1
|
Van Oekelen O, Amatangelo M, Guo M, Upadhyaya B, Cribbs AP, Kelly G, Patel M, Kim-Schulze S, Flynt E, Lagana A, Gooding S, Merad M, Jagganath S, Pierceall WE, Oppermann U, Thakurta A, Parekh S. Iberdomide increases innate and adaptive immune cell subsets in the bone marrow of patients with relapsed/refractory multiple myeloma. Cell Rep Med 2024; 5:101584. [PMID: 38776911 PMCID: PMC11228551 DOI: 10.1016/j.xcrm.2024.101584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/11/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Iberdomide is a potent cereblon E3 ligase modulator (CELMoD agent) with promising efficacy and safety as a monotherapy or in combination with other therapies in patients with relapsed/refractory multiple myeloma (RRMM). Using a custom mass cytometry panel designed for large-scale immunophenotyping of the bone marrow tumor microenvironment (TME), we demonstrate significant increases of effector T and natural killer (NK) cells in a cohort of 93 patients with multiple myeloma (MM) treated with iberdomide, correlating findings to disease characteristics, prior therapy, and a peripheral blood immune phenotype. Notably, changes are dose dependent, associated with objective response, and independent of prior refractoriness to MM therapies. This suggests that iberdomide broadly induces innate and adaptive immune activation in the TME, contributing to its antitumor efficacy. Our approach establishes a strategy to study treatment-induced changes in the TME of patients with MM and, more broadly, patients with cancer and establishes rational combination strategies for iberdomide with immune-enhancing therapies to treat MM.
Collapse
Affiliation(s)
- Oliver Van Oekelen
- Department of Medicine, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Manman Guo
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Bhaskar Upadhyaya
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam P Cribbs
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Geoffrey Kelly
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manishkumar Patel
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erin Flynt
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, USA
| | - Alessandro Lagana
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah Gooding
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Miriam Merad
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sundar Jagganath
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Udo Oppermann
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK; Oxford Translational Myeloma Centre (OTMC), Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Anjan Thakurta
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, USA; Oxford Translational Myeloma Centre (OTMC), Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Samir Parekh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
2
|
Lee H, Neri P, Bahlis NJ. Cereblon-Targeting Ligase Degraders in Myeloma: Mechanisms of Action and Resistance. Hematol Oncol Clin North Am 2024; 38:305-319. [PMID: 38302306 DOI: 10.1016/j.hoc.2024.01.001] [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] [Indexed: 02/03/2024]
Abstract
Cereblon-targeting degraders, including immunomodulatory imide drugs lenalidomide and pomalidomide alongside cereblon E3 ligase modulators like iberdomide and mezigdomide, have demonstrated significant anti-myeloma effects. These drugs play a crucial role in diverse therapeutic approaches for multiple myeloma (MM), emphasizing their therapeutic importance across various disease stages. Despite their evident efficacy, approximately 5% to 10% of MM patients exhibit primary resistance to lenalidomide, and resistance commonly develops over time. Understanding the intricate mechanisms of action and resistance to this drug class becomes imperative for refining and advancing novel therapeutic combinations.
Collapse
Affiliation(s)
- Holly Lee
- Arnie Charbonneau Cancer Institute, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive N.W., Calgary, Alberta T2N 4N1, Canada
| | - Paola Neri
- Arnie Charbonneau Cancer Institute, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive N.W., Calgary, Alberta T2N 4N1, Canada
| | - Nizar J Bahlis
- Arnie Charbonneau Cancer Institute, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive N.W., Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
3
|
Zarei M, Abdoli S, Farazmandfar T, Shahbazi M. Lenalidomide improves NKG2D-based CAR-T cell activity against colorectal cancer cells invitro. Heliyon 2023; 9:e20460. [PMID: 37790973 PMCID: PMC10543764 DOI: 10.1016/j.heliyon.2023.e20460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction Although CAR-based immunotherapy is viewed as a promising treatment for tumors, particularly hematological malignancies, solid tumors can pose challenges. It has been suggested that the immunomodulatory medication Lenalidomide (LEN) may increase the effectiveness of CAR T cells in the treatment of solid tumors. The purpose of our study was to investigate the effect of NKG2D-based CAR T cell therapy on colorectal cancer cell lines, and then we assessed combinatorial therapy using NKG2D CAR T cells and lenalidomide in vitro. Methods and results To prepare NKG2D CAR T cells, a second-generation NKG2D-CAR construct was designed and transfected into the T cells using a lentiviral system. The NKG2D CAR T cells showed significantly higher cytotoxic activity against colorectal cancer cell lines, HCT116 and SW480, compared to untransduced T cells. In addition, our data demonstrated that the cytotoxicity and cytokine secretion of NKG2D CAR T cells significantly increased in the presence of higher doses of lenalidomide. Conclusions The study findings suggest that combinational therapy, utilizing NKG2D-based CAR T cells and lenalidomide, has a high potential for effectively eliminating tumor cells in vitro.
Collapse
Affiliation(s)
- Mahdi Zarei
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shahriyar Abdoli
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Touraj Farazmandfar
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- AryaTina Gene (ATG) Biopharmaceutical Company Gorgan, Iran
| |
Collapse
|
4
|
Zhuang Y, Li C, Jiang H, Li L, Zhang Y, Yu W, Fu W. Multi-omics investigation of the resistance mechanisms of pomalidomide in multiple myeloma. Front Oncol 2023; 13:1264422. [PMID: 37799465 PMCID: PMC10549987 DOI: 10.3389/fonc.2023.1264422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023] Open
Abstract
Background Despite significant therapeutic advances over the last decade, multiple myeloma remains an incurable disease. Pomalidomide is the third Immunomodulatory drug that is commonly used to treat patients with relapsed/refractory multiple myeloma. However, approximately half of the patients exhibit resistance to pomalidomide treatment. While previous studies have identified Cereblon as a primary target of Immunomodulatory drugs' anti-myeloma activity, it is crucial to explore additional mechanisms that are currently less understood. Methods To comprehensively investigate the mechanisms of drug resistance, we conducted integrated proteomic and metabonomic analyses of 12 plasma samples from multiple myeloma patients who had varying responses to pomalidomide. Differentially expressed proteins and metabolites were screened, and were further analyzed using pathway analysis and functional correlation analysis. Also, we estimated the cellular proportions based on ssGSEA algorithm. To investigate the potential role of glycine in modulating the response of MM cells to pomalidomide, cell viability and apoptosis were analyzed. Results Our findings revealed a consistent decrease in the levels of complement components in the pomalidomide-resistant group. Additionally, there were significant differences in the proportion of T follicular helper cell and B cells in the resistant group. Furthermore, glycine levels were significantly decreased in pomalidomide-resistant patients, and exogenous glycine administration increased the sensitivity of MM cell lines to pomalidomide. Conclusion These results demonstrate distinct molecular changes in the plasma of resistant patients that could be used as potential biomarkers for identifying resistance mechanisms for pomalidomide in multiple myeloma and developing immune-related therapeutic strategies.
Collapse
Affiliation(s)
- Yan Zhuang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Department of Hematology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chenyu Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Hua Jiang
- Department of Hematology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lu Li
- Department of Hematology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanteng Zhang
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - WeiJun Fu
- Department of Hematology, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
5
|
Lurain K, Polizzotto MN, Krug LT, Shoemaker G, Singh A, Jensen SMR, Wyvill KM, Ramaswami R, Uldrick TS, Yarchoan R, Sereti I. Immunophenotypic analysis in participants with Kaposi sarcoma following pomalidomide administration. AIDS 2023; 37:1693-1703. [PMID: 37352498 PMCID: PMC10527758 DOI: 10.1097/qad.0000000000003627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2023]
Abstract
OBJECTIVE The aim of this study was to evaluate baseline differences by HIV status and the impact of pomalidomide on lymphocyte counts and T-cell subsets in patients with Kaposi sarcoma. DESIGN We prospectively evaluated CD4 + and CD8 + T-cell phenotypes in 19 participants with Kaposi sarcoma enrolled on a phase 1/2 study of pomalidomide (NCT01495598), seven without HIV and 12 with HIV on antiretroviral therapy. METHODS Trial participants received pomalidomide 5 mg orally for 21 days of 28-day cycles for up to 1 year. Flow cytometry was performed on peripheral blood mononuclear cells at baseline, after three cycles, and at end-of-treatment. Lymphocyte count and T-cell subset comparisons were evaluated by Wilcoxon signed-rank and Mann--Whitney tests. RESULTS At baseline, HIV + participants had lower CD4 + cell counts (median 416 vs. 742 CD4 + T cells/μl, P = 0.006), and a decreased proportion of CD57 + (senescent) CD8 + T cells ( P = 0.007) compared with HIV - participants. After three cycles, pomalidomide led to an increased proportion of CD45RO + CD27 + (central memory) CD4 + ( P = 0.002) and CD8 + ( P = 0.002) T cells, a decrease in CD45RO - CD27 - (effector) CD4 + cells ( P = 0.0002), and expansion of CD38 + /HLADR + (activated) CD4 + ( P = 0.002) and CD8 + ( P ≤ 0.0001) T cells. Increased numbers of activated CD8 + T cells persisted at end-of-treatment ( P = 0.002). After three cycles and at end-of-treatment, there was reduction in the proportion of CD57 + (senescent) CD4 + ( P = 0.001, 0.0006), and CD8 + ( P = < 0.0001, 0.0004) T cells. CONCLUSION Administration of pomalidomide decreased T-cell senescence and increased T-cell activation in patients with Kaposi sarcoma, suggesting pomalidomide activity in Kaposi sarcoma stems in part from its immunomodulatory effects.
Collapse
Affiliation(s)
- Kathryn Lurain
- HIV & AIDS Malignancy Branch, Center for Cancer Research (CCR), NCI
| | | | - Laurie T Krug
- HIV & AIDS Malignancy Branch, Center for Cancer Research (CCR), NCI
| | | | - Amrit Singh
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, Maryland, USA
| | - Stig M R Jensen
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, Maryland, USA
| | | | - Ramya Ramaswami
- HIV & AIDS Malignancy Branch, Center for Cancer Research (CCR), NCI
| | - Thomas S Uldrick
- HIV & AIDS Malignancy Branch, Center for Cancer Research (CCR), NCI
| | - Robert Yarchoan
- HIV & AIDS Malignancy Branch, Center for Cancer Research (CCR), NCI
| | - Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, Maryland, USA
| |
Collapse
|
6
|
Aghaee M, Ledzewicz U, Robbins M, Bezman N, Jay Cho H, Moore H. Determining Optimal Combination Regimens for Patients with Multiple Myeloma. Eur J Pharm Sci 2023:106492. [PMID: 37302768 DOI: 10.1016/j.ejps.2023.106492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/04/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
While many novel therapies have been approved in recent years for treating patients with multiple myeloma, there is still no established curative regimen, especially for patients with high-risk disease. In this work, we use a mathematical modeling approach to determine combination therapy regimens that maximize healthy lifespan for patients with multiple myeloma. We start with a mathematical model for the underlying disease and immune dynamics, which was presented and analyzed previously. We add the effects of three therapies to the model: pomalidomide, dexamethasone, and elotuzumab. We consider multiple approaches to optimizing combinations of these therapies. We find that optimal control combined with approximation outperforms other methods, in that it can quickly produce a combination regimen that is clinically-feasible and near-optimal. Implications of this work can be used to optimize doses and advance the scheduling of drugs.
