1
|
Lozano E, Mena MP, Garrabou G, Cardús O, Díaz T, Moreno DF, Mañé-Pujol J, Oliver-Caldés A, Battram A, Tovar N, Cibeira MT, Rodríguez-Lobato LG, Bladé J, Fernández de Larrea C, Rosiñol L. Increased PVR Expression on Bone Marrow Macrophages May Promote Resistance to TIGIT Blockade in Multiple Myeloma. Clin Cancer Res 2024; 30:3944-3955. [PMID: 38990101 DOI: 10.1158/1078-0432.ccr-24-0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/08/2024] [Accepted: 07/08/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE TIGIT blockade in our ex vivo model of bone marrow (BM) reduced the number of malignant plasma cells (PC) in only half of patients with multiple myeloma. Here, we wanted to investigate whether increased expression of TIGIT ligands may inhibit T-cell immune response promoting resistance to TIGIT blockade. EXPERIMENTAL DESIGN We first characterized the number and phenotype of BM macrophages in different stages of the disease by multiparameter flow cytometry. We assessed the effect of TIGIT ligands on PC survival by performing experiments in the ex vivo BM model and analyzed changes in gene expression by using NanoString technology and real-time PCR. RESULTS The frequency of BM macrophages was significantly decreased in multiple myeloma, which was accompanied by changes in their immunophenotype. Moreover, we found a higher number of malignant PC in ex vivo BM cells cultured onto the poliovirus receptor (PVR) and nectin-2 compared with control, suggesting that both ligands may support PC survival. In addition, the presence of PVR, but not nectin-2, overcame the therapeutic effect of TIGIT blockade or exogenous IL2. Furthermore, exogenous IL2 increased TIGIT expression on both CD4+ and CD8+ T cells and, indirectly, PVR on BM macrophages. Consistently, PVR reduced the number of cytotoxic T cells and promoted a gene signature with reduced effector molecules. CONCLUSIONS IL2 induced TIGIT on T cells in the BM, in which increased PVR expression resulted in cytotoxic T-cell inhibition, promoting PC survival and resistance to TIGIT blockade.
Collapse
Affiliation(s)
- Ester Lozano
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona (UB), Barcelona, Spain
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Mari-Pau Mena
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Glòria Garrabou
- Inherited Metabolic Diseases and Muscular Disorders Research Lab, Cellex-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine and Health Sciences-University of Barcelona, Barcelona, Spain
| | - Oriol Cardús
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Tania Díaz
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - David F Moreno
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Joan Mañé-Pujol
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Aina Oliver-Caldés
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Anthony Battram
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Natalia Tovar
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - María-Teresa Cibeira
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Luis-Gerardo Rodríguez-Lobato
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Joan Bladé
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Carlos Fernández de Larrea
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Laura Rosiñol
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| |
Collapse
|
2
|
Jafari-Raddani F, Davoodi-Moghaddam Z, Bashash D. Construction of immune-related gene pairs signature to predict the overall survival of multiple myeloma patients based on whole bone marrow gene expression profiling. Mol Genet Genomics 2024; 299:47. [PMID: 38649532 DOI: 10.1007/s00438-024-02140-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
Multiple myeloma (MM) is a plasma cell dyscrasia that is characterized by the uncontrolled proliferation of malignant PCs in the bone marrow. Due to immunotherapy, attention has returned to the immune system in MM, and it appears necessary to identify biomarkers in this area. In this study, we created a prognostic model for MM using immune-related gene pairs (IRGPs), with the advantage that it is not affected by technical bias. After retrieving microarray data of MM patients, bioinformatics analyses like COX regression and least absolute shrinkage and selection operator (LASSO) were used to construct the signature. Then its prognostic value is assessed via time-dependent receiver operating characteristic (ROC) and the Kaplan-Meier (KM) analysis. We also used XCELL to examine the status of immune cell infiltration among MM patients. 6-IRGP signatures were developed and proved to predict MM prognosis with a P-value of 0.001 in the KM analysis. Moreover, the risk score was significantly associated with clinicopathological characteristics and was an independent prognostic factor. Of note, the combination of age and β2-microglobulin with risk score could improve the accuracy of determining patients' prognosis with the values of the area under the curve (AUC) of 0.73 in 5 years ROC curves. Our model was also associated with the distribution of immune cells. This novel signature, either alone or in combination with age and β2-microglobulin, showed a good prognostic predictive value and might be used to guide the management of MM patients in clinical practice.
Collapse
Affiliation(s)
- Farideh Jafari-Raddani
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Davoodi-Moghaddam
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
4
|
Soekojo CY, Chng WJ. The Evolution Of Immune Dysfunction In Multiple Myeloma. Eur J Haematol 2022; 109:415-424. [PMID: 35880386 DOI: 10.1111/ejh.13839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES This review discusses the role of immune dysfunction at the different stages of MM. METHODS Narrative review RESULTS: Multiple myeloma (MM) is a complex disease and immune dysfunction has been known to play an important role in disease pathogenesis, progression, and drug resistance. MM is known to be preceded by asymptomatic precursor states and progression from the precursor states to MM is likely related to a progressive impairment of the immune system. CONCLUSIONS An understanding of the role of the immune system in the progression of MM is important to guide the development of immunotherapeutic strategies for this disease.
Collapse
Affiliation(s)
- Cinnie Yentia Soekojo
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System
| | - Wee Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System
| |
Collapse
|
5
|
Giannakoulas N, Ntanasis-Stathopoulos I, Terpos E. The Role of Marrow Microenvironment in the Growth and Development of Malignant Plasma Cells in Multiple Myeloma. Int J Mol Sci 2021; 22:ijms22094462. [PMID: 33923357 PMCID: PMC8123209 DOI: 10.3390/ijms22094462] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/16/2022] Open
Abstract
The development and effectiveness of novel therapies in multiple myeloma have been established in large clinical trials. However, multiple myeloma remains an incurable malignancy despite significant therapeutic advances. Accumulating data have elucidated our understanding of the genetic background of the malignant plasma cells along with the role of the bone marrow microenvironment. Currently, the interaction among myeloma cells and the components of the microenvironment are considered crucial in multiple myeloma pathogenesis. Adhesion molecules, cytokines and the extracellular matrix play a critical role in the interplay among genetically transformed clonal plasma cells and stromal cells, leading to the proliferation, progression and survival of myeloma cells. In this review, we provide an overview of the multifaceted role of the bone marrow microenvironment in the growth and development of malignant plasma cells in multiple myeloma.
