematology, Oncology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Rafael Duarte
- Servicio de Hematologia y Hemoterapia, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Carlo Dufour
- Hematology Unit, G. Gaslini Children's Institute, Genova, Italy
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Hospital Eppendorf, Hamburg, Germany
| | - Jürgen Kuball
- Department of Haematology, University Medical Centre, Utrecht, The Netherlands
| | - Arjan Lankester
- Department of Pediatrics, Leiden University Medical Centre Leiden, Leiden, The Netherlands
| | - Silvia Montoto
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Arnon Nagler
- Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - John A Snowden
- Sheffield Teaching Hospitals NHS Foundation, Royal Hallamshire Hospital, Sheffield, UK
| | - Jan Styczynski
- Pediatric Hematology and Oncology, University Hospital, Collegium Medicum UMK, Bydgoszcz, Poland
| | - Mohamad Mohty
- Department of Hematology, Hospital Saint Antoine, Paris, France
| | | |
Collapse
|
594
|
Wang ZH, Shen L. Management of gastrointestinal adverse events induced by immune-checkpoint inhibitors. Chronic Dis Transl Med 2018; 4:1-7. [PMID: 29756118 PMCID: PMC5938241 DOI: 10.1016/j.cdtm.2017.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Indexed: 12/26/2022] Open
Affiliation(s)
- Zheng-Hang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
595
|
Minson A, Douglas G, Bilmon I, Grigg A. Low dose PD-1 inhibition in relapsed refractory Hodgkin lymphoma after allogeneic stem cell transplant with concomitant active GVHD. Br J Haematol 2018. [DOI: 10.1111/bjh.15186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Adrian Minson
- Department of Haematology; Austin Hospital; Heidelberg Vic Australia
| | - Genevieve Douglas
- Department of Haematology; Austin Hospital; Heidelberg Vic Australia
| | - Ian Bilmon
- Department of Haematology; Westmead Hospital; Westmead NSW Australia
| | - Andrew Grigg
- Department of Haematology; Austin Hospital; Heidelberg Vic Australia
| |
Collapse
|
596
|
Menter T, Tzankov A. Mechanisms of Immune Evasion and Immune Modulation by Lymphoma Cells. Front Oncol 2018; 8:54. [PMID: 29564225 PMCID: PMC5845888 DOI: 10.3389/fonc.2018.00054] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/20/2018] [Indexed: 02/06/2023] Open
Abstract
Purpose Targeting cancer cells by modulating the immune system has become an important new therapeutic option in many different malignancies. Inhibition of CTLA4/B7 and PD1/PDL1 signaling is now also being investigated and already successfully applied to various hematologic malignancies. Methods A literature review of PubMed and results of our own studies were compiled in order to give a comprehensive overview on this topic. Results We elucidate the pathophysiological role of immunosuppressive networks in lymphomas, ranging from changes in the cellular microenvironment composition to distinct signaling pathways such as PD1/PDL1 or CTLA4/B7/CD28. The prototypical example of a lymphoma manipulating and thereby silencing the immune system is Hodgkin lymphoma. Also other lymphomas, e.g., primary mediastinal B-cell lymphoma and some Epstein–Barr virus (EBV)-driven malignancies, use analogous survival strategies, while diffuse large B-cell lymphoma of the activated B-cell type, follicular lymphoma and angioimmunoblastic T-cell lymphoma to name a few, exert further immune escape strategies each. These insights have already led to new treatment opportunities and results of the most important clinical trials based on this concept are briefly summarized. Immune checkpoint inhibition might also have severe side effects; the mechanisms of the rather un(der)recognized hematological side effects of this treatment approach are discussed. Conclusion Silencing the host’s immune system is an important feature of various lymphomas. Achieving a better understanding of distinct pathways of interactions between lymphomas and different immunological microenvironment compounds yields substantial potential for new treatment concepts.