Collapse
Affiliation(s)
- Mahya Aghaee
- Laboratory for Systems Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Urszula Ledzewicz
- Institute of Mathematics, Lodz University of Technology, Lodz, Poland; Dept. of Mathematics and Statistics, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | | | - Natalie Bezman
- Oncology Research and Development, Pfizer, La Jolla, California, USA
| | - Hearn Jay Cho
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Helen Moore
- Laboratory for Systems Medicine, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
7
|
Al-Haideri M, Tondok SB, Safa SH, maleki AH, Rostami S, Jalil AT, Al-Gazally ME, Alsaikhan F, Rizaev JA, Mohammad TAM, Tahmasebi S. CAR-T cell combination therapy: the next revolution in cancer treatment. Cancer Cell Int 2022; 22:365. [DOI: 10.1186/s12935-022-02778-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
AbstractIn recent decades, the advent of immune-based therapies, most notably Chimeric antigen receptor (CAR)-T cell therapy has revolutionized cancer treatment. The promising results of numerous studies indicate that CAR-T cell therapy has had a remarkable ability and successful performance in treating blood cancers. However, the heterogeneity and immunosuppressive tumor microenvironment (TME) of solid tumors have challenged the effectiveness of these anti-tumor fighters by creating various barriers. Despite the promising results of this therapeutic approach, including tumor degradation and patient improvement, there are some concerns about the efficacy and safety of the widespread use of this treatment in the clinic. Complex and suppressing tumor microenvironment, tumor antigen heterogeneity, the difficulty of cell trafficking, CAR-T cell exhaustion, and reduced cytotoxicity in the tumor site limit the applicability of CAR-T cell therapy and highlights the requiring to improve the performance of this treatment. With this in mind, in the last decade, many efforts have been made to use other treatments for cancer in combination with tuberculosis to increase the effectiveness of CAR-T cell therapy, especially in solid tumors. The combination therapy results have promising consequences for tumor regression and better cancer control compared to single therapies. Therefore, this study aimed to comprehensively discuss different cancer treatment methods in combination with CAR-T cell therapy and their therapeutic outcomes, which can be a helpful perspective for improving cancer treatment in the near future.
Collapse
|
8
|
Chen LY, Gooding S. Tumor and microenvironmental mechanisms of resistance to immunomodulatory drugs in multiple myeloma. Front Oncol 2022; 12:1038329. [PMID: 36439455 PMCID: PMC9682014 DOI: 10.3389/fonc.2022.1038329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/04/2022] [Indexed: 10/07/2023] Open
Abstract
Resistance to immunomodulatory drugs (IMiDs®) is a major cause of treatment failure, disease relapse and ultimately poorer outcomes in multiple myeloma (MM). In order to optimally deploy IMiDs and their newer derivates CRBN E3 ligase modulators (CELMoDs®) into future myeloma therapeutic regimens, it is imperative to understand the mechanisms behind the inevitable emergence of IMiD resistance. IMiDs bind and modulate Cereblon (CRBN), the substrate receptor of the CUL4CRBN E3 ubiquitin ligase, to target novel substrate proteins for ubiquitination and degradation. Most important of these are IKZF1 and IKZF3, key MM survival transcription factors which sustain the expression of myeloma oncogenes IRF4 and MYC. IMiDs directly target MM cell proliferation, but also stimulate T/NK cell activation by their CRBN-mediated effects, and therefore enhance anti-MM immunity. Thus, their benefits in myeloma are directed against tumor and immune microenvironment - and in considering the mechanisms by which IMiD resistance emerges, both these effects must be appraised. CRBN-dependent mechanisms of IMiD resistance, including CRBN genetic aberrations, CRBN protein loss and CRBN-substrate binding defects, are beginning to be understood. However, only a proportion of IMiD-resistant cases are related to CRBN and therefore additional mechanisms, which are currently less well described, need to be sought. These include resistance within the immune microenvironment. Here we review the existing evidence on both tumor and immune microenvironment mechanisms of resistance to IMiDs, pose important questions for future study, and consider how knowledge regarding resistance mechanism may be utilized to guide treatment decision making in the clinic.
Collapse
Affiliation(s)
- Lucia Y. Chen
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
| | - Sarah Gooding
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
9
|
Guo H, Yang J, Wang H, Liu X, Liu Y, Zhou K. Reshaping the tumor microenvironment: The versatility of immunomodulatory drugs in B-cell neoplasms. Front Immunol 2022; 13:1017990. [PMID: 36311747 PMCID: PMC9596992 DOI: 10.3389/fimmu.2022.1017990] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Immunomodulatory drugs (IMiDs) such as thalidomide, lenalidomide and pomalidomide are antitumor compounds that have direct tumoricidal activity and indirect effects mediated by multiple types of immune cells in the tumor microenvironment (TME). IMiDs have shown remarkable therapeutic efficacy in a set of B-cell neoplasms including multiple myeloma, B-cell lymphomas and chronic lymphocytic leukemia. More recently, the advent of immunotherapy has revolutionized the treatment of these B-cell neoplasms. However, the success of immunotherapy is restrained by immunosuppressive signals and dysfunctional immune cells in the TME. Due to the pleiotropic immunobiological properties, IMiDs have shown to generate synergetic effects in preclinical models when combined with monoclonal antibodies, immune checkpoint inhibitors or CAR-T cell therapy, some of which were successfully translated to the clinic and lead to improved responses for both first-line and relapsed/refractory settings. Mechanistically, despite cereblon (CRBN), an E3 ubiquitin ligase, is considered as considered as the major molecular target responsible for the antineoplastic activities of IMiDs, the exact mechanisms of action for IMiDs-based TME re-education remain largely unknown. This review presents an overview of IMiDs in regulation of immune cell function and their utilization in potentiating efficacy of immunotherapies across multiple types of B-cell neoplasms.
Collapse
Affiliation(s)
| | | | | | | | | | - Keshu Zhou
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
10
|
Yamamoto J, Ito T, Yamaguchi Y, Handa H. Discovery of CRBN as a target of thalidomide: a breakthrough for progress in the development of protein degraders. Chem Soc Rev 2022; 51:6234-6250. [PMID: 35796627 DOI: 10.1039/d2cs00116k] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Progress in strategies aimed at breaking down therapeutic target proteins has led to a paradigm shift in drug discovery. Thalidomide and its derivatives are the only protein degraders currently used in clinical practice. Our understanding of the molecular mechanism of action of thalidomide and its derivatives has advanced dramatically since the identification of cereblon (CRBN) as their direct target. The binding of thalidomide derivatives to CRBN, a substrate recognition receptor for Cullin 4 RING E3 ubiquitin ligase (CRL4), induces the recruitment of non-native substrates to CRL4CRBN and their subsequent degradation. This discovery was a breakthrough in the current rapid development of protein-degrading agents because clarification of the mechanism of action of thalidomide derivatives has demonstrated the clinical value of these compounds. This review provides an overview of the mechanism of action of thalidomide and its derivatives and describes perspectives for protein degraders.
Collapse
Affiliation(s)
- Junichi Yamamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Takumi Ito
- Institute of Medical Science, Tokyo Medical University, Shinjuku, Tokyo 160-8402, Japan
| | - Yuki Yamaguchi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Hiroshi Handa
- Center for Future Medical Research, Tokyo Medical University, Shinjuku, Tokyo 160-8402, Japan.
| |
Collapse
|
11
|
Zou Y, Liu B, Li L, Yin Q, Tang J, Jing Z, Huang X, Zhu X, Chi T. IKZF3 deficiency potentiates chimeric antigen receptor T cells targeting solid tumors. Cancer Lett 2022; 524:121-130. [PMID: 34687790 DOI: 10.1016/j.canlet.2021.10.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/28/2021] [Accepted: 10/13/2021] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has been successful in treating hematological malignancy, but solid tumors remain refractory. Here, we demonstrated that knocking out transcription factor IKZF3 in HER2-specific CAR T cells targeting breast cancer cells did not affect CAR expression or CAR T cell differentiation, but markedly enhanced killing of the cancer cells in vitro and in a xenograft model, which was associated with increased T cell activation and proliferation. Furthermore, IKZF3 KO had similar effects on the CD133-specific CAR T cells targeting glioblastoma cells. AlphaLISA and RNA-seq analyses indicate that IKZF3 KO increased the expression of genes involved in cytokine signaling, chemotaxis and cytotoxicity. Our results suggest a general strategy for enhancing CAR T efficacy on solid tumors.
Collapse
Affiliation(s)
- Yan Zou
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Bo Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Long Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Qinan Yin
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, Henan, 471000, China
| | - Jiaxing Tang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Zhengyu Jing
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xingxu Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xuekai Zhu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| | - Tian Chi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Department of Immunobiology, Yale University Medical School, New Haven, CT, USA.
| |
Collapse
|
12
|
Tawfik EA, Aldrak NA, Albrahim SH, Alzahrani DA, Alfassam HA, Alkoblan SM, Almalik AM, Chen KS, Abou-Khalil R, Shah K, Zaidan NM. Immunotherapy in hematological malignancies: recent advances and open questions. Immunotherapy 2021; 13:1215-1229. [PMID: 34498496 DOI: 10.2217/imt-2021-0065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Over recent years, tremendous advances in immunotherapy approaches have been observed, generating significant clinical progress. Cancer immunotherapy has been shown, in different types of blood cancers, to improve the overall survival of patients. Immunotherapy treatment of hematopoietic malignancies is a newly growing field that has been accelerating over the past years. Several US FDA approved drugs and cell-based therapies are being exploited in the late stage of clinical trials. This review attempt to highlight and discuss the numerous innovative immunotherapy approaches of hematopoietic malignancy ranging from nonmyeloablative transplantation, T-cell immunotherapy, natural killer cells and immune agonist to monoclonal antibodies and vaccination. In addition, a brief discussion on the future advances and accomplishments required to counterpart the current immunotherapeutic approaches for hematopoietic malignancies were also highlighted.
Collapse
Affiliation(s)
- Essam A Tawfik
- Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia.,National Center for Pharmaceutical Technology, Life Science & Environment Research Institute, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia
| | - Norah A Aldrak
- Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia
| | - Shahad H Albrahim
- Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia
| | - Dunia A Alzahrani
- National Center for Pharmaceutical Technology, Life Science & Environment Research Institute, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia
| | - Haya A Alfassam
- Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia
| | - Samar M Alkoblan
- Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia
| | - Abdulaziz M Almalik
- Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia.,National Center for Pharmaceutical Technology, Life Science & Environment Research Institute, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia
| | - Kok-Siong Chen
- BWH Center of Excellence for Biomedicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Center for Stem Cell Therapeutics & Imaging, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Rana Abou-Khalil
- Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia
| | - Khalid Shah
- BWH Center of Excellence for Biomedicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Center for Stem Cell Therapeutics & Imaging, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Nada M Zaidan
- Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science & Technology (KACST), PO Box 6086, Riyadh, 11442, Saudi Arabia
| |
Collapse
|
13
|
Tuscano JM, Poh C, Kaesberg P, Luxardi G, Merleev A, Marusina A, Brunson A, Rosenberg A, Jonas B, Maverakis E. Long-term Follow-up and Correlative Analysis of Two Phase II Trials of Rituximab and Lenalidomide Followed by Continuous Lenalidomide in Untreated and Relapsed/Refractory Indolent Lymphoma. Clin Cancer Res 2021; 27:4726-4736. [PMID: 34088724 PMCID: PMC9262142 DOI: 10.1158/1078-0432.ccr-20-4622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/01/2021] [Accepted: 05/27/2021] [Indexed: 01/26/2023]
Abstract
PURPOSE Rituximab and lenalidomide are effective for previously untreated and relapsed/refractory (R/R) indolent non-Hodgkin lymphoma (iNHL). However, long-term survival and predictive biomarkers are not well described. PATIENTS AND METHODS We conducted two phase II open-label trials involving 60 patients with previously untreated and R/R advanced-stage iNHL. Patients received lenalidomide and rituximab induction followed by continuous lenalidomide until disease progression or unacceptable toxicity. The primary endpoint was overall response rate (ORR). Correlative studies included plasma cytokine monitoring, flow cytometry of peripheral blood mononuclear cells (PBMC; days 0, 15, 30, and 60), and RNA sequencing (RNA-seq) of pretreatment tumor biopsies. RESULTS At a median follow-up of 63 months for previously untreated and 100 months for R/R, ORR was 82% for both. The 11 R/R patients who achieved complete remission remained in continuous remission for 16 to 141 months, thereafter. Median overall survival (OS) was not reached in the previously untreated and was 140 months (95% confidence interval, 53.4-140) in the R/R group. A mixed-effects linear regression model identified significant associations between Granzyme B+ (GranB+) CD8+ T cells and long-term complete response (LTCR; P = 5.3e-4). Furthermore, prior to start of therapy, treatment response could be predicted by B-cell and GranB+ CD8+ T-cell levels (% total lymphocytes). CONCLUSIONS Rituximab plus lenalidomide followed by continuous lenalidomide is effective with manageable toxicity in patients with previously untreated and R/R iNHL. This regimen produces durable remissions, even in heavily pretreated patients, with some lasting greater than 10 years. GranB+ CD8+ T cells, B cells, and plasma IFNγ allowed prediction of LTCR but need validation in larger trials.