Collapse
Affiliation(s)
- Nikolaos Giannakoulas
- Department of Hematology of University Hospital of Larisa, Faculty of Medicine, University of Thessaly, 41110 Larisa, Greece;
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
- Correspondence:
| |
Collapse
|
6
|
Díaz-Tejedor A, Lorenzo-Mohamed M, Puig N, García-Sanz R, Mateos MV, Garayoa M, Paíno T. Immune System Alterations in Multiple Myeloma: Molecular Mechanisms and Therapeutic Strategies to Reverse Immunosuppression. Cancers (Basel) 2021; 13:cancers13061353. [PMID: 33802806 PMCID: PMC8002455 DOI: 10.3390/cancers13061353] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary A common characteristic of multiple myeloma (MM) is the dysfunction of patients’ immune system, a condition termed immunosuppression. This state is mainly due to alterations in the number and functionality of the principal immune populations. In this setting, immunotherapy has acquired high relevance in the last years and the investigation of agents that boost the immune system represent a field of interest. In the present review, we will summarize the main cellular and molecular alterations observed in MM patients’ immune system. Furthermore, we will describe the mechanisms of action of the four immunotherapeutic drugs approved so far for the treatment of MM, which are part of the group of monoclonal antibodies (mAbs). Finally, the immune-stimulating effects of several therapeutic agents are described due to their potential role in reversing immunosuppression and, therefore, in favoring the efficacy of immunotherapy drugs, such as mAbs, as part of future pharmacological combinations. Abstract Immunosuppression is a common feature of multiple myeloma (MM) patients and has been associated with disease evolution from its precursor stages. MM cells promote immunosuppressive effects due to both the secretion of soluble factors, which inhibit the function of immune effector cells, and the recruitment of immunosuppressive populations. Alterations in the expression of surface molecules are also responsible for immunosuppression. In this scenario, immunotherapy, as is the case of immunotherapeutic monoclonal antibodies (mAbs), aims to boost the immune system against tumor cells. In fact, mAbs exert part of their cytotoxic effects through different cellular and soluble immune components and, therefore, patients’ immunosuppressive status could reduce their efficacy. Here, we will expose the alterations observed in symptomatic MM, as compared to its precursor stages and healthy subjects, in the main immune populations, especially the inhibition of effector cells and the activation of immunosuppressive populations. Additionally, we will revise the mechanisms responsible for all these alterations, including the interplay between MM cells and immune cells and the interactions among immune cells themselves. We will also summarize the main mechanisms of action of the four mAbs approved so far for the treatment of MM. Finally, we will discuss the potential immune-stimulating effects of non-immunotherapeutic drugs, which could enhance the efficacy of immunotherapeutic treatments.
Collapse
Affiliation(s)
- Andrea Díaz-Tejedor
- Centro de Investigación del Cáncer-IBMCC (CSIC-Universidad de Salamanca), Complejo Asistencial Universitario de Salamanca-IBSAL, Department of Hematology, 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (N.P.); (R.G.-S.); (M.-V.M.); (M.G.)
| | - Mauro Lorenzo-Mohamed
- Centro de Investigación del Cáncer-IBMCC (CSIC-Universidad de Salamanca), Complejo Asistencial Universitario de Salamanca-IBSAL, Department of Hematology, 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (N.P.); (R.G.-S.); (M.-V.M.); (M.G.)
| | - Noemí Puig
- Centro de Investigación del Cáncer-IBMCC (CSIC-Universidad de Salamanca), Complejo Asistencial Universitario de Salamanca-IBSAL, Department of Hematology, 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (N.P.); (R.G.-S.); (M.-V.M.); (M.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC, CB16/12/00233), Instituto de Salud Carlos III, 37007 Salamanca, Spain
| | - Ramón García-Sanz
- Centro de Investigación del Cáncer-IBMCC (CSIC-Universidad de Salamanca), Complejo Asistencial Universitario de Salamanca-IBSAL, Department of Hematology, 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (N.P.); (R.G.-S.); (M.-V.M.); (M.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC, CB16/12/00233), Instituto de Salud Carlos III, 37007 Salamanca, Spain
| | - María-Victoria Mateos
- Centro de Investigación del Cáncer-IBMCC (CSIC-Universidad de Salamanca), Complejo Asistencial Universitario de Salamanca-IBSAL, Department of Hematology, 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (N.P.); (R.G.-S.); (M.-V.M.); (M.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC, CB16/12/00233), Instituto de Salud Carlos III, 37007 Salamanca, Spain
| | - Mercedes Garayoa
- Centro de Investigación del Cáncer-IBMCC (CSIC-Universidad de Salamanca), Complejo Asistencial Universitario de Salamanca-IBSAL, Department of Hematology, 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (N.P.); (R.G.-S.); (M.-V.M.); (M.G.)
| | - Teresa Paíno
- Centro de Investigación del Cáncer-IBMCC (CSIC-Universidad de Salamanca), Complejo Asistencial Universitario de Salamanca-IBSAL, Department of Hematology, 37007 Salamanca, Spain; (A.D.-T.); (M.L.-M.); (N.P.); (R.G.-S.); (M.-V.M.); (M.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC, CB16/12/00233), Instituto de Salud Carlos III, 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-923-294-812; Fax: +34-923-294-743
| |
Collapse
|
7
|
Zou Z, Guo T, Cui J, Tang W, Li Y, Wang F, Dong T, Yang Y, Feng Y, Ho M, Zhang L, Pan L, Niu T. Real-world data combined with studies on Regulatory B Cells for newly diagnosed Multiple Myeloma from a tertiary referral Hospital in South-Western China. J Cancer 2021; 12:2633-2642. [PMID: 33854623 PMCID: PMC8040702 DOI: 10.7150/jca.53209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 01/30/2021] [Indexed: 02/05/2023] Open
Abstract
Multiple myeloma (MM) is a heterogeneous disease that remains incurable with significant interpatient variability in outcomes. Regulatory B cells (Bregs) were observed to be involved into specific defects in MM. Here, we provide our risk-adapted approach to newly diagnosed MM (NDMM), combining with the fundamental dysfunction of Bregs. We reported one hundred consecutive patients with NDMM from South-Western China, primarily treated with bortezomib plus dexamethasone with or without a 3rd agent, were enrolled from 2017. Bone marrow aspirates were obtained and flow cytometry (FCM) was used to quantify the percentage of Bregs from the bone marrow. The correlation between Bregs and clinical characters were further analyzed. This study found using bortezomib plus dexamethasone as backbone showed promising efficacy with acceptable tolerability in NDMM. The relatively compromised progression free survival (PFS) points to the essential synergy of bortezomib and lenalidomide here. This study also found that altered proportions of Bregs were closely correlated with treatment efficacy and prognosis in MM. Further understanding of Bregs biology might provide new opportunities to develop immunotherapy, which could prove beneficial in treating MM.
