1
|
Yue L, Li J, Yao M, Song S, Zhang X, Wang Y. Cutting edge of immune response and immunosuppressants in allogeneic and xenogeneic islet transplantation. Front Immunol 2024; 15:1455691. [PMID: 39346923 PMCID: PMC11427288 DOI: 10.3389/fimmu.2024.1455691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
As an effective treatment for diabetes, islet transplantation has garnered significant attention and research in recent years. However, immune rejection and the toxicity of immunosuppressive drugs remain critical factors influencing the success of islet transplantation. While immunosuppressants are essential in reducing immune rejection reactions and can significantly improve the survival rate of islet transplants, improper use of these drugs can markedly increase mortality rates following transplantation. Additionally, the current availability of islet organ donations fails to meet the demand for organ transplants, making xenotransplantation a crucial method for addressing organ shortages. This review will cover the following three aspects: 1) the immune responses occurring during allogeneic islet transplantation, including three stages: inflammation and IBMIR, allogeneic immune response, and autoimmune recurrence; 2) commonly used immunosuppressants in allogeneic islet transplantation, including calcineurin inhibitors (Cyclosporine A, Tacrolimus), mycophenolate mofetil, glucocorticoids, and Bortezomib; and 3) early and late immune responses in xenogeneic islet transplantation and the immune effects of triple therapy (ECDI-fixed donor spleen cells (ECDI-SP) + anti-CD20 + Sirolimus) on xenotransplantation.
Collapse
Affiliation(s)
- Liting Yue
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jisong Li
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingjun Yao
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Xiaoqin Zhang
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
2
|
Popek-Marciniec S, Styk W, Wojcierowska-Litwin M, Szudy-Szczyrek A, Dudek P, Swiderska-Kolacz G, Czerwik-Marcinkowska J, Zmorzynski S. The Relationship of CCL5 and CCR1 Variants with Response Rate and Survival Taking into Account Thalidomide/Bortezomib Treatment in Patients with Multiple Myeloma. J Clin Med 2023; 12:jcm12062384. [PMID: 36983384 PMCID: PMC10056693 DOI: 10.3390/jcm12062384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
(1) Background: Chemokines and chemokine receptors play an important role in tumor development. The aim of this study was to check the significance of CCL5 and CCR1 variants with response rate, survival, and the level of regulated on activation, normal T cells expressed and secreted (RANTES/CCL5) in multiple myeloma (MM) patients; (2) Methods: Genomic DNA from 101 newly diagnosed MM patients and 100 healthy blood donors were analyzed by Real-time PCR method (for CCL5 and CCR1 genotyping). In a subgroup of 70 MM patients, serum samples were collected to determine the level of RANTES; (3) Results: multivariate Cox regression showed increased risk of disease relapse or progression (HR = 4.77; p = 0.01) in MM patients with CG + CC genotypes of CCL5 rs2280788. In contrast, CT + TT genotypes of CCL5 rs2107538 were associated withdecreased risk of death (HR = 0.18; p = 0.028) and disease relapse or progression (HR = 0.26; p = 0.01). In MM patients with major genotypes of rs2280789, rs2280788, and rs2107538, higher survival rates were observed in response to treatment with thalidomide and bortezomib. Statistically significant lower RANTES levels were seen in minor genotypes and heterozygotes of CCL5 and CCR1 variants; (4) Conclusions: Major genotypes of CCL5 variants may be independent positive prognostic factors in MM.