Collapse
Affiliation(s)
- Joseph M. Tuscano
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA USA,Veterans Administration Northern California Healthcare System, Sacramento, CA, USA
| | - Christina Poh
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA USA,University of Washington, Division of Medical Oncology, Seattle, WA USA
| | - Paul Kaesberg
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA USA
| | - Guillaume Luxardi
- University of California, Davis, Department of Dermatology, Sacramento, CA USA
| | - Alexander Merleev
- University of California, Davis, Department of Dermatology, Sacramento, CA USA
| | - Alina Marusina
- University of California, Davis, Department of Dermatology, Sacramento, CA USA
| | - Ann Brunson
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA USA
| | - Aaron Rosenberg
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA USA
| | - Brian Jonas
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA USA
| | - Emanual Maverakis
- University of California, Davis, Department of Dermatology, Sacramento, CA USA
| |
Collapse
|
14
|
Thalidomide alleviates neuropathic pain through microglial IL-10/β-endorphin signaling pathway. Biochem Pharmacol 2021; 192:114727. [PMID: 34390739 DOI: 10.1016/j.bcp.2021.114727] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 01/08/2023]
Abstract
Thalidomide is an antiinflammatory, antiangiogenic and immunomodulatory agent which has been used for the treatment of erythema nodosum leprosum and multiple myeloma. It has also been employed in treating complex regional pain syndromes. The current study aimed to reveal the molecular mechanisms underlying thalidomide-induced pain antihypersensitive effects in neuropathic pain. Thalidomide gavage, but not its more potent analogs lenalidomide and pomalidomide, inhibited mechanical allodynia and thermal hyperalgesia in neuropathic pain rats induced by tight ligation of spinal nerves, with ED50 values of 44.9 and 23.5 mg/kg, and Emax values of 74% and 84% MPE respectively. Intrathecal injection of thalidomide also inhibited mechanical allodynia and thermal hyperalgesia in neuropathic pain. Treatment with thalidomide, lenalidomide and pomalidomide reduced peripheral nerve injury-induced proinflammatory cytokines (TNFα, IL-1β and IL-6) in the ipsilateral spinal cords of neuropathic rats and LPS-treated primary microglial cells. In contrast, treatment with thalidomide, but not lenalidomide or pomalidomide, stimulated spinal expressions of IL-10 and β-endorphin in neuropathic rats. Particularly, thalidomide specifically stimulated IL-10 and β-endorphin expressions in microglia but not astrocytes or neurons. Furthermore, pretreatment with the IL-10 antibody blocked upregulation of β-endorphin in neuropathic rats and cultured microglial cells, whereas it did not restore thalidomide-induced downregulation of proinflammatory cytokine expression. Importantly, pretreatment with intrathecal injection of the microglial metabolic inhibitor minocycline, IL-10 antibody, β-endorphin antiserum, and preferred or selective μ-opioid receptor antagonist naloxone or CTAP entirely blocked thalidomide gavage-induced mechanical antiallodynia. Our results demonstrate that thalidomide, but not lenalidomide or pomalidomide, alleviates neuropathic pain, which is mediated by upregulation of spinal microglial IL-10/β-endorphin expression, rather than downregulation of TNFα expression.
Collapse
|
15
|
Developing next generation immunomodulatory drugs and their combinations in multiple myeloma. Oncotarget 2021; 12:1555-1563. [PMID: 34316334 PMCID: PMC8310669 DOI: 10.18632/oncotarget.27973] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Multiple Myeloma (MM) is an incurable malignancy with current treatment choices primarily comprising combination regimens implemented with a risk-adapted approach. Cereblon (CRBN)-targeting immunomodulatory agents (IMiDs®) lenalidomide (LEN) and pomalidomide (POM) play a central role in combination regimens due to their pleiotropic antitumor/immunomodulatory mechanisms that synergize with many anti-myeloma approved or developmental agents. Currently, more potent next generation cereblon E3 ligase modulators (CELMoDs®) - iberdomide (IBER) and CC-92480 are in clinical development. With an expanding number of active agents/therapeutic modalities and a myriad of combinatorial possibilities, physicians and drug developers share an opportunity and challenge to combine and sequence therapies to maximize long-term patient benefit. Understanding drug mechanisms and their application in combination settings as well as the unique disease biology considerations from newly diagnosed (NDMM), relapsed/refractory (RRMM), and maintenance settings will be vital to guide the development of future MM therapies centered on a backbone of IMiD or CELMoD agents. Key aspects of drug activity are critical to consider while evaluating potential combinations: direct antitumor effects, indirect antitumor cytotoxicity, immune surveillance, and adverse side effects. In addition, the treatment journey from NDMM to early and late MM relapses are connected to genomic and immune changes associated with disease progression and acquisition of resistance mechanisms. Based on the types of combinations used and the goals of therapy, insights into mechanisms of drug activity and resistance may inform treatment decisions for patients with MM. Here we focus on the evolving understanding of the molecular mechanisms of CRBN-binding drugs and how they can be differentiated and suggest a strategic framework to optimize efficacy and safety of combinations using these agents.
Collapse
|
16
|
Abstract
Bispecific T-cell recruiting antibodies are emerging as a potent immunotherapeutic class in the treatment of B-cell malignancies and act by simultaneously targeting antigens on T-cells and malignant cells to effect tumor cell death. Glofitamab is a novel full-length IgG-like CD20-CD3 bispecific with a unique 2:1 configuration that provides an extended half-life and superior CD20 binding. Phase 1 monotherapy and combination data demonstrate clear activity in heavily treated aggressive and indolent B-cell lymphoma, including >50% complete responses at the recommended phase 2 dose. In this review, we provide an overview of the structure, mechanism of action and pharmacokinetics of glofitamab. Available efficacy and safety data from ongoing clinical trials are also presented. Glofitamab appears to be a welcome addition to the treatment possibilities for patients with B-cell lymphomas who otherwise have limited therapeutic options. The current data are sufficient to evaluate its role in combination and in earlier lines of therapy.
Collapse
Affiliation(s)
- Adrian Minson
- Clinical Haematology, Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Michael Dickinson
- Clinical Haematology, Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
17
|
Wang Z, Zhou G, Risu N, Fu J, Zou Y, Tang J, Li L, Liu H, Liu Q, Zhu X. Lenalidomide Enhances CAR-T Cell Activity Against Solid Tumor Cells. Cell Transplant 2021; 29:963689720920825. [PMID: 32967454 PMCID: PMC7784582 DOI: 10.1177/0963689720920825] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell immunotherapy still faces many challenges in the treatment of solid tumors, one of which is T-cell dysfunction or exhaustion. Immunomodulator lenalidomide may improve CAR T-cell function. In this study, the effects of lenalidomide on CAR T-cell functions (cytotoxicity, cytokine secretion, and cell proliferation) were investigated. Two different CAR T cells (CD133-specific CAR and HER2-specific CAR) were prepared, and the corresponding target cells including human glioma cell line U251 CD133-OE that overexpress CD133 and human breast cancer cell line MDA-MB-453 were used for functional assay. We found that lenalidomide promoted the killing of U251 CD133-OE by CD133-CAR T cells, the cytokine secretion, and the proliferation of CD133-CAR T cells. Lenalidomide also enhanced the cytotoxicity against MDA-MB-453 and the cytokine secretion of HER2-CAR T cells but did not affect their proliferation significantly. Furthermore, lenalidomide may regulate the function of CAR T cells by inducing the degradation of transcription factors Ikaros and Aiolos.
Collapse
Affiliation(s)
- Zhixiong Wang
- School of Medical Instrument and Food Engineering, 47863University of Shanghai for Science and Technology, China.,Ma'anshan University, China.,Division of Health Science, Graduate School of Medicine, 13013Osaka University, Japan
| | - Guomin Zhou
- School of Medical Instrument and Food Engineering, 47863University of Shanghai for Science and Technology, China
| | - Na Risu
- Division of Health Science, Graduate School of Medicine, 13013Osaka University, Japan
| | - Jiayu Fu
- Division of Health Science, Graduate School of Medicine, 13013Osaka University, Japan
| | - Yan Zou
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), 387433ShanghaiTech University, China
| | - Jiaxing Tang
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), 387433ShanghaiTech University, China
| | - Long Li
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), 387433ShanghaiTech University, China
| | - Hui Liu
- School of Medical Instrument and Food Engineering, 47863University of Shanghai for Science and Technology, China
| | - Qian Liu
- School of Medical Instrument and Food Engineering, 47863University of Shanghai for Science and Technology, China
| | - Xuekai Zhu
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), 387433ShanghaiTech University, China
| |
Collapse
|
18
|
A randomized phase 2 trial of pomalidomide in subjects failing prior therapy for chronic graft-versus-host disease. Blood 2021; 137:896-907. [PMID: 32976576 DOI: 10.1182/blood.2020006892] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Steroid-refractory chronic graft-versus-host disease (cGVHD) is a therapeutic challenge. Sclerotic skin manifestations are especially difficult to treat. We conducted a randomized phase 2 clinical trial (#NCT01688466) to determine the safety, efficacy, and preferred dose of pomalidomide in persons with moderate to severe cGVHD unresponsive to corticosteroids and/or subsequent lines of therapy. Thirty-four subjects were randomized to receive pomalidomide 0.5 mg per day orally (n = 17; low-dose cohort) or 2 mg per day at a starting dose of 0.5 mg per day increasing to 2 mg per day over 6 weeks (n = 17; high-dose cohort). The primary endpoint was overall response rate (ORR) at 6 months according to the 2005 National Institutes of Health cGVHD Response Criteria. Thirty-two patients had severe sclerotic skin and received a median of 5 (range, 2-10) previous systemic therapies. ORR was 47% (95% confidence interval, 30-65) in the intention-to-treat analyses. All were partial responses, with no difference in ORR between the cohorts. ORR was 67% (45%-84%) in the 24 evaluable subjects at 6 months. Nine had improvement in National Institutes of Health joint/fascia scores (P = .018). Median change from the baseline in body surface area involvement of skin cGVHD was -7.5% (-10% to 35%; P = .002). The most frequent adverse events were lymphopenia, infection, and fatigue. Eight subjects in the high-dose cohort had dose decreases because of adverse events. There was 1 death in the low-dose cohort from bacterial pneumonia. Our data indicate antifibrotic effects of pomalidomide and possible association with increases in concentrations of blood regulatory T-cell and interleukin-2. Pomalidomide 0.5 mg per day is a safe and effective therapy for advanced corticosteroid-refractory cGVHD.