Collapse
Affiliation(s)
- Zhongqing Zou
- Department of Hematology, West China Hospital, Sichuan University, China
- Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Tingting Guo
- Department of Hematology, West China Hospital, Sichuan University, China
- Center for Precision Medicine, West China Hospital, Sichuan University, China
| | - Jian Cui
- Department of Hematology, West China Hospital, Sichuan University, China
| | - Wenjiao Tang
- Department of Hematology, West China Hospital, Sichuan University, China
| | - Yan Li
- Department of Hematology, West China Hospital, Sichuan University, China
| | - Fangfang Wang
- Department of Hematology, West China Hospital, Sichuan University, China
- Hematology Research Laboratory, Department of Hematology, West China Hospital, Sichuan University, China
| | - Tian Dong
- Department of Hematology, West China Hospital, Sichuan University, China
| | - Yunfan Yang
- Department of Hematology, West China Hospital, Sichuan University, China
| | - Yan Feng
- Department of Hematology, West China Hospital, Sichuan University, China
| | - Matthew Ho
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Li Zhang
- Department of Hematology, West China Hospital, Sichuan University, China
| | - Ling Pan
- Department of Hematology, West China Hospital, Sichuan University, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, China
| |
Collapse
|
8
|
Lozano E, Mena MP, Díaz T, Martin-Antonio B, León S, Rodríguez-Lobato LG, Oliver-Caldés A, Cibeira MT, Bladé J, Prat A, Rosiñol L, Fernández de Larrea C. Nectin-2 Expression on Malignant Plasma Cells Is Associated with Better Response to TIGIT Blockade in Multiple Myeloma. Clin Cancer Res 2020; 26:4688-4698. [PMID: 32513837 DOI: 10.1158/1078-0432.ccr-19-3673] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/08/2020] [Accepted: 06/03/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE T-cell immunoreceptor with Ig and ITIM domain (TIGIT) blockade could represent an alternative therapeutic option to release the immune response in patients with multiple myeloma. Here we analyzed the expression of TIGIT and its ligands poliovirus receptor (PVR) and nectin-2 in the bone marrow (BM) of patients with monoclonal gammopathies and the efficacy of TIGIT blockade activating antimyeloma immunity. EXPERIMENTAL DESIGN Expression levels of TIGIT and its ligands were characterized by flow cytometry and ELISA. TIGIT blockade was analyzed in in vitro functional assays with peripheral T cells. BM cells were studied with NanoString technology, real-time PCR, and ex vivo patient BM cell models. RESULTS TIGIT and its ligands are highly expressed in the BM of patients with multiple myeloma, suggesting that may play a role in restraining immune activation. TIGIT blockade depleted FoxP3+ Tregs while increasing proliferation of IFNγ-producing CD4+ T cells from patients with multiple myeloma. PVR ligation inhibited CD8+ T-cell signaling and cell proliferation which could be overcome with anti-TIGIT mAb. However, BM cells showed a remarkable heterogeneity in immune signature. Accordingly, functional ex vivo BM assays revealed that only some patients respond to checkpoint blockade. Thus, response to TIGIT blockade correlated with low frequency of TIGIT+ cells and high nectin-2 expression on malignant plasma cells. CONCLUSIONS TIGIT blockade efficiently reinvigorated peripheral T cells from patients with multiple myeloma. However, in the BM, the efficacy of blocking anti-TIGIT mAb to achieve tumor cell death may depend on the expression of TIGIT and nectin-2, becoming potential predictive biomarkers for identifying patients who may benefit from TIGIT blockade.
Collapse
Affiliation(s)
- Ester Lozano
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Mari-Pau Mena
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Tania Díaz
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Beatriz Martin-Antonio
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Sheila León
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Luis-Gerardo Rodríguez-Lobato
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Aina Oliver-Caldés
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Teresa Cibeira
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Joan Bladé
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Aleix Prat
- Department of Medical Oncology, Hospital Clinic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Laura Rosiñol
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Carlos Fernández de Larrea
- Department of Hematology, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
9
|
De Keersmaecker B, Fostier K, Corthals J, Wilgenhof S, Heirman C, Aerts JL, Thielemans K, Schots R. Immunomodulatory drugs improve the immune environment for dendritic cell-based immunotherapy in multiple myeloma patients after autologous stem cell transplantation. Cancer Immunol Immunother 2014; 63:1023-36. [PMID: 24947180 PMCID: PMC11028612 DOI: 10.1007/s00262-014-1571-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 06/06/2014] [Indexed: 10/25/2022]
Abstract
Multiple myeloma (MM) is characterized by a malignant proliferation of plasma cells in the bone marrow with associated organ damage. Although the prognosis of MM has improved recently, the disease remains incurable for the large majority of patients. The eradication of residual disease in the bone marrow is a main target on the road toward cure. Immune cells play a role in the control of cancer and can be tools to attack residual MM cells. However, the myeloma-associated immune deficiency is a major hurdle to immunotherapy. We evaluated ex vivo the effects of low doses of the immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide on several immune cell types from MM patients after autologous stem cell transplantation and with low tumor burden. We observed that these drugs increased CD4(+) and CD8(+) T-cell proliferation and cytokine production, enhanced the lytic capacity of cytotoxic T lymphocytes and reduced the suppressive effects of regulatory T cells on CD8(+) T-cell responses. In addition, we found that functional dendritic cells (DCs) can be generated from mononuclear cells from MM patients. The presence of IMiDs improved the quality of antigen-specific T cells induced or expanded by these DCs as evidenced by a higher degree of T-cell polyfunctionality. Our results provide a rationale for the design of early phase clinical studies to assess the efficacy of DC-based immunotherapy in combination with posttransplant maintenance treatment with IMiDs in MM.
Collapse
Affiliation(s)
- Brenda De Keersmaecker
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090, Brussels, Belgium,
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Cellular immunotherapy in multiple myeloma: lessons from preclinical models. Biochim Biophys Acta Rev Cancer 2014; 1846:392-404. [PMID: 25109893 DOI: 10.1016/j.bbcan.2014.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/31/2014] [Accepted: 08/02/2014] [Indexed: 12/20/2022]
Abstract
The majority of multiple myeloma patients relapse with the current treatment strategies, raising the need for alternative therapeutic approaches. Cellular immunotherapy is a rapidly evolving field and currently being translated into clinical trials with encouraging results in several cancer types, including multiple myeloma. Murine multiple myeloma models are of critical importance for the development and refinement of cellular immunotherapy. In this review, we summarize the immune cell changes that occur in multiple myeloma patients and we discuss the cell-based immunotherapies that have been tested in multiple myeloma, with a focus on murine models.
Collapse
|
11
|
Immunological dysregulation in multiple myeloma microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:198539. [PMID: 25013764 PMCID: PMC4071780 DOI: 10.1155/2014/198539] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/20/2014] [Indexed: 12/22/2022]
Abstract
Multiple Myeloma (MM) is a systemic hematologic disease due to uncontrolled proliferation of monoclonal plasma cells (PC) in bone marrow (BM). Emerging in other solid and liquid cancers, the host immune system and the microenvironment have a pivotal role for PC growth, proliferation, survival, migration, and resistance to drugs and are responsible for some clinical manifestations of MM. In MM, microenvironment is represented by the cellular component of a normal bone marrow together with extracellular matrix proteins, adhesion molecules, cytokines, and growth factors produced by both stromal cells and PC themselves. All these components are able to protect PC from cytotoxic effect of chemo- and radiotherapy. This review is focused on the role of immunome to sustain MM progression, the emerging role of myeloid derived suppressor cells, and their potential clinical implications as novel therapeutic target.
Collapse
|
12
|
Tete SM, Bijl M, Sahota SS, Bos NA. Immune defects in the risk of infection and response to vaccination in monoclonal gammopathy of undetermined significance and multiple myeloma. Front Immunol 2014; 5:257. [PMID: 24917865 PMCID: PMC4042361 DOI: 10.3389/fimmu.2014.00257] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 05/18/2014] [Indexed: 12/13/2022] Open
Abstract
The plasma cell proliferative disorders monoclonal gammopathy of undetermined significance (MGUS) and malignant multiple myeloma (MM) are characterized by an accumulation of transformed clonal plasma cells in the bone marrow and production of monoclonal immunoglobulin. They typically affect an older population, with median age of diagnosis of approximately 70 years. In both disorders, there is an increased risk of infection due to the immunosuppressive effects of disease and conjointly of therapy in MM, and response to vaccination to counter infection is compromised. The underlying factors in a weakened immune response in MGUS and MM are as yet not fully understood. A confounding factor is the onset of normal aging, which quantitatively and qualitatively hampers humoral immunity to affect response to infection and vaccination. In this review, we examine the status of immune alterations in MGUS and MM and set these against normal aging immune responses. We focus primarily on quantitative and functional aspects of B-cell immunity. Furthermore, we review the current knowledge relating to susceptibility to infectious disease in MGUS and MM, and how efficacy of conventional vaccination is affected by proliferative disease-related and therapy-related factors.