Collapse
Affiliation(s)
- Sylwia Popek-Marciniec
- Department of Cancer Genetics with Cytogenetic Laboratory, Medical University of Lublin, 20-059 Lublin, Poland
| | - Wojciech Styk
- Department of Psychology, Medical University of Lublin, 20-059 Lublin, Poland
| | | | - Aneta Szudy-Szczyrek
- Chair and Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-059 Lublin, Poland
| | - Paul Dudek
- Department of Cancer Genetics with Cytogenetic Laboratory, Medical University of Lublin, 20-059 Lublin, Poland
| | | | | | - Szymon Zmorzynski
- Department of Cancer Genetics with Cytogenetic Laboratory, Medical University of Lublin, 20-059 Lublin, Poland
- Correspondence:
| |
Collapse
|
3
|
Alwahsh M, Farhat J, Talhouni S, Hamadneh L, Hergenröder R. Bortezomib advanced mechanisms of action in multiple myeloma, solid and liquid tumors along with its novel therapeutic applications. EXCLI JOURNAL 2023; 22:146-168. [PMID: 36998701 PMCID: PMC10043448 DOI: 10.17179/excli2022-5653] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/12/2023] [Indexed: 04/01/2023]
Abstract
Bortezomib (BTZ) is a first-in-class reversible and selective proteasome inhibitor. It inhibits the ubiquitin proteasome pathway that leads to the degradation of many intracellular proteins. Initially, BTZ was FDA approved for the treatment of refractory or relapsed multiple myeloma (MM) in 2003. Later, its usage was approved for patients with previously untreated MM. In 2006, BTZ was approved for the treatment of relapsed or refractory Mantle Cell Lymphoma (MCL) and, in 2014, for previously untreated MCL. BTZ has been extensively studied either alone or in combination with other drugs for the treatment of different liquid tumors especially in MM. However, limited data evaluated the efficacy and safety of using BTZ in patients with solid tumors. In this review, we will discuss the advanced and novel mechanisms of action of BTZ documented in MM, solid tumors and liquid tumors. Moreover, we will shed the light on the newly discovered pharmacological effects of BTZ in other prevalent diseases.
Collapse
Affiliation(s)
- Mohammad Alwahsh
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany
- Institute of Pathology and Medical Research Center (ZMF), University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany
- *To whom correspondence should be addressed: Mohammad Alwahsh, Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan, E-mail:
| | - Joviana Farhat
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, PO Box 127788, United Arab Emirates
| | - Shahd Talhouni
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan
| | - Lama Hamadneh
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan
| | - Roland Hergenröder
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany
| |
Collapse
|
4
|
Claveau JS, LeBlanc R, Ahmad I, Delisle JS, Cohen S, Kiss T, Bambace NM, Bernard L, Lachance S, Roy DC, Sauvageau G, Veilleux O, Roy J. Bortezomib Maintenance After Allogeneic Transplantation in Newly Diagnosed Myeloma Patients Results in Decreased Incidence and Severity of Chronic GVHD. Transplant Cell Ther 2023; 29:44.e1-44.e9. [PMID: 36334654 DOI: 10.1016/j.jtct.2022.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/11/2022] [Accepted: 10/26/2022] [Indexed: 11/05/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) has curative potential in myeloma but remains hampered by high rates of relapse and chronic graft-versus-host disease (GVHD). We hypothesized that bortezomib (BTZ) as maintenance therapy after allo HCT could not only decrease the incidence of relapse but also the incidence and severity of chronic GVHD. The primary endpoint of this study was to determine whether BTZ maintenance decreases the incidence and severity of chronic GVHD using National Institutes of Health (NIH) criteria. The secondary endpoints were to determine the immunosuppression burden, organ involvement and survival (overall survival, progression-free survival) in patients either receiving or not receiving BTZ. In this retrospective study, we compared the outcome of 46 myeloma patients who received BTZ after upfront tandem auto-allo HCT between 2008 and 2020 to 61 patients without maintenance. We explored the impact of BTZ maintenance on incidence and severity of chronic GVHD using the 2014 NIH criteria. At 2 years, incidences of overall (61.2% versus 83.6%; P = .001), and moderate/severe chronic GVHD (44.5% versus 77.0%; P = .001) were significantly lower in BTZ recipients who had less mouth (43% versus 67%; P = .018) and eyes (9% versus 41%; P = .001) involvement at initial diagnosis. We report a lower use of systemic steroids (45.1% versus 76.4%; P < .001), mycophenolate mofetil (15.5% versus 28.2%; P = .031) and tacrolimus (34.5% versus 70.6%; P < .001) in BTZ recipients. Probability of being alive and off systemic immunosuppressants at 3 years was 77% in BTZ recipients and 56% in controls (P = .046). To date, there is no difference in survival between both groups. In summary, BTZ maintenance improved incidence and severity of chronic GVHD and should be considered as a valid option in myeloma patients receiving upfront tandem auto-allo HCT.