Collapse
|
19
|
Fangusaro J, Cefalo MG, Garré ML, Marshall LV, Massimino M, Benettaib B, Biserna N, Poon J, Quan J, Conlin E, Lewandowski J, Simcock M, Jeste N, Hargrave DR, Doz F, Warren KE. Phase 2 Study of Pomalidomide (CC-4047) Monotherapy for Children and Young Adults With Recurrent or Progressive Primary Brain Tumors. Front Oncol 2021; 11:660892. [PMID: 34168987 PMCID: PMC8218626 DOI: 10.3389/fonc.2021.660892] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Treatment of recurrent primary pediatric brain tumors remains a major challenge, with most children succumbing to their disease. We conducted a prospective phase 2 study investigating the safety and efficacy of pomalidomide (POM) in children and young adults with recurrent and progressive primary brain tumors. Methods Patients with recurrent and progressive high-grade glioma (HGG), diffuse intrinsic pontine glioma (DIPG), ependymoma, or medulloblastoma received POM 2.6 mg/m2/day (the recommended phase 2 dose [RP2D]) on days 1-21 of a 28-day cycle. A Simon's Optimal 2-stage design was used to determine efficacy. Primary endpoints included objective response (OR) and long-term stable disease (LTSD) rates. Secondary endpoints included duration of response, progression-free survival (PFS), overall survival (OS), and safety. Results 46 patients were evaluable for response (HGG, n = 19; DIPG, ependymoma, and medulloblastoma, n = 9 each). Two patients with HGG achieved OR or LTSD (10.5% [95% CI, 1.3%-33.1%]; 1 partial response and 1 LTSD) and 1 patient with ependymoma had LTSD (11.1% [95% CI, 0.3%-48.2%]). There were no ORs or LTSD in the DIPG or medulloblastoma cohorts. The median PFS for patients with HGG, DIPG, ependymoma, and medulloblastoma was 7.86, 11.29, 8.43, and 8.43 weeks, respectively. Median OS was 5.06, 3.78, 12.02, and 11.60 months, respectively. Neutropenia was the most common grade 3/4 adverse event. Conclusions Treatment with POM monotherapy did not meet the primary measure of success in any cohort. Future studies are needed to evaluate if POM would show efficacy in tumors with specific molecular signatures or in combination with other anticancer agents. Clinical Trial Registration ClinicalTrials.gov, identifier NCT03257631; EudraCT, identifier 2016-002903-25.
Collapse
Affiliation(s)
- Jason Fangusaro
- Department of Pediatrics, Children's Healthcare of Atlanta and Aflac Cancer Center at Emory University Medical School, Atlanta, GA, United States
| | - Maria Giuseppina Cefalo
- Department of Hematology/Oncology and Stem Cell Transplantation, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Lynley V Marshall
- Children and Young People's Unit, The Royal Marsden Hospital and The Institute of Cancer Research, London, United Kingdom
| | - Maura Massimino
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Noha Biserna
- Bristol Myers Squibb, Princeton, NJ, United States
| | | | - Jackie Quan
- Bristol Myers Squibb, Princeton, NJ, United States
| | - Erin Conlin
- Bristol Myers Squibb, Princeton, NJ, United States
| | | | | | - Neelum Jeste
- Bristol Myers Squibb, Princeton, NJ, United States
| | - Darren R Hargrave
- Pediatric Oncology Unit, UCL Great Ormond Street Hospital for Children, London, United Kingdom
| | - François Doz
- Department of Pediatric Oncology, Institut Curie and University of Paris, Paris, France
| | - Katherine E Warren
- National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
20
|
Immunomodulatory drugs suppress Th1-inducing ability of dendritic cells but enhance Th2-mediated allergic responses. Blood Adv 2021; 4:3572-3585. [PMID: 32761232 DOI: 10.1182/bloodadvances.2019001410] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/01/2020] [Indexed: 11/20/2022] Open
Abstract
Immunomodulatory drugs (IMiDs), lenalidomide and pomalidomide, are widely used treatments for multiple myeloma; however, they occasionally lead to episodes of itchy skin and rashes. Here, we analyzed the effects of IMiDs on human myeloid dendritic cells (mDCs) as major regulators of Th1 or Th2 responses and the role they play in allergy. We found that lenalidomide and pomalidomide used at clinical concentrations did not affect the survival or CD86 and OX40-ligand expression of blood mDCs in response to lipopolysaccharide (LPS) and thymic stromal lymphopoietin (TSLP) stimulation. Both lenalidomide and pomalidomide dose-dependently inhibited interleukin-12 (IL-12) and TNF production and STAT4 expression, and enhanced IL-10 production in response to LPS. When stimulated with TSLP, both IMiDs significantly enhanced CCL17 production and STAT6 and IRF4 expression and promoted memory Th2-cell responses. In 46 myeloma patients, serum CCL17 levels at the onset of lenalidomide-associated rash were significantly higher than those without rashes during lenalidomide treatment and those before treatment. Furthermore, serum CCL17 levels in patients who achieved a very good partial response (VGPR) were significantly higher compared with a less than VGPR during lenalidomide treatment. The median time to next treatment was significantly longer in lenalidomide-treated patients with rashes than those without. Collectively, IMiDs suppressed the Th1-inducing capacity of DCs, instead promoting a Th2 response. Thus, the lenalidomide-associated rashes might be a result of an allergic response driven by Th2-axis activation. Our findings suggest clinical efficacy and rashes as a side effect of IMiDs are inextricably linked through immunostimulation.
Collapse
|
21
|
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
|
22
|
Effective degradation of EGFR L858R+T790M mutant proteins by CRBN-based PROTACs through both proteosome and autophagy/lysosome degradation systems. Eur J Med Chem 2021; 218:113328. [PMID: 33773286 DOI: 10.1016/j.ejmech.2021.113328] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/21/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023]
Abstract
Targeted therapy of treating patients with specific tyrosine kinase inhibitors (TKIs) is currently the standard care for epidermal growth factor receptor (EGFR) mutant non-small cell lung cancer. However, the inevitably developed drug resistance in patients to EGFR TKIs is the biggest obstacle for cancer targeted therapy. About 60% of drug resistance to the 1st generation of EGFR TKIs was resulted from an acquired T790M mutation in the kinase domain of EGFR protein. Proteolysis targeting chimera (PROTAC) is a lately-developed technology to target point of interest proteins for degradation. Because EGFR-mutant lung cancers are highly dependent on EGFR proteins, designing specific PROTAC molecules to degrade EGFR proteins from cancer cells provides a very promising strategy to treat such patients and eradicate drug resistance. Currently, there is no cereblon (CRBN)-based PROTAC reported able to degrade T790M-containing EGFR resistant proteins. In this study, we synthesized two novel CRBN-based EGFR PROTACs, SIAIS125 and SIAIS126, based on EGFR inhibitor canertinib and cereblon ligand pomalidomide. These two degraders displayed potent and selective antitumor activities in EGFR TKI resistant lung cancer cells. Firstly, they could selectively degrade EGFRL858R+T790M resistant proteins in H1975 cells at the concentration of 30-50 nM, and EGFREx19del proteins in PC9 cells. But they did not degrade EGFREx19del+T790M mutant proteins in PC9Brca1 cells or wild type EGFR in A549 lung cancer cells. They could also selectively inhibit the growth of EGFR mutant lung cancer cells but not that of normal cells or A549 cells. Secondly, the degradation of EGFRL858R+T790M proteins was long lasting up to 72 h. Thirdly, these degraders displayed better inhibition of EGFR phosphorylation in H1975 cells and PC9Brca1 cells comparing to canertinib. Finally, these degraders could also induce significant apoptosis and cell cycles arrest in H1975 cells. Pre-incubation with canertinib, pomalidomide or ubiquitination inhibitor MLN4924 totally blocked EGFR degradation by PROTACs. Mechanistic studies showed that PROTAC could induce autophagy in lung cancer cells. PROTAC-induced EGFR degradation acted through both ubiquitin/proteosome system and ubiquitin/autophagy/lysosome system. Elevating autophagy activities enhanced EGFR degradation and cell apoptosis induced by PROTACs. Our research not only offered a novel PROTAC tool to target EGFR TKI drug resistance in lung cancer, but also firstly demonstrated that the involvement of autophagy/lysosome system in PROTAC- mediated target protein degradation.
Collapse
|
23
|
Brayshaw LL, Martinez-Fleites C, Athanasopoulos T, Southgate T, Jespers L, Herring C. The role of small molecules in cell and gene therapy. RSC Med Chem 2021; 12:330-352. [PMID: 34046619 PMCID: PMC8130622 DOI: 10.1039/d0md00221f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/25/2020] [Indexed: 01/22/2023] Open
Abstract
Cell and gene therapies have achieved impressive results in the treatment of rare genetic diseases using gene corrected stem cells and haematological cancers using chimeric antigen receptor T cells. However, these two fields face significant challenges such as demonstrating long-term efficacy and safety, and achieving cost-effective, scalable manufacturing processes. The use of small molecules is a key approach to overcome these barriers and can benefit cell and gene therapies at multiple stages of their lifecycle. For example, small molecules can be used to optimise viral vector production during manufacturing or used in the clinic to enhance the resistance of T cell therapies to the immunosuppressive tumour microenvironment. Here, we review current uses of small molecules in cell and gene therapy and highlight opportunities for medicinal chemists to further consolidate the success of cell and gene therapies.