Collapse
Affiliation(s)
- Sarah M Tete
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen , Groningen , Netherlands ; Cancer Sciences Unit, Faculty of Medicine, University of Southampton , Southampton , UK
| | - Marc Bijl
- Department of Internal Medicine and Rheumatology, Martini Hospital , Groningen , Netherlands
| | - Surinder S Sahota
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton , Southampton , UK
| | - Nicolaas A Bos
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen , Groningen , Netherlands
| |
Collapse
|
13
|
Teh BW, Harrison SJ, Pellegrini M, Thursky KA, Worth LJ, Slavin MA. Changing treatment paradigms for patients with plasma cell myeloma: impact upon immune determinants of infection. Blood Rev 2014; 28:75-86. [PMID: 24582081 DOI: 10.1016/j.blre.2014.01.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 01/22/2014] [Accepted: 01/30/2014] [Indexed: 12/28/2022]
Abstract
Plasma cell myeloma (PCM) is increasing in prevalence in older age groups and infective complications are a leading cause of mortality. Patients with PCM are at increased risk of severe infections, having deficits in many arms of the immune system due to disease and treatment-related factors. Treatment of PCM has evolved over time with significant impacts on immune function resulting in changing rates and pattern of infection. Recently, there has been a paradigm shift in the treatment of PCM with the use of immunomodulatory drugs and proteasome inhibitors becoming the standard of care. These drugs have wide-ranging effects on the immune system but their impact on infection risk and aetiology remain unclear. The aims of this review are to discuss the impact of patient, disease and treatment factors on immune function over time for patients with PCM and to correlate immune deficits with the incidence and aetiology of infections seen clinically in these patients. Preventative measures and the need for clinically relevant tools to enable infective profiling of patients with PCM are discussed.
Collapse
Affiliation(s)
- Benjamin W Teh
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Simon J Harrison
- Department of Haematology, Peter MacCallum Cancer Centre, East Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Marc Pellegrini
- Walter and Eliza Hall Institute for Medical Research, Parkville, Australia
| | - Karin A Thursky
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne, Australia; Victorian Infectious Diseases Service, Royal Melbourne Hospital, Parkville, Australia
| | - Leon J Worth
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Monica A Slavin
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne, Australia; Victorian Infectious Diseases Service, Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
14
|
Teh B, Khot A, Harrison S, Prince H, Slavin M. A messenger at the door: cytomegalovirus retinitis in myeloma patients with progressive disease. Transpl Infect Dis 2013; 15:E134-8. [DOI: 10.1111/tid.12106] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/14/2013] [Accepted: 03/05/2013] [Indexed: 11/27/2022]
Affiliation(s)
- B.W. Teh
- Department of Infectious Diseases and Infection Control; Peter MacCallum Cancer Centre; East Melbourne; Victoria; Australia
| | - A.S. Khot
- Department of Haematology; Peter MacCallum Cancer Centre; East Melbourne; Victoria; Australia
| | | | | | | |
Collapse
|
15
|
Rossi M, Botta C, Correale P, Tassone P, Tagliaferri P. Immunologic microenvironment and personalized treatment in multiple myeloma. Expert Opin Biol Ther 2013; 13 Suppl 1:S83-93. [PMID: 23692463 DOI: 10.1517/14712598.2013.799130] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is characterized by generalized immune suppression and increased susceptibility to infections and secondary malignancies. Malignant plasma cells (PCs) modulate the bone marrow microenvironment to favor their own survival and proliferation. These events lead to a severe deregulation of immune effectors. Extensive studies have been conducted to unveil the mechanisms through which MM cells negatively modulate immunity and to develop therapeutical approaches for restoring an efficient anti-MM immune response. AREAS COVERED This review article covers both the immunosuppressive effects exerted by MM and the immunomodulatory potential of novel anti-MM agents. A brief overview on the most promising immunotherapeutic approaches in the field is also provided. EXPERT OPINION MM leads to a progressive impairment of the immune system. Different approaches have been evaluated or are currently under investigation to boost a specific anti-MM immune response. The discovery that anti-MM agents like bortezomib also retain immunomodulatory properties provides evidence to support the development of combined treatment modalities. In the next future, immunotherapy will be likely included in selective treatments in early stages or in the post-transplantation setting with non toxic modalities that control or clear the neoplastic clone.
Collapse
Affiliation(s)
- Marco Rossi
- Magna Graecia University and T. Campanella Cancer Center, Department of Experimental and Clinical Medicine, Medical Oncology Unit, Viale Europa, 88100 Catanzaro, Italy
| | | | | | | | | |
Collapse
|
16
|
Biological activity of lenalidomide and its underlying therapeutic effects in multiple myeloma. Adv Hematol 2012; 2012:842945. [PMID: 22919394 PMCID: PMC3417169 DOI: 10.1155/2012/842945] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 05/01/2012] [Accepted: 05/02/2012] [Indexed: 01/22/2023] Open
Abstract
Lenalidomide is a synthetic compound derived by modifying the chemical structure of thalidomide. It belongs to the second generation of immunomodulatory drugs (IMiDs) and possesses pleiotropic properties. Even if lenalidomide has been shown to be active in the treatment of several hematologic malignancies, this review article is mostly focalized on its mode of action in multiple myeloma. The present paper is about the direct and indirect antitumor effects of lenalidomide on malignant plasmacells, bone marrow microenvironment, bone resorption and host's immune response. The molecular mechanisms and targets of lenalidomide remain largely unknown, but recent evidence shows cereblon (CRBN) as a possible mediator of its therapeutical effects.
Collapse
|
17
|
Nguyen-Pham TN, Lee YK, Lee HJ, Kim MH, Yang DH, Kim HJ, Lee JJ. Cellular immunotherapy using dendritic cells against multiple myeloma. THE KOREAN JOURNAL OF HEMATOLOGY 2012; 47:17-27. [PMID: 22479274 PMCID: PMC3317466 DOI: 10.5045/kjh.2012.47.1.17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 02/09/2012] [Accepted: 03/02/2012] [Indexed: 11/17/2022]
Abstract
Cellular therapy with dendritic cells (DCs) is emerging as a useful immunotherapeutic tool to treat multiple myeloma (MM). DC-based idiotype vaccination was recently suggested to induce idiotype-specific immune responses in MM patients. However, the clinical results so far have been largely disappointing, and the clinical effectiveness of such vaccinations in MM still needs to be demonstrated. DC-based therapies against MM may need to be boosted with other sources of tumor-associated antigens, and potent DCs should be recruited to increase the effectiveness of treatment. DCs with both high migratory capacity and high cytokine production are very important for effective DC-based cancer vaccination in order to induce high numbers of Th1-type CD4+ T cells and CD8+ cytotoxic T lymphocytes. The tumor microenvironment is also important in the regulation of tumor cell growth, proliferation, and the development of therapeutic resistance after treatment. In this review, we discuss how the efficacy of DC vaccination in MM can be improved. In addition, novel treatment strategies that target not only myeloma cells but also the tumor microenvironment are urgently needed to improve treatment outcomes.