Collapse
Affiliation(s)
- Jean-Sébastien Claveau
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada.
| | - Richard LeBlanc
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Imran Ahmad
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Sébastien Delisle
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Sandra Cohen
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Thomas Kiss
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Nadia M Bambace
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Léa Bernard
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Silvy Lachance
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Denis Claude Roy
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Guy Sauvageau
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Olivier Veilleux
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | - Jean Roy
- Division of Hematology, Oncology and Transplantation, Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
5
|
Firer MA, Shapira MY, Luboshits G. The Impact of Induction Regimes on Immune Responses in Patients with Multiple Myeloma. Cancers (Basel) 2021; 13:4090. [PMID: 34439244 PMCID: PMC8393868 DOI: 10.3390/cancers13164090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/16/2022] Open
Abstract
Current standard frontline therapy for newly diagnosed patients with multiple myeloma (NDMM) involves induction therapy, autologous stem cell transplantation (ASCT), and maintenance therapy. Major efforts are underway to understand the biological and the clinical impacts of each stage of the treatment protocols on overall survival statistics. The most routinely used drugs in the pre-ASCT "induction" regime have different mechanisms of action and are employed either as monotherapies or in various combinations. Aside from their direct effects on cancer cell mortality, these drugs are also known to have varying effects on immune cell functionality. The question remains as to how induction therapy impacts post-ASCT immune reconstitution and anti-tumor immune responses. This review provides an update on the known immune effects of melphalan, dexamethasone, lenalidomide, and bortezomib commonly used in the induction phase of MM therapy. By analyzing the actions of each individual drug on the immune system, we suggest it might be possible to leverage their effects to rationally devise more effective induction regimes. Given the genetic heterogeneity between myeloma patients, it may also be possible to identify subgroups of patients for whom particular induction drug combinations would be more appropriate.
Collapse
Affiliation(s)
- Michael A. Firer
- Department Chemical Engineering, Ariel University, Ariel 40700, Israel;
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel
- Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel
| | - Michael Y. Shapira
- The Hematology Institute, Assuta Medical Center, Tel Aviv 6971028, Israel;
| | - Galia Luboshits
- Department Chemical Engineering, Ariel University, Ariel 40700, Israel;
- Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel
| |
Collapse
|
6
|
Rodriguez N, Lee J, Flynn L, Murray F, Devlin SM, Soto C, Cho C, Dahi P, Giralt S, Perales MA, Sauter C, Ponce DM. Oral Proteasome Inhibitor Ixazomib for Switch-Maintenance Prophylaxis of Recurrent or Late Acute and Chronic Graft-versus-Host Disease after Day 100 in Allogeneic Stem Cell Transplantation. Transplant Cell Ther 2021; 27:920.e1-920.e9. [PMID: 34029766 DOI: 10.1016/j.jtct.2021.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
Graft-versus-host disease (GVHD) is a frequent complication in the first year after allogeneic stem cell transplantation (allo-HCT). Recipients of reduced-intensity (RI) or nonmyeloablative (NMA) conditioning combined with calcineurin inhibitor (CNI)-based GVHD prophylaxis frequently develop GVHD in the context of immunosuppression taper. Ixazomib is an oral proteasome inhibitor with a wide safety profile that has demonstrated immunomodulatory properties, inhibition of pro-inflammatory cytokines, and anti-tumor activity. We hypothesized that switch-maintenance GVHD prophylaxis using ixazomib would facilitate CNI taper without increased GVHD frequency and severity while maintaining graft-versus-tumor (GVT) effect and an acceptable safety profile. We conducted an open-label, prospective, single-center pilot study in patients with hematologic malignancies who received an RI or NMA conditioning and CNI-based GVHD prophylaxis that were within day 100 to 150 after HCT (n = 18). Patients were treated with ixazomib once weekly on a 28-day cycle (3 weeks on, 1 week off). Treatment was safe; most adverse events were grade 1 or 2, with cytopenia and elevation in transaminases the most common. Five patients were removed from the study because of toxicity or side effects. Only 5 of 18 patients developed GVHD during the study, and its severity was driven by acute manifestations while chronic involvement was mild. The cumulative incidence of grade II-IV acute and chronic GVHD at 1-year after HCT was 33% (95% confidence interval [CI], 13-55). No patients died during the study, and only 1 had malignant relapse. An additional patient relapsed after completion of the study but within 1 year after HCT. The probability of progression-free survival and GVHD-free/relapse-free survival (composite endpoint) at 1 year were 89% (95% CI, 75-100) and 78% (95% CI, 61-100), respectively. Immune reconstitution analysis showed a rapid and sustained recovery in T-cell subpopulations and B cell reconstitution, and vaccine response in a subset of patients demonstrated continuing or de novo positive protective antibody titers. This study demonstrated low incidence of recurrent and late acute and chronic GVHD within 1 year after HCT possible associated with switch-maintenance GVHD prophylaxis using ixazomib. This approach allowed for CNI taper while preserving GVT effect, without aggravating GVHD. Our findings support further development of this approach and provide a proof-of-concept for switch-maintenance GVHD prophylaxis.