Collapse
Affiliation(s)
- Lewis L Brayshaw
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Carlos Martinez-Fleites
- Protein Degradation Group, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Takis Athanasopoulos
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Thomas Southgate
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Laurent Jespers
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| | - Christopher Herring
- Cell & Gene Therapy Discovery Research, Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
| |
Collapse
|
24
|
Shrestha P, Davis DA, Jaeger HK, Stream A, Aisabor AI, Yarchoan R. Pomalidomide restores immune recognition of primary effusion lymphoma through upregulation of ICAM-1 and B7-2. PLoS Pathog 2021; 17:e1009091. [PMID: 33411730 PMCID: PMC7817053 DOI: 10.1371/journal.ppat.1009091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 01/20/2021] [Accepted: 10/23/2020] [Indexed: 01/08/2023] Open
Abstract
Pomalidomide (Pom) is an immunomodulatory drug that has efficacy against Kaposi’s sarcoma, a tumor caused by Kaposi’s sarcoma-associated herpesvirus (KSHV). Pom also induces direct cytotoxicity in primary effusion lymphoma (PEL), a B-cell malignancy caused by KSHV, in part through downregulation of IRF4, cMyc, and CK1α as a result of its interaction with cereblon, a cellular E3 ubiquitin ligase. Additionally, Pom can reverse KSHV-induced downregulation of MHCI and co-stimulatory immune surface molecules ICAM-1 and B7-2 on PELs. Here, we show for the first time that Pom-induced increases in ICAM-1 and B7-2 on PEL cells lead to an increase in both T-cell activation and NK-mediated cytotoxicity against PEL. The increase in T-cell activation can be prevented by blocking ICAM-1 and/or B7-2 on the PEL cell surface, suggesting that both ICAM-1 and B7-2 are important for T-cell co-stimulation by PELs. To gain mechanistic insights into Pom’s effects on surface markers, we generated Pom-resistant (PomR) PEL cells, which showed about 90% reduction in cereblon protein level and only minimal changes in IRF4 and cMyc upon Pom treatment. Pom no longer upregulated ICAM-1 and B7-2 on the surface of PomR cells, nor did it increase T-cell and NK-cell activation. Cereblon-knockout cells behaved similarly to the pomR cells upon Pom-treatment, suggesting that Pom’s interaction with cereblon is necessary for these effects. Further mechanistic studies revealed PI3K signaling pathway as being important for Pom-induced increases in these molecules. These observations provide a rationale for the study of Pom as therapy in treating PEL and other KSHV-associated tumors. Primary effusion lymphoma (PEL) is an aggressive B-cell lymphoma caused by Kaposi’s sarcoma-associated herpesvirus (KSHV). KSHV encodes various genes that enable infected cells to evade recognition and elimination by the immune system. PEL cells are poorly recognized by T-cells and NK cells, partly due to KSHV-induced downregulation of immune stimulatory surface molecules ICAM-1 and B7-2. We previously found that a cereblon-binding immunomodulatory drug pomalidomide (Pom) can restore the levels of these markers on PELs. Here, we show that the increases in ICAM-1 and B7-2 induced by Pom leads to a functional increase in the recognition and killing of PELs by both T-cells and NK cells. Further, exposure of both the PEL cells and T-cells to Pom lead to an even higher T-cell stimulation providing strong evidence that Pom could help PEL patients by providing specific immune-stimulatory effect. We further perform mechanistic studies and show that Pom’s cellular binding partner cereblon as well as the PI3K pathway are important for Pom-mediated increases in these surface markers.
Collapse
Affiliation(s)
- Prabha Shrestha
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - David A. Davis
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Hannah K. Jaeger
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Alexandra Stream
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Ashley I. Aisabor
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
25
|
Bansal R, Reshef R. Revving the CAR - Combination strategies to enhance CAR T cell effectiveness. Blood Rev 2021; 45:100695. [PMID: 32402724 DOI: 10.1016/j.blre.2020.100695] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/22/2020] [Accepted: 04/11/2020] [Indexed: 12/14/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is currently approved for treatment of refractory B-cell malignancies. Response rates in these diseases are impressive by historical standards, but most patients do not have a durable response and there remains room for improvement. To date, CAR T cell activity has been even more limited in solid malignancies. These limitations are thought to be due to several pathways of resistance to CAR T cells, including cell-intrinsic mechanisms and the immunosuppressive tumor microenvironment. In this review, we discuss current experimental strategies that combine small molecules and monoclonal antibodies with CAR T cells to overcome these resistance mechanisms. We describe the biological rationale, pre-clinical data and clinical trials in progress that test the efficacy and safety of these combinations.
Collapse
Affiliation(s)
- Rajat Bansal
- Division of Hematology/Oncology, Columbia University Irving Medical Center, 177 Ft. Washington Ave, Floor: 6GN-435, New York, NY 10032, USA.
| | - Ran Reshef
- Division of Hematology/Oncology, Columbia University Irving Medical Center, 630 W. 168(th) Street Mailbox 127, New York, NY 10032, USA.
| |
Collapse
|
26
|
Soekojo CY, Ooi M, de Mel S, Chng WJ. Immunotherapy in Multiple Myeloma. Cells 2020; 9:E601. [PMID: 32138182 PMCID: PMC7140529 DOI: 10.3390/cells9030601] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma is a complex disease and immune dysfunction has been known to play an important role in the disease pathogenesis, progression, and drug resistance. Recent efforts in drug development have been focused on immunotherapies to modify the MM disease process. Here, we summarize the emerging immunotherapies in the MM treatment landscape.
Collapse
Affiliation(s)
| | | | | | - Wee Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore; (C.Y.S.); (M.O.); (S.d.M.)
| |
Collapse
|
27
|
Suzuki K, Nishiwaki K, Gunji T, Katori M, Masuoka H, Yano S. Elevated eosinophil level predicted long time to next treatment in relapsed or refractory myeloma patients treated with lenalidomide. Cancer Med 2020; 9:1694-1702. [PMID: 31950647 PMCID: PMC7050101 DOI: 10.1002/cam4.2828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 12/19/2022] Open
Abstract
Lenalidomide is an immunomodulatory drug that is administered commonly in patients with relapsed or refractory multiple myeloma (RRMM). Eosinophils have immunological functions, for instance, in allergic diseases and asthma. The purpose of this study was to investigate the clinical significance of elevated eosinophil levels in patients with RRMM treated with lenalidomide. A total of 59 patients were included. Elevated eosinophil level was defined as an increase in the eosinophil count of ≥250/µL from the eosinophil count on day 1 during the first cycle. The percentage of patients with elevated eosinophil levels was 22.0%. The overall response ratio in the elevated eosinophil group and nonelevated eosinophil group was 84.6% and 63.0% (P = .189), respectively. The median time to next treatment (TTNT) in the elevated eosinophil group was significantly longer than that in the nonelevated group (40.3 months vs 8.4 months; P = .017). Additionally, TTNT in the elevated eosinophil group with partial response (PR) or better was significantly longer than that in the nonelevated eosinophil group with PR or better (40.3 months vs 11.9 months; P = .021). We concluded that elevated eosinophil levels were frequently observed and might predict a longer TTNT in patients with RRMM treated with lenalidomide.
Collapse
Affiliation(s)
- Kazuhito Suzuki
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Chiba, Japan
| | - Kaichi Nishiwaki
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Chiba, Japan
| | - Tadahiro Gunji
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Mitsuji Katori
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Chiba, Japan
| | - Hidekazu Masuoka
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Chiba, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
28
|
A New Salvage Regimen for Aggressive Lymphomas Based on Gemcitabine, Rituximab, and Oxaliplatin Followed by Lenalidomide (GROC-Rev). CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:776-783. [DOI: 10.1016/j.clml.2019.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 06/22/2019] [Accepted: 07/01/2019] [Indexed: 01/13/2023]
|
29
|
Kibata K, Ito T, Inaba M, Tanaka A, Iwata R, Inagaki-Katashiba N, Phan V, Satake A, Nomura S. The immunomodulatory-drug, lenalidomide, sustains and enhances interferon-α production by human plasmacytoid dendritic cells. J Blood Med 2019; 10:217-226. [PMID: 31372079 PMCID: PMC6635835 DOI: 10.2147/jbm.s206459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/31/2019] [Indexed: 11/23/2022] Open
Abstract
Background: Lenalidomide (LEN), an immunomodulatory drug (IMiD), is currently used for treatment of multiple myeloma (MM). LEN potentiates T cell and natural killer cell functions. However, the cellular and molecular mechanisms underlying the immunomodulatory effects of LEN remain unclear. We focused on the effects of LEN on human plasmacytoid dendritic cells (pDCs), which are the major source of interferon (IFN)-α in the blood and play a central role in innate immune responses. Results: We found that bortezomib, a proteasome inhibitor used to treat MM, killed pDCs but that 0.1-3 μM LEN (covering clinical plasma concentration range) did not affect pDC survival or CD86 expression. Bortezomib inhibited pDC-derived IFN-α production in a dose-dependent fashion, but 0.1-3 µM LEN sustained pDC-derived IFN-α production when stimulated with an optimal concentration of CpG-ODN 2216 (3 μM). In pDCs stimulated with a low concentration of CpG-ODN (0.1 μM), LEN enhanced IFN-α production. These results indicated that LEN, when used at a clinically relevant concentration, can potentially enhance IFN-α production by pDCs. Conclusion: Collectively, our findings unveiled a novel target of LEN and extend the repertoire of the drug's known immunomodulatory effects. These effects may explain the low incidence of herpes zoster viral infection observed during LEN treatment compared with bortezomib treatment. LEN may function as an IMiD affecting a wide array of immune cells, including pDCs, leading to amplification of a positive immune axis able to eliminate MM cells.
Collapse
Affiliation(s)
- Kayoko Kibata
- Kansai Medical University, First Department of Internal Medicine, Osaka, Japan
| | - Tomoki Ito
- Kansai Medical University, First Department of Internal Medicine, Osaka, Japan
| | - Muneo Inaba
- Kansai Medical University, First Department of Internal Medicine, Osaka, Japan
| | - Akihiro Tanaka
- Kansai Medical University, First Department of Internal Medicine, Osaka, Japan
| | - Ryoichi Iwata
- Kansai Medical University, Department of Neurosurgery, Osaka, Japan
| | | | - Vien Phan
- Kansai Medical University, First Department of Internal Medicine, Osaka, Japan
| | - Atsushi Satake
- Kansai Medical University, First Department of Internal Medicine, Osaka, Japan
| | - Shosaku Nomura
- Kansai Medical University, First Department of Internal Medicine, Osaka, Japan
| |
Collapse
|
30
|
Kannagi M, Hasegawa A, Nagano Y, Iino T, Okamura J, Suehiro Y. Maintenance of long remission in adult T-cell leukemia by Tax-targeted vaccine: A hope for disease-preventive therapy. Cancer Sci 2019; 110:849-857. [PMID: 30666755 PMCID: PMC6398881 DOI: 10.1111/cas.13948] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/27/2018] [Accepted: 01/13/2019] [Indexed: 12/19/2022] Open
Abstract
Adult T-cell leukemia/lymphoma (ATL) is an aggressive lymphoproliferative disease caused by human T-cell leukemia virus type 1 (HTLV-1). Multi-agent chemotherapy can reduce ATL cells but frequently allows relapses within a short period of time. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) following chemotherapy is now a standard therapy for ATL in Japan as it can achieve long-term remission in approximately one-third of recipient ATL patients; however, it also has a risk of treatment-related mortality. Allo-HSCT often induces HTLV-1 Tax-specific cytotoxic T cells (CTL) as well as graft-versus-host (GVH) response in ATL patients. This observation led to development of a new therapeutic vaccine to activate Tax-specific CTL, anticipating anti-ATL effects without GVH response. The newly developed Tax-DC vaccine consists of autologous dendritic cells pulsed with Tax peptides corresponding to CTL epitopes that have been identified in post-allo-HSCT ATL patients. In a pilot study of Tax-DC therapy in three ATL patients after various initial therapies, two patients survived for more than 4 years after vaccination without severe adverse effects (UMIN000011423). The Tax-DC vaccine is currently under phase I trial, showing a promising clinical outcome so far. These findings indicate the importance of patients' own HTLV-1-specific T-cell responses in maintaining remission and provide a new approach to anti-ATL immunotherapy targeting Tax. Although Tax-targeted vaccination is ineffective against Tax-negative ATL cells, it can be a safe alternative maintenance therapy for Tax-positive ATL and may be further applicable for treatment of indolent ATL or even prophylaxis of ATL development among HTLV-1-carriers.