Collapse
Affiliation(s)
- Thanh-Nhan Nguyen-Pham
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | | | | | | | | | | | | |
Collapse
|
18
|
Kobold S, Luetkens T, Bartels BM, Cao Y, Hildebrandt Y, Sezer O, Reinhard H, Templin J, Bartels K, Lajmi N, Haag F, Bokemeyer C, Kröger N, Atanackovic D. Longitudinal analysis of tetanus- and influenza-specific IgG antibodies in myeloma patients. Clin Dev Immunol 2012; 2012:134081. [PMID: 22481961 PMCID: PMC3310258 DOI: 10.1155/2012/134081] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 12/25/2011] [Accepted: 12/28/2011] [Indexed: 11/21/2022]
Abstract
BACKGROUND Multiple myeloma (MM) and its therapies may induce a severely compromised humoral immunity. We have performed a longitudinal analysis of IgG-antibody responses against influenza virus (FLU) and tetanus toxoid (TT) as surrogate markers for the B cell-mediated immunity in MM patients. METHODS 1094 serum samples of 190 MM patients and samples from 100 healthy donors were analyzed by ELISA for FLU- and TT-specific antibodies. RESULTS MM patients evidenced lower levels of FLU- and TT-specific antibodies than healthy controls (P < 0.001). Immunoreactivity decreased with progressing disease and worsening clinical status. Levels of FLU- and TT-specific antibodies increased shortly (0-6 months) after alloSCT (P < 0.001), a time-period during which intravenous immunoglobulin (IVIG) is routinely applied. Thereafter, antibody concentrations declined and remained suppressed for 3 years in the case of FLU-specific and for more than 5 years in the case of TT-specific antibodies. CONCLUSIONS We found that MM is associated with a profound disease- and therapy-related immunosuppression, which is compensated for a few months after alloSCT, most likely by application of IVIG. This and the differences regarding the recovery of anti-FLU and anti-TT antibody titers during the following years need to be taken into account for optimizing IVIG application and immunization after alloSCT.
Collapse
Affiliation(s)
- Sebastian Kobold
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Division of Clinical Pharmacology, Department of Internal Medicine, Ludwig-Maximilian University, 80336 Munich, Germany
| | - Tim Luetkens
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Britta Marlen Bartels
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Yanran Cao
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - York Hildebrandt
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Orhan Sezer
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Henrike Reinhard
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Julia Templin
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Katrin Bartels
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nesrine Lajmi
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Friedrich Haag
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Djordje Atanackovic
- Department of Internal Medicine II, and Department of Oncology, Hematology, Bone Marrow Transplantation Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Hubertus Wald Tumorzentrum, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
19
|
Brimnes MK, Vangsted AJ, Knudsen LM, Gimsing P, Gang AO, Johnsen HE, Svane IM. Increased level of both CD4+FOXP3+ regulatory T cells and CD14+HLA-DR⁻/low myeloid-derived suppressor cells and decreased level of dendritic cells in patients with multiple myeloma. Scand J Immunol 2011; 72:540-7. [PMID: 21044128 DOI: 10.1111/j.1365-3083.2010.02463.x] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Patients with multiple myeloma (MM) suffer from a general impaired immunity comprising deficiencies in humoral responses, T-cell responses as well as dendritic cell (DC) function. Thus, to achieve control of tumour growth through immune therapy constitutes a challenge. Careful evaluation of the immune status in patients with MM seems crucial prior to active immune therapy. We evaluated the proportion of both, DC, Treg cells and myeloid-derived suppressor cells (MDSC) in peripheral blood from patients with MM at diagnosis and in remission as well as patients with monoclonal gammopathy of undetermined significance (MGUS). We found that the proportion of both myeloid (m) DC and plasmacytoid (p) DC in patients at diagnosis was lowered compared to control donors, while only the proportion of pDC in patients in remission and with MGUS was significantly lower than in controls. The proportion of CD4+FOXP3+ Treg cells was increased in patients at diagnosis and not in patients in remission or with MGUS. Also, Treg cells from patients with MM were functionally intact as they were able to inhibit proliferation of both CD4 and CD8 T cells. Finally, we observed an increase in the proportion of CD14+HLA-DR⁻/low MDSC in patients with MM at diagnosis, illustrating that this cell fraction is also distorted in patients with MM. Taken together, our results illustrate that, both mDC, pDC, Treg cells and MDSC are affected in patients with MM underlining the fact that the immune system is dysregulated as a consequence of the disease.
Collapse
Affiliation(s)
- M K Brimnes
- Center for Cancer Immune Therapy, Department of Haematology, Herlev Hospital, University of Copenhagen, Herlev, Denmark.
| | | | | | | | | | | | | |
Collapse
|
20
|
Ritchie DS, Quach H, Fielding K, Neeson P. Drug-mediated and cellular immunotherapy in multiple myeloma. Immunotherapy 2010; 2:243-55. [PMID: 20635931 DOI: 10.2217/imt.10.9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Multiple myeloma is an immunologically relevant disease, which subverts and suppresses immunity, but that may also be amenable to immunological control. Novel drug and cell-based therapies provide an opportunity for the design of antimyeloma immunotherapy. Reversing the immunosuppression associated myeloma remains a substantial challenge. The minimal residual disease setting achieved by autologous stem cell transplant or highly efficacious induction therapy may reverse this immunoparesis and provide a setting for induction of antimyeloma T-cell responses. Adoptive cytotoxic T-lymphocyte/NK therapy and comprehensive treatment with immunomodulatory drug therapy represent means by which antimyeloma immune responses may be promoted. In addition, apoptosis-inducing therapies may prime endogenous antigen presentation via immunogenic cell death, which again may be enhanced by the addition of immunomodulatory drug therapy.
Collapse
Affiliation(s)
- David S Ritchie
- Department of Haematology & Medical Oncology, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia.
| | | | | | | |
Collapse
|
21
|
Tramontana AR, George B, Hurt AC, Doyle JS, Langan K, Reid AB, Harper JM, Thursky K, Worth LJ, Dwyer DE, Morrissey CO, Johnson PDR, Buising KL, Harrison SJ, Seymour JF, Ferguson PE, Wang B, Denholm JT, Cheng AC, Slavin M. Oseltamivir resistance in adult oncology and hematology patients infected with pandemic (H1N1) 2009 virus, Australia. Emerg Infect Dis 2010; 16:1068-75. [PMID: 20587176 PMCID: PMC3321901 DOI: 10.3201/eid1607.091691] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We describe laboratory-confirmed influenza A pandemic (H1N1) 2009 in 17 hospitalized recipients of a hematopoietic stem cell transplant (HSCT) (8 allogeneic) and in 15 patients with malignancy treated at 6 Australian tertiary centers during winter 2009. Ten (31.3%) patients were admitted to intensive care, and 9 of them were HSCT recipients. All recipients of allogeneic HSCT with infection <100 days posttransplantation or severe graft-versus-host disease were admitted to an intensive care unit. In-hospital mortality rate was 21.9% (7/32). The H275Y neuraminidase mutation, which confers oseltamivir resistance developed in 4 of 7 patients with PCR positive for influenza after > or = 4 days of oseltamivir therapy. Three of these 4 patients were critically ill. Oseltamivir resistance in 4 (13.3%) of 30 patients who were administered oseltamivir highlights the need for ongoing surveillance of such resistance and further research on optimal antiviral therapy in the immunocompromised.