Collapse
Affiliation(s)
- Thomas Menter
- Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| |
Collapse
|
597
|
Optimal management of immune-related adverse events resulting from treatment with immune checkpoint inhibitors: a review and update. Int J Clin Oncol 2018. [PMID: 29516216 DOI: 10.1007/s10147-018-1259-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Over the last two decades, molecular-targeted agents have become mainstream treatment for many types of malignancies and have improved the overall survival of patients. However, most patients eventually develop resistance to these targeted therapies. Recently, immunotherapies such as immune checkpoint inhibitors have revolutionized the treatment paradigm for many types of malignancies. Immune checkpoint inhibitors have been approved for treatment of melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck squamous cell carcinoma, Hodgkin's lymphoma, bladder cancer and gastric cancer. However, oncologists have been faced with immune-related adverse events caused by immune checkpoint inhibitors; these are generally mild but can be fatal in some cases. Because immune checkpoint inhibitors have distinct toxicity profiles from those of chemotherapy or targeted therapy, many oncologists are not familiar with the principles for optimal management of immune-related adverse events, which require early recognition and appropriate treatment without delay. To achieve this, oncologists must educate patients and health-care workers, develop checklists of appropriate tests for immune-related adverse events and collaborate closely with organ specialists. Clinical questions that remain include whether immune checkpoint inhibitors should be administered to patients with autoimmune disease and whether patients for whom immune-related adverse events lead to delays in immunotherapy should be retreated. In addition, the predicted use of combination immunotherapies in the near future means that oncologists will face a higher incidence and severity of immune-related adverse events. This review provides an overview of the optimal management of immune-related adverse events attributed to immune checkpoint inhibitors.
Collapse
|
598
|
Abstract
INTRODUCTION Urothelial bladder cancer is one of the most predominant malignancies worldwide with a poor prognosis when presented at an advanced or metastatic stage. Improving the therapeutic landscape in this setting has been an unmet medical need. Palliative cisplatin-based chemotherapy is currently the standard of care in first line therapies, but many patients are ineligible and few alternative therapies exist. Moreover second-line chemotherapy has minimal activity. Recently, immune-checkpoint inhibitors have shifted the therapeutic armamentarium of bladder cancer and it is now necessary to redesign the therapeutic paradigm. Areas covered: In this article, we focus on the development of durvalumab and provide an overview of the safety, activity, efficacy and future perspectives of this drug in urothelial carcinoma. Expert commentary: Durvalumab is a well-tolerated drug and demonstrated major and durable response in advanced bladder cancer. Combinations with durvalumab will probably emerge as promising therapeutic strategies for the treatment of urothelial carcinoma. Further research efforts are needed to identify predictive biomarkers of response to immune-oncology agents.
Collapse
Affiliation(s)
- Pernelle Lavaud
- a Gustave Roussy, Department de Medicine Oncologique & INSERM U981 , Université Paris-Saclay , Villejuif , France
| | - Zineb Hamilou
- a Gustave Roussy, Department de Medicine Oncologique & INSERM U981 , Université Paris-Saclay , Villejuif , France
| | - Yohann Loriot
- a Gustave Roussy, Department de Medicine Oncologique & INSERM U981 , Université Paris-Saclay , Villejuif , France
| | - Christophe Massard
- b Drug Development Department (DITEP), Gustave Roussy , Université Paris-Sud, Université Paris-Saclay , Villejuif , France
| |
Collapse
|
599
|
Nieto Y, Thall PF, Ma J, Valdez BC, Ahmed S, Anderlini P, Popat U, Jones RB, Shpall EJ, Hosing C, Qazilbash M, Kebriaei P, Alousi A, Timmons M, Gulbis A, Myers A, Oki Y, Fanale M, Dabaja B, Pinnix C, Milgrom S, Champlin R, Andersson BS. Phase II Trial of High-Dose Gemcitabine/Busulfan/Melphalan with Autologous Stem Cell Transplantation for Primary Refractory or Poor-Risk Relapsed Hodgkin Lymphoma. Biol Blood Marrow Transplant 2018; 24:1602-1609. [PMID: 29501779 DOI: 10.1016/j.bbmt.2018.02.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/26/2018] [Indexed: 12/30/2022]
Abstract