Collapse
Affiliation(s)
- Mari Kannagi
- Department of Immunotherapeutics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsuhiko Hasegawa
- Department of Immunotherapeutics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshiko Nagano
- Department of Immunotherapeutics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tadafumi Iino
- Center for Advanced Medicine Innovation, Kyushu University, Fukuoka, Japan
| | - Jun Okamura
- Institute for Clinical Research, National Kyushu Cancer Center, Fukuoka, Japan
| | - Youko Suehiro
- Department of Hematology, National Kyushu Cancer Center, Fukuoka, Japan
| |
Collapse
|
31
|
Ma LY, Su L. Application of Lenalidomide on Diffused Large B-cell Lymphoma: Salvage, Maintenance, and Induction Treatment. Chin Med J (Engl) 2019; 131:2510-2513. [PMID: 30334543 PMCID: PMC6202603 DOI: 10.4103/0366-6999.243567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Li-Yangxue Ma
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Li Su
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
32
|
Fuchs O. Treatment of Lymphoid and Myeloid Malignancies by Immunomodulatory Drugs. Cardiovasc Hematol Disord Drug Targets 2019; 19:51-78. [PMID: 29788898 DOI: 10.2174/1871529x18666180522073855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/05/2018] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
Thalidomide and its derivatives (lenalidomide, pomalidomide, avadomide, iberdomide hydrochoride, CC-885 and CC-90009) form the family of immunomodulatory drugs (IMiDs). Lenalidomide (CC5013, Revlimid®) was approved by the US FDA and the EMA for the treatment of multiple myeloma (MM) patients, low or intermediate-1 risk transfusion-dependent myelodysplastic syndrome (MDS) with chromosome 5q deletion [del(5q)] and relapsed and/or refractory mantle cell lymphoma following bortezomib. Lenalidomide has also been studied in clinical trials and has shown promising activity in chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL). Lenalidomide has anti-inflammatory effects and inhibits angiogenesis. Pomalidomide (CC4047, Imnovid® [EU], Pomalyst® [USA]) was approved for advanced MM insensitive to bortezomib and lenalidomide. Other IMiDs are in phases 1 and 2 of clinical trials. Cereblon (CRBN) seems to have an important role in IMiDs action in both lymphoid and myeloid hematological malignancies. Cereblon acts as the substrate receptor of a cullin-4 really interesting new gene (RING) E3 ubiquitin ligase CRL4CRBN. This E3 ubiquitin ligase in the absence of lenalidomide ubiquitinates CRBN itself and the other components of CRL4CRBN complex. Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1α (CK1α) and marks them for degradation in proteasomes. Both these transcription factors (IKZF1 and IKZF3) stimulate proliferation of MM cells and inhibit T cells. Low CRBN level was connected with insensitivity of MM cells to lenalidomide. Lenalidomide decreases expression of protein argonaute-2, which binds to cereblon. Argonaute-2 seems to be an important drug target against IMiDs resistance in MM cells. Lenalidomide decreases also basigin and monocarboxylate transporter 1 in MM cells. MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. The CK1α gene (CSNK1A1) is located on 5q32 in commonly deleted region (CDR) in del(5q) MDS. Inhibition of CK1α sensitizes del(5q) MDS cells to lenalidomide. CK1α mediates also survival of malignant plasma cells in MM. Though, inhibition of CK1α is a potential novel therapy not only in del(5q) MDS but also in MM. High level of full length CRBN mRNA in mononuclear cells of bone marrow and of peripheral blood seems to be necessary for successful therapy of del(5q) MDS with lenalidomide. While transfusion independence (TI) after lenalidomide treatment is more than 60% in MDS patients with del(5q), only 25% TI and substantially shorter duration of response with occurrence of neutropenia and thrombocytopenia were achieved in lower risk MDS patients with normal karyotype treated with lenalidomide. Shortage of the biomarkers for lenalidomide response in these MDS patients is the main problem up to now.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| |
Collapse
|
33
|
Bassi ZI, Fillmore MC, Miah AH, Chapman TD, Maller C, Roberts EJ, Davis LC, Lewis DE, Galwey NW, Waddington KE, Parravicini V, Macmillan-Jones AL, Gongora C, Humphreys PG, Churcher I, Prinjha RK, Tough DF. Modulating PCAF/GCN5 Immune Cell Function through a PROTAC Approach. ACS Chem Biol 2018; 13:2862-2867. [PMID: 30200762 DOI: 10.1021/acschembio.8b00705] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
P300/CBP-associated factor (PCAF) and general control nonderepressible 5 (GCN5) are closely related epigenetic proteins, each containing an acetyltransferase domain and a bromodomain. Consistent with reported roles for these proteins in immune function, we find that PCAF-deficient macrophages exhibit a markedly reduced ability to produce cytokines upon stimulation with lipopolysaccharide (LPS). Investigating the potential to target this pathway pharmacologically, we show that chemical inhibition of the PCAF/GCN5 bromodomains is insufficient to recapitulate the diminished inflammatory response of PCAF-deficient immune cells. However, by generating the first PCAF/GCN5 proteolysis targeting chimera (PROTAC), we identify small molecules able to degrade PCAF/GCN5 and to potently modulate the expression of multiple inflammatory mediators in LPS-stimulated macrophages and dendritic cells. Our data illustrate the power of the PROTAC approach in the context of multidomain proteins, revealing a novel anti-inflammatory therapeutic opportunity for targeting PCAF/GCN5.
Collapse
Affiliation(s)
- Zuni I. Bassi
- Protein Degradation DPU, Future Pipelines Discovery, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - Martin C. Fillmore
- NCE-MD Medicinal Chemistry UK Team, R&D Platform Technology & Science, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - Afjal H. Miah
- Protein Degradation DPU, Future Pipelines Discovery, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - Trevor D. Chapman
- Epigenetics DPU, Immuno-Inflammation and Oncology Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - Claire Maller
- Epigenetics DPU, Immuno-Inflammation and Oncology Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - Emma J. Roberts
- Epigenetics DPU, Immuno-Inflammation and Oncology Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - Lauren C. Davis
- Epigenetics DPU, Immuno-Inflammation and Oncology Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - Darcy E. Lewis
- Epigenetics DPU, Immuno-Inflammation and Oncology Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - Nicholas W. Galwey
- Target Sciences Statistics, R&D Target Sciences, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | | | | | - Abigail L. Macmillan-Jones
- Protein Degradation DPU, Future Pipelines Discovery, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - Celine Gongora
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Montpellier F-34298, France
| | - Philip G. Humphreys
- Epigenetics DPU, Immuno-Inflammation and Oncology Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | | | - Rab K. Prinjha
- Epigenetics DPU, Immuno-Inflammation and Oncology Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| | - David F. Tough
- Epigenetics DPU, Immuno-Inflammation and Oncology Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage SG1 2NY, United Kingdom
| |
Collapse
|
34
|
Chen CI, Paul H, Snitzler S, Kakar S, Le LW, Wei EN, Lau A, Johnston JB, Gibson SB, Queau M, Spaner D, Croucher D, Sherry B, Trudel S. A phase 2 study of lenalidomide and dexamethasone in previously untreated patients with chronic lymphocytic leukemia (CLL). Leuk Lymphoma 2018; 60:980-989. [DOI: 10.1080/10428194.2018.1508669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
| | | | - Susi Snitzler
- Princess Margaret Cancer Centre, Toronto, (ON) Canada
| | - Sumeet Kakar
- Princess Margaret Cancer Centre, Toronto, (ON) Canada
| | - Lisa W. Le
- Princess Margaret Cancer Centre, Toronto, (ON) Canada
| | - Ellen N. Wei
- Princess Margaret Cancer Centre, Toronto, (ON) Canada
| | - Anthea Lau
- Princess Margaret Cancer Centre, Toronto, (ON) Canada
| | | | | | - Michelle Queau
- Manitoba Institute of Cell Biology, Winnipeg, (MN), Canada
| | - David Spaner
- Sunnybrook Health Sciences Centre, Toronto, (ON), Canada
| | | | - Barbara Sherry
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, (NY) USA
| | | |
Collapse
|
35
|
Ito T, Konishi A, Tsubokura Y, Azuma Y, Hotta M, Yoshimura H, Nakanishi T, Fujita S, Nakaya A, Satake A, Ishii K, Nomura S. Combined Use of Ninjin'yoeito Improves Subjective Fatigue Caused by Lenalidomide in Patients With Multiple Myeloma: A Retrospective Study. Front Nutr 2018; 5:72. [PMID: 30186837 PMCID: PMC6110850 DOI: 10.3389/fnut.2018.00072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 07/30/2018] [Indexed: 11/25/2022] Open
Abstract
Lenalidomide is an immunomodulating derivative of thalidomide, which shows anti-tumor activity against myeloma cells with immunomodulation including augmentation of T-cell and natural killer cell function. Continuous treatment with this agent shows better survival benefit in patients with multiple myeloma and combined lenalidomide with dexamethasone (LEN-DEX) is a standard treatment regimen. However, fatigue is a frequent symptom resulting from lenalidomide administration. This side-effect therefore reduces quality of life for elderly patients and, furthermore, is a reason for treatment discontinuation. Unfortunately, appropriate preventive countermeasures against lenalidomide-related fatigue have not been established. Ninjin'yoeito is a traditional Chinese medicine made from the extracts of 12 herbal plants, which positively affects immunity and inflammation. It is used to treat fatigue, decreased appetite, anemia, and general malaise associated with malignant tumors and chemotherapy. We have previously reported that ninjin'yoeito significantly improved patients' subjective fatigue symptoms treated with melphalan-prednisone for multiple myeloma. In the present study, we assessed the benefits of ninjin'yoeito as a supplementary treatment for patients with myeloma, and its effect on lenalidomide treatment regime compliance. We retrospectively analyzed 36 cases of newly diagnosed or relapsed/refractory multiple myeloma. The study included patients receiving LEN-DEX with onset of general fatigue after lenalidomide administration (13 and 23 patients with or without ninjin'yoeito, respectively). Frequency of subjective fatigue was significantly decreased in patients administered ninjin'yoeito, compared to those treated with LEN-DEX alone (92.3 and 47.8 % of patients with and without ninjin'yoeito, respectively; p = 0.008). In addition, combined use of ninjin'yoeito and LEN-DEX showed a trend toward reduced rates of treatment discontinuation (7.7 % and 34.8 % of patients with and without ninjin'yoeito, respectively; p = 0.076). Our results suggest that ninjin'yoeito is an effective method for treating subjective fatigue caused by lenalidomide and may have the potential to extend lenalidomide treatment duration.
Collapse
Affiliation(s)
- Tomoki Ito
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Akiko Konishi
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Yukie Tsubokura
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Yoshiko Azuma
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Masaaki Hotta
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Hideaki Yoshimura
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Takahisa Nakanishi
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Shinya Fujita
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Aya Nakaya
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Atsushi Satake
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Kazuyoshi Ishii
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Shosaku Nomura
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
36
|
Kubasch AS, Platzbecker U. Beyond the Edge of Hypomethylating Agents: Novel Combination Strategies for Older Adults with Advanced MDS and AML. Cancers (Basel) 2018; 10:E158. [PMID: 29795051 PMCID: PMC6025349 DOI: 10.3390/cancers10060158] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/19/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022] Open
Abstract
Higher-risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) of the elderly exhibit several commonalities, including first line treatment with hypomethylating agents (HMA) like azacitidine (AZA) or decitabine (DAC). Until today, response to treatment occurs in less than 50 percent of patients, and is often short-lived. Moreover, patients failing HMA have a dismal prognosis. Current developments include combinations of HMA with novel drugs targeting epigenetic or immunomodulatory pathways. Other efforts focus on the prevention of resistance to HMA using checkpoint inhibitors to enhance immune attack. This review focuses on recent advances in the field of HMA-based front-line therapies in elderly patients with myeloid diseases.
Collapse
Affiliation(s)
- Anne Sophie Kubasch
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
| | - Uwe Platzbecker
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), 01307 Dresden, Germany.