Collapse
Affiliation(s)
- Natasia Rodriguez
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jasme Lee
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lisa Flynn
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fiona Murray
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cristina Soto
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christina Cho
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Parastoo Dahi
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Sergio Giralt
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Craig Sauter
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Doris M Ponce
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York.
| |
Collapse
|
7
|
Jin X, Zhang W, Wang Y, Liu J, Hao F, Li Y, Tian M, Shu H, Dong J, Feng Y, Wei M. Pyruvate Kinase M2 Promotes the Activation of Dendritic Cells by Enhancing IL-12p35 Expression. Cell Rep 2021; 31:107690. [PMID: 32460017 DOI: 10.1016/j.celrep.2020.107690] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/06/2020] [Accepted: 05/01/2020] [Indexed: 12/30/2022] Open
Abstract
Dendritic cells (DCs) play a central role in both innate and adaptive immunity. Emerging evidence has demonstrated metabolic reprogramming during DC activation. However, how DC activation is linked with metabolic reprogramming remains unclear. Here we show that pyruvate kinase M2 (PKM2), the rate-limiting enzyme in the last step of glycolysis, is critical for LPS-induced DC activation. Upon DC activation, JNK signaling stimulated p300 association with PKM2 for the acetylation of lysine 433, a classic posttranslational modification critical for PKM2 destabilization and nuclear re-localization. Subsequently, nuclear PKM2 partnered with c-Rel to enhance Il12p35 expression, which is important for Th1 cell differentiation. Meanwhile, decreased enzymatic activity of PKM2 due to detetramerization facilitated glycolysis and fatty acid synthesis, helping DCs meet their need for biomacromolecules. Together, we provide evidence for metabolic control of DC activation and offer insights into aberrant immune responses due to dysregulated Th1 functions.
Collapse
Affiliation(s)
- Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Wenxia Zhang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Yang Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Jia Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Fengqi Hao
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Yunlong Li
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Miaomiao Tian
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Hengyao Shu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Jiaxin Dong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China
| | - Yunpeng Feng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China.
| | - Min Wei
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, 5268 Renmin Street, Changchun, Jilin, 130024, People's Republic of China.
| |
Collapse
|
8
|
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
|
9
|
Uhelski ML, Li Y, Fonseca MM, Romero-Snadoval EA, Dougherty PM. Role of innate immunity in chemotherapy-induced peripheral neuropathy. Neurosci Lett 2021; 755:135941. [PMID: 33961945 DOI: 10.1016/j.neulet.2021.135941] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 01/18/2023]
Abstract
It has become increasingly clear that the innate immune system plays an essential role in the generation of many types of neuropathic pain including that which accompanies cancer treatment. In this article we review current findings of the role of the innate immune system in contributing to cancer treatment pain at the distal endings of peripheral nerve, in the nerve trunk, in the dorsal root ganglion and in the spinal dorsal horn.