Collapse
|
22
|
Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood 2010; 115:5385-92. [PMID: 20395418 DOI: 10.1182/blood-2009-10-246660] [Citation(s) in RCA: 283] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Elevated cytokines in bone marrow (BM) micro-environment (interleukin-6 [IL-6], transforming growth factor-beta [TGF-beta], and IL-1beta) may play an important role in observed immune dysfunction in multiple myeloma (MM). As IL-6 and TGF-beta are important for the generation of T-helper 17 (T(H)17) cells, we evaluated and observed a significantly elevated baseline and induced frequency of T(h)17 cells in peripheral blood mononuclear cells (PBMCs) and BM mononuclear cells (BMMCs) from MM patients compared with healthy donors. We observed significant increase in levels of serum IL-17, IL-21, IL-22, and IL-23 in blood and BM in MM compared with healthy donors. We also observed that myeloma PBMCs after T(H)17 polarization significantly induced IL-1alpha, IL-13, IL-17, and IL-23 production compared with healthy donor PBMCs. We next observed that IL-17 promotes myeloma cell growth and colony formation via IL-17 receptor, adhesion to bone marrow stromal cells (BMSCs) as well as increased growth in vivo in murine xenograft model of human MM. Additionally, we have observed that combination of IL-17 and IL-22 significantly inhibited the production of T(H)1-mediated cytokines, including interferon-gamma (IFN-gamma), by healthy donor PBMCs. In conclusion, IL-17-producing T(h)17 cells play an important role in MM pathobiology and may be an important therapeutic target for anti-MM activity and to improve immune function.
Collapse
|
23
|
Abstract
Immunomodulatory drugs (IMiDs) are thalidomide analogues, which possess pleiotropic anti-myeloma properties including immune-modulation, anti-angiogenic, anti-inflammatory and anti-proliferative effects. Their development was facilitated by an improved understanding in myeloma (MM) biology and initiated a profound shift in the therapeutic approach towards MM. Despite the diverse effects of IMiDs in vitro, the relative contribution of each effect towards their ultimate anti-MM activity is still unclear. Based on in vitro data, it appears that anti-proliferative effects and downregulation of crucial cytokines are their most important anti-MM attributes. Although the co-stimulatory effects on T and NK cells have been heralded as a unique and important property of IMiDs towards enhancing anti-MM immune activity, these in vitro effects have yet to be firmly corroborated in vivo. Much is yet to be elucidated regarding the complex interplay of immunomodulatory cytokines that occurs in vivo, which ultimately dictates the net effects of IMiDs in MM-the understanding of which is necessary to facilitate optimal manipulation of these drugs in future MM management.
Collapse
|
24
|
Abstract
Immune cells with specific functions and abilities are vital to cancer treatment prevention. Although there have been many accomplishments made in the areas of immunotherapy and immunobiology of myeloma, there are still many obstacles in the way of conceptualizing the interrelationships between immune cells and tumor cells. To provide better understanding of these concepts and to move toward improved therapies for myeloma, cell-based therapeutic approaches should be developed.
Collapse
Affiliation(s)
- Nikhil C. Munshi
- Associate Director, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Associate Professor, Department of Medicine, Harvard Medical School, Boston, MA
- Staff Physician, VA Boston Healthcare System, Harvard Medical School, Boston, MA
| | - Rao H. Prabhala
- Instructor, Department of Medicine, Brigham and Women's Hospital/Dana Farber Cancer Institute, Boston, MA
- Research Health Scientist, VA Boston Healthcare System, Harvard Medical School, Boston, MA
| |
Collapse
|
25
|
Abstract
Multiple myeloma is a malignant tumour of plasma cells that remains incurable for the vast majority of patients, with a median survival of 2-3 years. It is characterized by the patchy accumulation of tumour cells within bone marrow leading to variable anaemia, bone destruction, hypercalcaemia, renal failure and infections. Immune dysfunction is an important feature of the disease and leads to infections that are both a major cause of morbidity and mortality and may promote tumour growth and resistance to chemotherapy. Numerous defects of the immune system have been described in multiple myeloma although the relative clinical importance of these remains elusive. There has been considerable interest in the identification of an autologous response against myeloma. Although T cells and humoral responses directed against myeloma-associated antigens have been described, it is uncertain if the immune system plays a role in preventing or controlling myeloma cell growth. There is increasing interest in the potential role of immunotherapy but the success of these interventions is likely to be modified by the immunologically hostile environment associated with multiple myeloma. This review attempts to summarize the current knowledge relating to the immune defects found in multiple myeloma.
Collapse
Affiliation(s)
- Guy Pratt
- CRUK Institute for Cancer Studies, University of Birmingham, Birmingham, UK.
| | | | | |
Collapse
|
26
|
Liu J, Hamrouni A, Wolowiec D, Coiteux V, Kuliczkowski K, Hetuin D, Saudemont A, Quesnel B. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-γ and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood 2007; 110:296-304. [PMID: 17363736 DOI: 10.1182/blood-2006-10-051482] [Citation(s) in RCA: 500] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Multiple myeloma (MM) cells inhibit certain T-cell functions. We examined the expression of B7-H1 (PD-L1), a B7-related protein that inhibits T-cell responses, in CD138-purified plasma cells isolated from MM patients, monoclonal gammopathy of undetermined significance patients, and healthy donors. We observed that B7-H1 was expressed in most MM plasma cells, but not cells isolated from monoclonal gammopathy of undetermined significance or healthy donors. This expression was increased or induced by IFN-γ and Toll-like receptor (TLR) ligands in isolated MM plasma cells. Blocking the MEK/ERK pathway inhibited IFN-γ–mediated and TLR-mediated expression of B7-H1. Inhibition of the MyD88 and TRAF6 adaptor proteins of the TLR pathway blocked not only B7-H1 expression induced by TLR ligands but also that mediated by IFN-γ. IFN-γ–induced STAT1 activation, via MEK/ERK and MyD88/TRAF6, and inhibition of STAT1 reduced B7-H1 expression. MM plasma cells stimulated with IFN-γ or TLR ligands inhibited cytotoxic T lymphocytes (CTLs) generation and this immunosuppressive effect was inhibited by preincubation with an anti-B7-H1 antibody, the UO126 MEK inhibitor, or by transfection of a dominant-negative mutant of MyD88. Thus, B7-H1 expression by MM cells represents a possible immune escape mechanism that could be targeted therapeutically through inhibition of MyD88/TRAF6 and MEK/ERK/STAT1.
Collapse
Affiliation(s)
- Jizhong Liu
- Institut National de la Santé et de la Recherche Médicale, Unité 837, Institut de Recherche sur le Cancer de Lille, France
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Beyer M, Kochanek M, Giese T, Endl E, Weihrauch MR, Knolle PA, Classen S, Schultze JL. In vivo peripheral expansion of naive CD4+CD25highFoxP3+ regulatory T cells in patients with multiple myeloma. Blood 2006; 107:3940-9. [PMID: 16410445 DOI: 10.1182/blood-2005-09-3671] [Citation(s) in RCA: 244] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In solid tumors, leukemias, and lymphomas, increased frequencies of functional CD4+CD25high regulatory T cells (Treg cells) have been previously demonstrated. In healthy individuals, Treg cells consist not only of memory but also of naive T cells, which can undergo peripheral expansion and are characterized by a relative enrichment for autoreactive T-cell receptors. Here, we demonstrate in patients with premalignant monoclonal gammopathy of undetermined significance and patients with multiple myeloma that functional FoxP3+ Treg cells of naive, central, and effector memory phenotype as determined by CCR7 and CD45RA expression are significantly expanded. Low frequencies of T-cell receptor excision circles in naive Treg cells in both healthy controls and multiple myeloma patients point to peripheral expansion as the prominent mechanism of increased frequencies of naive Treg cells in these cancer patients. These findings strongly suggest that the increase of functional Treg cells in cancer patients is a response to the process of malignant transformation.