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
37
|
Ji Z, Su J, Wu D, Peng H, Zhao W, Nlong Zhao B, Zhou X. Predicting the impact of combined therapies on myeloma cell growth using a hybrid multi-scale agent-based model. Oncotarget 2018; 8:7647-7665. [PMID: 28032590 PMCID: PMC5352350 DOI: 10.18632/oncotarget.13831] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 11/30/2016] [Indexed: 11/25/2022] Open
Abstract
Multiple myeloma is a malignant still incurable plasma cell disorder. This is due to refractory disease relapse, immune impairment, and development of multi-drug resistance. The growth of malignant plasma cells is dependent on the bone marrow (BM) microenvironment and evasion of the host's anti-tumor immune response. Hence, we hypothesized that targeting tumor-stromal cell interaction and endogenous immune system in BM will potentially improve the response of multiple myeloma (MM). Therefore, we proposed a computational simulation of the myeloma development in the complicated microenvironment which includes immune cell components and bone marrow stromal cells and predicted the effects of combined treatment with multi-drugs on myeloma cell growth. We constructed a hybrid multi-scale agent-based model (HABM) that combines an ODE system and Agent-based model (ABM). The ODEs was used for modeling the dynamic changes of intracellular signal transductions and ABM for modeling the cell-cell interactions between stromal cells, tumor, and immune components in the BM. This model simulated myeloma growth in the bone marrow microenvironment and revealed the important role of immune system in this process. The predicted outcomes were consistent with the experimental observations from previous studies. Moreover, we applied this model to predict the treatment effects of three key therapeutic drugs used for MM, and found that the combination of these three drugs potentially suppress the growth of myeloma cells and reactivate the immune response. In summary, the proposed model may serve as a novel computational platform for simulating the formation of MM and evaluating the treatment response of MM to multiple drugs.
Collapse
Affiliation(s)
- Zhiwei Ji
- Division of Radiologic Sciences and Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA 27157
| | - Jing Su
- Division of Radiologic Sciences and Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA 27157
| | - Dan Wu
- Division of Radiologic Sciences and Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA 27157
| | - Huiming Peng
- Division of Radiologic Sciences and Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA 27157
| | - Weiling Zhao
- Division of Radiologic Sciences and Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA 27157
| | - Brian Nlong Zhao
- Division of Radiologic Sciences and Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA 27157
| | - Xiaobo Zhou
- Division of Radiologic Sciences and Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA 27157
| |
Collapse
|
38
|
Hagner PR, Chiu H, Ortiz M, Apollonio B, Wang M, Couto S, Waldman MF, Flynt E, Ramsay AG, Trotter M, Gandhi AK, Chopra R, Thakurta A. Activity of lenalidomide in mantle cell lymphoma can be explained by NK cell-mediated cytotoxicity. Br J Haematol 2017; 179:399-409. [DOI: 10.1111/bjh.14866] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/05/2017] [Indexed: 01/13/2023]
Affiliation(s)
| | | | | | - Benedetta Apollonio
- School of Cancer Sciences; Faculty of Life Sciences & Medicine; King's College London; London UK
| | | | | | | | | | - Alan G. Ramsay
- School of Cancer Sciences; Faculty of Life Sciences & Medicine; King's College London; London UK
| | | | | | - Rajesh Chopra
- Celgene Corporation; Summit NJ USA
- Division of Cancer Therapeutics; Institute of Cancer Research; London UK
| | | |
Collapse
|
39
|
Maier SK, Hammond JM. Role of Lenalidomide in the Treatment of Multiple Myeloma and Myelodysplasic Syndrome. Ann Pharmacother 2016; 40:286-9. [PMID: 16403850 DOI: 10.1345/aph.1g170] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: To evaluate lenalidomide in the treatment of multiple myeloma and myelodysplastic syndrome (MDS). Data Sources: Clinical literature was accessed through MEDLINE (1966–August 2005), Science Citation Index (1980–August 2005), and Proceedings of the American Society of Hematology (2000–2004). Data Synthesis: New analogs of thalidomide have been synthesized that are more potent and less toxic. Lenalidomide (CC-5013) is currently in Phase III trials for the treatment of multiple myeloma and MDS. Phase II trials demonstrated lenalidomide's efficacy in patients refractory to thalidomide. The full potential of this agent has yet to be proven, but preliminary data seem promising. Conclusions: Lenalidomide is a potent immunomodulating drug that offers different mechanisms of action and therapeutic potential for the treatment of multiple myeloma, MDS, and other malignancies.
Collapse
Affiliation(s)
- Sara K Maier
- Department of Pharmacy, Duke University Medical Center, Durham, NC 27710-3089, USA.
| | | |
Collapse
|
40
|
Xie L, Jozwik B, Weeks P, Buja LM, Brown R, Nathan S, Rajagopal K, Paruthi C, Kar B, Loyalka P, Rajapreyar I. Treatment of Multiple Myeloma in a Heart Transplant Recipient. Prog Transplant 2016; 27:65-68. [PMID: 27885144 DOI: 10.1177/1526924816679842] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Malignancy following solid organ transplant remains a significant threat to the survival of cardiac transplant recipients. Plasma cell dyscrasias including multiple myeloma have been encountered in this population, and medication treatments traditionally used to treat these disorders demonstrate immunomodulatory effects that may have implications on the transplanted allograft. Lenalidomide is an immunomodulatory agent that has been used to treat plasma cell disorders, including light-chain amyloidosis (AL) and multiple myeloma, and represents such a class of medications in which the risks and benefits in the solid organ transplant population remain to be fully elucidated. This report highlights a clinical practice issue where the treatment of a patient's multiple myeloma with lenalidomide may have potentiated an episode of severe acute cellular rejection and further demonstrates the need for future investigation of the optimal treatment of plasma cell disorders including AL amyloidosis and multiple myeloma following solid organ transplantation.
Collapse
Affiliation(s)
- Lola Xie
- 1 Division of Cardiovascular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Bartosz Jozwik
- 1 Division of Cardiovascular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Phillip Weeks
- 2 Department of Pharmacy, Memorial Hermann-Texas Medical Center, Houston, TX, USA
| | - L Maximilian Buja
- 3 Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Robert Brown
- 3 Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sriram Nathan
- 4 Center for Advanced Heart Failure, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Keshava Rajagopal
- 4 Center for Advanced Heart Failure, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christina Paruthi
- 1 Division of Cardiovascular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Biswajit Kar
- 4 Center for Advanced Heart Failure, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Pranav Loyalka
- 4 Center for Advanced Heart Failure, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Indranee Rajapreyar
- 4 Center for Advanced Heart Failure, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
41
|
Zhang L, Bi E, Hong S, Qian J, Zheng C, Wang M, Yi Q. CD4⁺ T cells play a crucial role for lenalidomide in vivo anti-tumor activity in murine multiple myeloma. Oncotarget 2016; 6:36032-40. [PMID: 26447613 PMCID: PMC4742159 DOI: 10.18632/oncotarget.5506] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/21/2015] [Indexed: 11/25/2022] Open
Abstract
Lenalidomide modulates the host immune response against myeloma via multiple actions. Although these effects have been elucidated in vitro, the central action of lenalidomide-mediated anti-myeloma immune response in vivo is not clear. To investigate its immune action in vivo, we selected the murine myeloma cell line 5TGM1, which is resistant to direct tumoricidal effects of lenalidomide in vitro and in immunodeficient mice, but sensitive to lenalidomide treatment in 5TGM1-bearing immunocompetent mice. Depletion of CD4+ T cells, but not NK cells, B cells, or CD8+ T cells, deprived lenalidomide of its therapeutic effects on 5TGM1-bearing immunocompetent mice. Lenalidomide significantly increased the numbers of IFN-γ-secreting CD4+ and CD8+ T cells but had no effects on NK cells and B cells in this mouse model. Lenalidomide slightly decreased the number of CD25+Foxp3+ T cells but increased perforin expression in CD8+ T cells in vivo. Using this mouse model for investigation of anti-tumor immune action of lenalidomide, we demonstrated that lenalidomide facilitated a type-1 anti-tumor immune response in vivo. The CD4+ T cell subset may play a critical role in the lenalidomide-mediated anti-myeloma immune response in vivo.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Enguang Bi
- Deparment of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Sungyoul Hong
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jianfei Qian
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Deparment of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Chengyun Zheng
- Department of Hematology, Second Hospital of Shandong University, Jinan, PR China
| | - Michael Wang
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Qing Yi
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Deparment of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
42
|
Torigoe K, Nakayama N, Achiwa H. [Pomalidomide (Pomalyst(®) capsule 1 mg/2 mg/3 mg/4 mg): pharmacokinetics, pharmacodynamics and clinical study outcome]. Nihon Yakurigaku Zasshi 2016; 148:154-161. [PMID: 27581964 DOI: 10.1254/fpj.148.154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
|
43
|
Gupta NK, Nolan A, Omuro A, Reid EG, Wang CC, Mannis G, Jaglal M, Chavez JC, Rubinstein PG, Griffin A, Abrams DI, Hwang J, Kaplan LD, Luce JA, Volberding P, Treseler PA, Rubenstein JL. Long-term survival in AIDS-related primary central nervous system lymphoma. Neuro Oncol 2016; 19:99-108. [PMID: 27576871 DOI: 10.1093/neuonc/now155] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The optimal therapeutic approach for patients with AIDS-related primary central nervous system lymphoma (AR-PCNSL) remains undefined. While its incidence declined substantially with combination antiretroviral therapy (cART), AR-PCNSL remains a highly aggressive neoplasm for which whole brain radiotherapy (WBRT) is considered a standard first-line intervention. METHODS To identify therapy-related factors associated with favorable survival, we first retrospectively analyzed outcomes of AR-PCNSL patients treated at San Francisco General Hospital, a public hospital with a long history of dedicated care for patients with HIV and AIDS-related malignancies. Results were validated in a retrospective, multicenter analysis that evaluated all newly diagnosed patients with AR-PCNSL treated with cART plus high-dose methotrexate (HD-MTX). RESULTS We provide evidence that CD4+ reconstitution with cART administered during HD-MTX correlates with long-term survival among patients with CD4 <100. This was confirmed in a multicenter analysis which demonstrated that integration of cART regimens with HD-MTX was generally well tolerated and resulted in longer progression-free survival than other treatments. No profound differences in immunophenotype were identified in an analysis of AR-PCNSL tumors that arose in the pre- versus post-cART eras. However, we detected evidence for a demographic shift, as the proportion of minority patients with AR-PCNSL increased since advent of cART. CONCLUSION Long-term disease-free survival can be achieved in AR-PCNSL, even among those with histories of opportunistic infections, limited access to health care, and medical non-adherence. Given this, as well as the long-term toxicities of WBRT, we recommend that integration of cART plus first-line HD-MTX be considered for all patients with AR-PCNSL.