We conducted a prospective phase 2 trial of gemcitabine, busulfan and melphalan (Gem/Bu/Mel) with autologous stem cell transplantation (ASCT) in patients with primary refractory or poor-risk relapsed Hodgkin lymphoma (HL) (ie, extranodal relapse or within 1 year of frontline therapy). The trial was powered to detect an improvement in 2-year progression-free survival (PFS) from a historical 50% using a BEAM regimen (carmustine/etoposide/cytarabine/melphalan) to 65%. We compared the study population with all other concurrent patients who were eligible for the trial but instead received the BEAM regimen at our center. No patient received post-ASCT maintenance therapy. The Gem/Bu/Mel trial enrolled 80 patients with a median age of 31 years, 41% with primary refractory HL and 59% with relapsed HL (36% extranodal relapses), and 30% with positron emission tomography (PET)-positive lesions at ASCT. The concurrent BEAM (n = 45) and Gem/Bu/Mel cohorts were well balanced except for higher rates of bulky relapse and PET-positive tumors in the Gem/Bu/Mel cohort. There were no transplantation-related deaths in either cohort. At a median follow-up of 34.5 months (range, 26 to 72 months), Gem/Bu/Mel was associated with better 2-year PFS (65% versus 51%; P = .008) and overall survival (89% versus 73%; P = .0003). In conclusion, our data show that Gem/Bu/Mel is safe, in this nonrandomized comparison yielding improved outcomes compared with a concurrently treated and prognostically matched cohort of patients with primary refractory or poor-risk relapsed HL receiving BEAM.
Collapse
Affiliation(s)
- Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| | - Peter F Thall
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Junsheng Ma
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Benigno C Valdez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Sairah Ahmed
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Paolo Anderlini
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Roy B Jones
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Muzaffar Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Melissa Timmons
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Alison Gulbis
- Department of Pharmacy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Alan Myers
- Department of Pharmacy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Yasuhiro Oki
- Department of Lymphoma and Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Michelle Fanale
- Department of Lymphoma and Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Bouthaina Dabaja
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Chelsea Pinnix
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Sarah Milgrom
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Richard Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
600
|
Biology of classical Hodgkin lymphoma: implications for prognosis and novel therapies. Blood 2018; 131:1654-1665. [PMID: 29500175 DOI: 10.1182/blood-2017-09-772632] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/09/2018] [Indexed: 12/18/2022] Open
Abstract
Hodgkin lymphoma is considered a prime example of treatment success, with cure rates exceeding 80% using modern combined modality therapies. However, especially in adolescents and young adults, treatment-related toxicity and long-term morbidity still represent persistent challenges. Moreover, outcomes in patients with relapsed or refractory disease remain unfavorable in the era of high-dose chemotherapy and stem-cell transplantation. Hence, there is a high demand for novel and innovative alternative treatment approaches. In recent years, many new therapeutic agents have emerged from preclinical and clinical studies that target molecular hallmarks of Hodgkin lymphoma, including the aberrant phenotype of the tumor cells, deregulated oncogenic pathways, and immune escape. The antibody-drug conjugate brentuximab vedotin and immune checkpoint inhibitors have already shown great success in patients with relapsed/refractory disease, leading to US Food and Drug Administration approval and new trials testing these agents in various clinical settings. The expanding knowledge and understanding of Hodgkin lymphoma biology and disease progression, as well as the development of robust tools for biomarker-driven risk stratification and therapeutic decision making, continue to be fundamentally important for the success of these and other novel agents. We anticipate that the availability and clinical implementation of novel molecular assays will be instrumental in an era of rapid shifts in the treatment landscape of this disease. Here, we review the current knowledge of Hodgkin lymphoma pathobiology, highlighting the related development of novel treatment strategies and prognostic models that hold the promise to continually challenge and change the current standard of care in classical Hodgkin lymphoma.
Collapse
|