Collapse
MESH Headings
- Antigens, CD/blood
- Antigens, CD/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Flow Cytometry
- Forkhead Transcription Factors/blood
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Humans
- Leukocyte Common Antigens/blood
- Leukocyte Common Antigens/genetics
- Multiple Myeloma/blood
- Multiple Myeloma/immunology
- Receptors, Antigen, T-Cell/blood
- Receptors, Antigen, T-Cell/immunology
- Receptors, CCR7
- Receptors, Chemokine/blood
- Receptors, Chemokine/genetics
- Receptors, Interleukin-2/blood
Collapse
Affiliation(s)
- Marc Beyer
- Molecular Tumor Biology and Tumor Immunology, Clinic I for Internal Medicine, University of Cologne, Joseph-Stelzmann Strasse 9/Haus 16, 50931 Cologne, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Weihrauch MR, Ansén S, Jurkiewicz E, Geisen C, Xia Z, Anderson KS, Gracien E, Schmidt M, Wittig B, Diehl V, Wolf J, Bohlen H, Nadler LM. Phase I/II combined chemoimmunotherapy with carcinoembryonic antigen-derived HLA-A2-restricted CAP-1 peptide and irinotecan, 5-fluorouracil, and leucovorin in patients with primary metastatic colorectal cancer. Clin Cancer Res 2005; 11:5993-6001. [PMID: 16115944 DOI: 10.1158/1078-0432.ccr-05-0018] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We conducted a phase I/II randomized trial to evaluate the clinical and immunologic effect of chemotherapy combined with vaccination in primary metastatic colorectal cancer patients with a carcinoembryonic antigen-derived peptide in the setting of adjuvants granulocyte macrophage colony-stimulating factor, CpG-containing DNA molecules (dSLIM), and dendritic cells. EXPERIMENTAL DESIGN HLA-A2-positive patients with confirmed newly diagnosed metastatic colorectal cancer and elevated serum carcinoembryonic antigen (CEA) were randomized to receive three cycles of standard chemotherapy (irinotecan/high-dose 5-fluorouracil/leucovorin) and vaccinations with CEA-derived CAP-1 peptide admixed with different adjuvants [CAP-1/granulocyte macrophage colony-stimulating factor/interleukin-2 (IL-2), CAP-1/dSLIM/IL-2, and CAP-1/IL-2]. After completion of chemotherapy, patients received weekly vaccinations until progression of disease. Immune assessment was done at baseline and after three cycles of combined chemoimmunotherapy. HLA-A2 tetramers complexed with the peptides CAP-1, human T-cell lymphotrophic virus type I TAX, cytomegalovirus (CMV) pp65, and EBV BMLF-1 were used for phenotypic immune assessment. IFN-gamma intracellular cytokine assays were done to evaluate CTL reactivity. RESULTS Seventeen metastatic patients were recruited, of whom 12 completed three cycles. Therapy resulted in five complete response, one partial response, five stable disease, and six progressive disease. Six grade 1 local skin reactions and one mild systemic reaction to vaccination treatment were observed. Overall survival after a median observation time of 29 months was 17 months with a survival rate of 35% (6 of 17) at that time. Eight patients (47%) showed elevation of CAP-1-specific CTLs. Neither of the adjuvants provided superiority in eliciting CAP-1-specific immune responses. During three cycles of chemotherapy, EBV/CMV recall antigen-specific CD8+ cells decreased by an average 14%. CONCLUSIONS The presented chemoimmunotherapy is a feasible and safe combination therapy with clinical and immunologic efficacy. Despite concurrent chemotherapy, increases in CAP-1-specific T cells were observed in 47% of patients after vaccination.
Collapse
Affiliation(s)
- Martin R Weihrauch
- Center for Experimental Medicine, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Lymphoproliferative disorders, including follicular lymphoma (FL), multiple myeloma (MM) and chronic lymphatic leukaemia (CLL), are slowly progressive malignancies which remain incurable despite advances in therapy. Harnessing the immune system to recognise and destroy tumours is a promising new approach to treating these diseases. Dendritic cells (DC) are unique antigen-presenting cells that play a central role in the initiation and direction of immune responses. DC loaded ex vivo with tumour-associated antigens and administered as a vaccine have already shown promise in early clinical trials for a number of lymphoproliferative disorders, but the need for improvement is widely agreed. Recent advances in the understanding of basic DC biology and lessons from early clinical trials have provided exciting new insights into the generation of anti-tumour immune responses and the design of vaccine strategies. In this review we provide an overview of our current understanding of DC biology and their function in patients with lymphoproliferative disorders. We discuss the current status of clinical trials and new approaches to exploit the antigen presenting capacity of DC to design vaccines of the future.
Collapse
MESH Headings
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Clinical Trials as Topic
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/pathology
- Lymphoma, Follicular/therapy
- Lymphoproliferative Disorders/immunology
- Lymphoproliferative Disorders/pathology
- Lymphoproliferative Disorders/therapy
- Multiple Myeloma/immunology
- Multiple Myeloma/pathology
- Multiple Myeloma/therapy
- Vaccination
Collapse
Affiliation(s)
- Kristen J Radford
- Mater Medical Research Institute, Dendritic Cell Laboratory, South Brisbane, Queensland, Australia.
| | | | | |
Collapse
|
30
|
Radford KJ, Turtle CJ, Kassianos AJ, Vuckovic S, Gardiner D, Khalil D, Taylor K, Wright S, Gill D, Hart DNJ. Immunoselection of functional CMRF-56+ blood dendritic cells from multiple myeloma patients for immunotherapy. J Immunother 2005; 28:322-31. [PMID: 16000950 DOI: 10.1097/01.cji.0000163592.66910.e4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Dendritic cells (DCs) loaded with tumor-associated antigens are a promising treatment to prevent disease relapse in patients with multiple myeloma (MM). Early-phase clinical trials have shown safety, efficacy, and immunologic responses in MM, but a key issue now is the isolation of a functional, clinically relevant DC preparation. The authors have described a unique blood DC (BDC) isolation platform based on positive immunoselection with the CMRF-56 antibody. To validate this as a feasible source of BDCs for immunotherapy, the authors undertook a quantitative and functional analysis of BDCs in MM patients and healthy donors. These data show that MM patients have similar numbers of CD11c+CD16+ and CD11c+CD16- BDCs but about half the number of CD11c-CD123+ BDCs in whole blood compared with healthy donors. BDCs could be isolated by CMRF-56+ immunoselection from all MM patients tested, with similar yields and purity to healthy donors. These BDCs could be activated ex vivo with poly I:C or LPS. Furthermore, CMRF-56+ preparations could induce potent CD4+ and CD8+ T-lymphocyte responses in both MM patients and healthy donors. These data suggest that BDCs with in vitro functional integrity can be isolated from MM patients in sufficient numbers to justify a clinical trial.