Collapse
Affiliation(s)
- Neel K Gupta
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Amber Nolan
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Antonio Omuro
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Erin G Reid
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Chia-Ching Wang
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Gabriel Mannis
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Michael Jaglal
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Julio C Chavez
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Paul G Rubinstein
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Ann Griffin
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Donald I Abrams
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Jimmy Hwang
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Lawrence D Kaplan
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Judith A Luce
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Paul Volberding
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - Patrick A Treseler
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| | - James L Rubenstein
- Division of Hematology/Oncology, University of California, San Francisco (N.K.G., C.W., G.M., D.I.A., L.D.K., J.A.L., P.V., J.L.R.); Department of Pathology, University of California, San Francisco (A.N., P.A.T.); Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY (A.O.); Division of Hematology/Oncology, University of California, San Diego (E.G.R.); Division of Hematology/Oncology, San Francisco General Hospital (C.W., D.I.A., J.A.L.); Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL (M.J., J.C.C.); Department of Medicine, Section of Hematology/Oncology, John H. Stroger Jr. Hospital of Cook County, Ruth M. Rothstein CORE Center, Developmental Center for AIDS Research, Chicago, IL (P.G.R.); Department of Internal Medicine, Rush University Medical Center, Chicago, IL (P.G.R.); UCSF Cancer Registry, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (A.G.); Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (D.I.A., J.H., L.D.K., J.A.L., P.V., P.A.T., J.L.R.); Biostatistics and Computational Biology Core, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco (J.H.); Center for AIDS Research; UCSF Gladstone Institute of Virology and Immunology (P.V.)
| |
Collapse
|
44
|
Carroll D, Howard D, Zhu H, Paumi CM, Vore M, Bondada S, Liang Y, Wang C, St Clair DK. Simultaneous quantitation of oxidized and reduced glutathione via LC-MS/MS: An insight into the redox state of hematopoietic stem cells. Free Radic Biol Med 2016; 97:85-94. [PMID: 27212018 PMCID: PMC4996720 DOI: 10.1016/j.freeradbiomed.2016.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 05/02/2016] [Accepted: 05/06/2016] [Indexed: 01/01/2023]
Abstract
Cellular redox balance plays a significant role in the regulation of hematopoietic stem-progenitor cell (HSC/MPP) self-renewal and differentiation. Unregulated changes in cellular redox homeostasis are associated with the onset of most hematological disorders. However, accurate measurement of the redox state in stem cells is difficult because of the scarcity of HSC/MPPs. Glutathione (GSH) constitutes the most abundant pool of cellular antioxidants. Thus, GSH metabolism may play a critical role in hematological disease onset and progression. A major limitation to studying GSH metabolism in HSC/MPPs has been the inability to measure quantitatively GSH concentrations in small numbers of HSC/MPPs. Current methods used to measure GSH levels not only rely on large numbers of cells, but also rely on the chemical/structural modification or enzymatic recycling of GSH and therefore are likely to measure only total glutathione content accurately. Here, we describe the validation of a sensitive method used for the direct and simultaneous quantitation of both oxidized and reduced GSH via liquid chromatography followed by tandem mass spectrometry (LC-MS/MS) in HSC/MPPs isolated from bone marrow. The lower limit of quantitation (LLOQ) was determined to be 5.0ng/mL for GSH and 1.0ng/mL for GSSG with lower limits of detection at 0.5ng/mL for both glutathione species. Standard addition analysis utilizing mouse bone marrow shows that this method is both sensitive and accurate with reproducible analyte recovery. This method combines a simple extraction with a platform for the high-throughput analysis, allows for efficient determination of GSH/GSSG concentrations within the HSC/MPP populations in mouse, chemotherapeutic treatment conditions within cell culture, and human normal/leukemia patient samples. The data implicate the importance of the modulation of GSH/GSSG redox couple in stem cells related diseases.
Collapse
Affiliation(s)
- Dustin Carroll
- Department of Toxicology and Cancer Biology, University of Kentucky, College of Medicine, Lexington, KY, United States
| | - Diana Howard
- Hematology and Oncology Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, United States
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, College of Medicine, Lexington, KY, United States
| | - Christian M Paumi
- Department of Chemistry, Eastern Kentucky University, Richmond, KY, United States
| | - Mary Vore
- Department of Toxicology and Cancer Biology, University of Kentucky, College of Medicine, Lexington, KY, United States
| | - Subbarao Bondada
- Department of Microbiology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY, United States
| | - Ying Liang
- Department of Toxicology and Cancer Biology, University of Kentucky, College of Medicine, Lexington, KY, United States
| | - Chi Wang
- Division of Cancer Biostatistics, Markey Cancer Center, University of Kentucky, College of Medicine, Lexington, KY, United States
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, College of Medicine, Lexington, KY, United States
| |
Collapse
|
45
|
Jones JR, Pawlyn C, Davies FE, Morgan GJ. The safety of pomalidomide for the treatment of multiple myeloma. Expert Opin Drug Saf 2016; 15:535-47. [DOI: 10.1517/14740338.2016.1154039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- J. R. Jones
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - C. Pawlyn
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - F. E. Davies
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - G. J. Morgan
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
46
|
Lenalidomide potentiates CD4+CD25+Treg-related suppression of lymphoma B-cell proliferation. Clin Exp Med 2016; 17:193-207. [DOI: 10.1007/s10238-016-0411-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 02/12/2016] [Indexed: 10/22/2022]
|
47
|
Therapeutic Strategies for Neuropathic Pain: Potential Application of Pharmacosynthetics and Optogenetics. Mediators Inflamm 2016; 2016:5808215. [PMID: 26884648 PMCID: PMC4738689 DOI: 10.1155/2016/5808215] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/27/2015] [Accepted: 12/17/2015] [Indexed: 11/17/2022] Open
Abstract
Chronic pain originating from neuronal damage remains an incurable symptom debilitating patients. Proposed molecular modalities in neuropathic pain include ion channel expressions, immune reactions, and inflammatory substrate diffusions. Recent advances in RNA sequence analysis have discovered specific ion channel expressions in nociceptors such as transient receptor potential (TRP) channels, voltage-gated potassium, and sodium channels. G protein-coupled receptors (GPCRs) also play an important role in triggering surrounding immune cells. The multiple protein expressions complicate therapeutic development for neuropathic pain. Recent progress in optogenetics and pharmacogenetics may herald the development of novel therapeutics for the incurable pain. Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) facilitate the artificial manipulation of intracellular signaling through excitatory or inhibitory G protein subunits activated by biologically inert synthetic ligands. Expression of excitatory channelrhodopsins and inhibitory halorhodopsins on injured neurons or surrounding cells can attenuate neuropathic pain precisely controlled by light stimulation. To achieve the discrete treatment of injured neurons, we can exploit the transcriptome database obtained by RNA sequence analysis in specific neuropathies. This can recommend the suitable promoter information to target the injury sites circumventing intact neurons. Therefore, novel strategies benefiting from pharmacogenetics, optogenetics, and RNA sequencing might be promising for neuropathic pain treatment in future.
Collapse
|
48
|
Otáhal P, Průková D, Král V, Fabry M, Vočková P, Latečková L, Trněný M, Klener P. Lenalidomide enhances antitumor functions of chimeric antigen receptor modified T cells. Oncoimmunology 2015; 5:e1115940. [PMID: 27141398 PMCID: PMC4839314 DOI: 10.1080/2162402x.2015.1115940] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/16/2015] [Accepted: 10/29/2015] [Indexed: 12/11/2022] Open
Abstract
Tumor immunotherapy based on the use of chimeric antigen receptor modified T cells (CAR T cells) is a promising approach for the treatment of refractory hematological malignancies. However, a robust response mediated by CAR T cells is observed only in a minority of patients and the expansion and persistence of CAR T cells in vivo is mostly unpredictable.Lenalidomide (LEN) is an immunomodulatory drug currently approved for the treatment of multiple myeloma (MM) and mantle cell lymphoma, while it is clinically tested in the therapy of diffuse large B-cell lymphoma of activated B cell immunophenotype. LEN was shown to increase antitumor immune responses at least partially by modulating the activity of E3 ubiquitin ligase Cereblon, which leads to increased ubiquitinylation of Ikaros and Aiolos transcription factors, which in turn results in changed expression of various receptors on the surface of tumor cells. In order to enhance the effectiveness of CAR-based immunotherapy, we assessed the anti-lymphoma efficacy of LEN in combination with CAR19 T cells or CAR20 T cells in vitro and in vivo using various murine models of aggressive B-cell non-Hodgkin lymphomas (B-NHL).Immunodeficient NSG mice were transplanted with various human B-NHL cells followed by treatment with CAR19 or CAR20 T cells with or without LEN. Next, CAR19 T cells were subjected to series of tests in vitro to evaluate their response and signaling capacity following recognition of B cell in the presence or absence of LEN.Our data shows that LEN significantly enhances antitumor functions of CAR19 and CAR20 T cells in vivo. Additionally, it enhances production of interferon gamma by CAR19 T cells and augments cell signaling via CAR19 protein in T cells in vitro. Our data further suggests that LEN works through direct effects on T cells but not on B-NHL cells. The biochemical events underlying this costimulatory effect of LEN are currently being investigated. In summary, our data supports the use of LEN for augmentation of CAR-based immunotherapy in the clinical grounds.
Collapse
Affiliation(s)
- Pavel Otáhal
- Department of Hematology, Charles University General Hospital in Prague, Czech Republic; Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Dana Průková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague , Czech Republic
| | - Vlastimil Král
- Institute of Molecular Genetics, Czech Academy of Sciences , Prague, Czech Republic
| | - Milan Fabry
- Institute of Molecular Genetics, Czech Academy of Sciences , Prague, Czech Republic
| | - Petra Vočková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague , Czech Republic
| | - Lucie Latečková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague , Czech Republic
| | - Marek Trněný
- Department of Hematology, Charles University General Hospital in Prague , Czech Republic
| | - Pavel Klener
- Department of Hematology, Charles University General Hospital in Prague, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, Czech Republic
| |
Collapse
|
49
|
Vaccination of multiple myeloma: Current strategies and future prospects. Crit Rev Oncol Hematol 2015; 96:339-54. [PMID: 26123319 DOI: 10.1016/j.critrevonc.2015.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 05/06/2015] [Accepted: 06/09/2015] [Indexed: 01/21/2023] Open
Abstract
Tumor immunotherapy holds great promise in controlling multiple myeloma (MM) and may provide an alternative treatment modality to conventional chemotherapy for MM patients. For this reason, a major area of investigation is the development of cancer vaccines to generate myeloma-specific immunity. Several antigens that are able to induce specific T-cell responses are involved in different critical mechanisms for cell differentiation, inhibition of apoptosis, demethylation and proliferation. Strategies under development include infusion of vaccine-primed and ex vivo expanded/costimulated autologous T cells after high-dose melphalan, genetic engineering of autologous T cells with receptors for myeloma-specific epitopes, administration of dendritic cell/plasma cell fusions and administration expanded marrow-infiltrating lymphocytes. In addition, novel immunomodulatory drugs may synergize with immunotherapies. The task ahead is to evaluate these approaches in appropriate clinical settings, and to couple them with strategies to overcome mechanisms of immunoparesis as a means to induce more robust clinically significant immune responses.
Collapse
|
50
|
Kuroda J, Kobayashi T, Taniwaki M. Prognostic indicators of lenalidomide for multiple myeloma: consensus and controversy. Expert Rev Anticancer Ther 2015; 15:787-804. [PMID: 25947283 DOI: 10.1586/14737140.2015.1044249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The long-term outcome of multiple myeloma (MM) has been greatly improved through new agents, one being lenalidomide (LEN). Based upon the findings of in vitro experiments, its mode of action against MM occurs through a combination of direct tumoricidal effects on myeloma cells, modulatory effects on tumor immunity and tumor microenvironment-regulatory effects. However, it has not been clearly defined whether the clinical response and long-term outcome of MM with LEN treatment truly reflect the mechanisms of action of LEN proposed by in vitro studies. To ascertain what is known and what remains to be elucidated with LEN, we review the current literature on the mode of action of LEN in association with myeloma pathophysiology, and discuss the prognostic indicators in the treatment of MM with LEN.
Collapse
Affiliation(s)
- Junya Kuroda
- Department of Medicine, Division of Hematology and Oncology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | | | | |
Collapse
|