Collapse
|
31
|
Maecker B, von Bergwelt-Baildon MS, Anderson KS, Vonderheide RH, Anderson KC, Nadler LM, Schultze JL. Rare naturally occurring immune responses to three epitopes from the widely expressed tumour antigens hTERT and CYP1B1 in multiple myeloma patients. Clin Exp Immunol 2005; 141:558-62. [PMID: 16045747 PMCID: PMC1809449 DOI: 10.1111/j.1365-2249.2005.02879.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2005] [Indexed: 12/01/2022] Open
Abstract
The widely expressed tumour antigens hTERT and CYP1B1 are commonly expressed in multiple myeloma (MM) cells. Several trials targeting these antigens by immunotherapy have been initiated. The aim of this study was to explore whether patients with MM have an endogenous pre-existing immune response against recently identified epitopes from hTERT and CYP1B1. Peripheral blood T cells from 27 HLA-A*0201+ multiple myeloma patients at different stages of disease and 20 healthy HLA-A*0201+ donors were enriched and studied for the presence of hTERT- and CYP1B1-specific cytotoxic T cells using MHC tetramer detection and short-term ex vivo expansion. No significant expansion of tetramer-positive cells was detected in the peripheral blood of either MM patients or healthy controls when cells were stained with tetramers containing the dominant hTERT-derived epitope or two peptides derived from CYP1B1. A single ex vivo peptide stimulation led to the detection of a small population (0.3-0.5%) of hTERT-specific cells in two of 27 patients with MM. None of the patients or controls showed significant expansion of CYP1B1-specific cells after a single peptide stimulation. Thus, endogenous in vivo priming of T cells against hTERT and CYP1B1 is a rare event in MM patients. These results suggest that strategies targeting hTERT and CYP1B1 may have to utilize techniques to induce T cell responses from a naive precursor frequency.
Collapse
Affiliation(s)
- B Maecker
- Department of Adult Oncology, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Coleman S, Clayton A, Mason MD, Jasani B, Adams M, Tabi Z. Recovery of CD8+ T-Cell Function During Systemic Chemotherapy in Advanced Ovarian Cancer. Cancer Res 2005; 65:7000-6. [PMID: 16061686 DOI: 10.1158/0008-5472.can-04-3792] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunologic approaches are emerging as new treatment options in several types of cancer. However, whereas the ability of patients to develop potent CD8+ T-cell responses is crucial for efficient antitumor responses, immunocompetence and T-cell function are not tested routinely in patients entering immunotherapy. The objective of our study was to monitor T-cell function in advanced cancer and during chemotherapy. CD8+ T-cell function of 21 patients with advanced ovarian cancer (stages III-IV) was assessed by cytokine flow cytometry following stimulation of 42 PBMC samples with a panel of synthetic viral peptides in vitro, consisting of pan-Caucasian epitopes. CD8+ T-cell responses were significantly lower in patients with high levels (>200 units/mL) of Ca125 (marker of tumor load and progression) than in those with low Ca125 levels (P = 0.0013). In longitudinal studies of nine patients, chemotherapy was associated with decreasing Ca125 levels in seven cases and also with improvement or maintenance of CD8+ T-cell function in seven cases. After the full course of chemotherapy, five of nine patients in remission displayed potent CD8+ T-cell responses, whereas four of nine patients in progression displayed low or decreasing T-cell responses, pointing toward a correlation between T-cell function and clinical response. Our results show for the first time that CD8+ T-cell function is not permanently suppressed in advanced cancer and successful chemotherapy is associated with improved antigen-specific T-cell reactivity. We suggest that functional assays determining T-cell immunocompetence can be valuable tools for optimizing cancer immunotherapy for improved clinical success.
Collapse
Affiliation(s)
- Sharon Coleman
- Department of Oncology and Palliative Medicine, Velindre Hospital, Whitchurch, Cardiff, UK
| | | | | | | | | | | |
Collapse
|
33
|
Maecker B, von Bergwelt-Baildon MS, Sherr DH, Nadler LM, Schultze JL. Identification of a new HLA-A*0201-restricted cryptic epitope from CYP1B1. Int J Cancer 2005; 115:333-6. [PMID: 15688394 DOI: 10.1002/ijc.20906] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cytochrome P450 1B1 (CYP1B1) was recently shown to be a candidate tumor antigen broadly expressed in solid and hematologic malignancies. Nevertheless, use of such self-antigens as targets for immune intervention can be limited because of loss of high-avidity T cells during negative selection in the thymus. Recent data suggest that targeting of cryptic epitopes may represent a way to circumvent such self-tolerance and induce efficient antitumor CTL responses. Here, we present the identification and characterization of a novel, cryptic HLA-A*0201-binding peptide from CYP1B1. The nanomer CYP246 was identified by epitope deduction using algorithms to predict HLA-A*0201-binding peptides. CYP246 is characterized by strong initial HLA-A*0201 binding but a short MHC/peptide binding half-life. Expansion of high-avidity CTL was readily possible using autologous CD40-activated B cells from normal donors and cancer patients as antigen-presenting cells, suggesting that an intact T-cell repertoire can be expanded for this epitope. Lysis of CYP1B1-expressing, HLA-A*0201+ tumor cell lines and primary tumor cells confirmed that sufficient levels of CYP246 are presented by tumor cells for effector CTL killing. These findings indicate that CYP246 is a candidate cryptic epitope for immune interventions in which tumor CYP1B1 is targeted.
Collapse
Affiliation(s)
- Britta Maecker
- Department of Adult Oncology, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
34
|
Brown R, Murray A, Pope B, Sze DM, Gibson J, Ho PJ, Hart D, Joshua D. Either interleukin-12 or interferon-γ can correct the dendritic cell defect induced by transforming growth factor β
1
in patients with myeloma. Br J Haematol 2004; 125:743-8. [PMID: 15180863 DOI: 10.1111/j.1365-2141.2004.04984.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The poor response to immunotherapy in patients with multiple myeloma (MM) indicates that a better understanding of any defects in the immune response in these patients is required before effective therapeutic strategies can be developed. Recently we reported that high potency (CMRF44(+)) dendritic cells (DC) in the peripheral blood of patients with MM failed to significantly up-regulate the expression of the B7 co-stimulatory molecules, CD80 and CD86, in response to an appropriate signal from soluble trimeric human CD40 ligand. This defect was caused by transforming growth factor beta(1) (TGFbeta(1)) and interleukin (IL)-10, produced by malignant plasma cells, and the defect was neutralized in vitro with anti-TGFbeta(1). As this defect could impact on immunotherapeutic strategies and may be a major cause of the failure of recent trials, it was important to identify a more clinically useful agent that could correct the defect in vivo. In this study of 59 MM patients, the relative and absolute numbers of blood DC were only significantly decreased in patients with stage III disease and CD80 up-regulation was reduced in both stage I and stage III. It was demonstrated that both IL-12 and interferon-gamma neutralized the failure to stimulate CD80 up-regulation by huCD40LT in vitro. IL-12 did not cause a change in the distribution of DC subsets that were predominantly myeloid (CD11c+ and CDw123-) suggesting that there would be a predominantly T-helper cell type response. The addition of IL-12 or interferon-gamma to future immunotherapy trials involving these patients should be considered.
Collapse
Affiliation(s)
- Ross Brown
- Institute of Haematology, Royal Prince Alfred Hospital, Sydney, Australia.
| | | | | | | | | | | | | | | |
Collapse
|