Collapse
Affiliation(s)
- Megan L Uhelski
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States
| | - Yan Li
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States
| | - Miriam M Fonseca
- The Department of Anesthesiology, Wake Forest School of Medicine, United States
| | | | - Patrick M Dougherty
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States.
| |
Collapse
|
10
|
Elotuzumab spares dendritic cell integrity and functionality. J Cancer Res Clin Oncol 2021; 147:2167-2170. [PMID: 33651141 PMCID: PMC8164573 DOI: 10.1007/s00432-021-03572-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/15/2021] [Indexed: 11/24/2022]
|
11
|
A clinical perspective on plasma cell leukemia; current status and future directions. Blood Cancer J 2021; 11:23. [PMID: 33563906 PMCID: PMC7873074 DOI: 10.1038/s41408-021-00414-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/15/2020] [Accepted: 01/14/2021] [Indexed: 02/08/2023] Open
Abstract
Primary plasma cell leukemia (pPCL) is an aggressive plasma cell disorder with a guarded prognosis. The diagnosis is confirmed when peripheral blood plasma cells (PCs) exceed 20% of white blood cells or 2000/μL. Emerging data demonstrates that patients with lower levels of circulating (PCs) have the same adverse prognosis, challenging the clinical disease definition, but supporting the adverse impact of circulating PCs. The cornerstone of treatment consists of combination therapy incorporating a proteasome inhibitor, an immunomodulatory agent, steroids, and/or anthracyclines and alkylators as part of more-intensive chemotherapy, followed by consolidative autologous hematopoietic cell transplantation in eligible patients and then maintenance therapy. Monoclonal antibodies are also currently being evaluated in this setting with a strong rationale for their use based on their activity in multiple myeloma (MM). Due to limited therapeutic studies specifically evaluating pPCL, patients with pPCL should be considered for clinical trials. In contrast to MM, the outcomes of patients with pPCL have only modestly improved with novel therapies, and secondary PCL arising from MM in particular is associated with a dismal outlook. Newer drug combinations, immunotherapy, and cellular therapy are under investigation, and these approaches hopefully will demonstrate efficacy to improve the prognosis of pPCL.
Collapse
|
12
|
Chhabra S, Visotcky A, Pasquini MC, Zhu F, Tang X, Zhang MJ, Thompson R, Abedin S, D'Souza A, Dhakal B, Drobyski WR, Fenske TS, Jerkins JH, Douglas Rizzo J, Runaas L, Saber W, Shah NN, Shaw BE, Horowitz MM, Hari PN, Hamadani M. Ixazomib for Chronic Graft-versus-Host Disease Prophylaxis following Allogeneic Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2020; 26:1876-1885. [PMID: 32653622 PMCID: PMC7571859 DOI: 10.1016/j.bbmt.2020.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/12/2020] [Accepted: 07/02/2020] [Indexed: 11/16/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) is major cause of morbidity and mortality following allogeneic hematopoietic cell transplantation (HCT). Ixazomib is an oral, second-generation, proteasome inhibitor that has been shown in preclinical models to prevent GVHD. We conducted a phase I/II trial in 57 patients to evaluate the safety and efficacy of ixazomib administration for cGVHD prophylaxis in patients undergoing allogeneic HCT. Oral ixazomib was administered on a weekly basis for a total of 4 doses, beginning days +60 through +90, to recipients of matched related donor (MRD, n = 25) or matched unrelated donor (MUD, n = 26) allogeneic HCT in phase II portion of the study, once the recommended phase II dose of 4 mg was identified in phase I (n = 6). All patients received peripheral blood graft and standard GVHD prophylaxis of tacrolimus and methotrexate. Ixazomib administration was safe and well tolerated, with thrombocytopenia, leukopenia, gastrointestinal complaints, and fatigue the most common adverse events (>10%). In phase II (n = 51), the cumulative incidence of cGVHD at 1 year was 36% (95% confidence interval [CI], 19% to 54%) in the MRD cohort and 39% (95% CI, 21% to 56%) in the MUD cohort. One-year cumulative incidence of nonrelapse mortality (NRM) and relapse was 0% and 20% (95% CI, 8% to 36%) in the MRD cohort, respectively. In the MUD cohort, the respective NRM and relapse rates were 4% (0% to 16%) and 34% (17% to 52%). The outcomes on the study were compared post hoc with contemporaneous matched Center for International Blood and Marrow Transplant Research (CIBMTR) controls. This post hoc analysis showed no significant improvement in cGVHD rates in both the MRD (hazard ratio [HR] = 0.85, P = .64) or MUD cohorts (HR = 0.68, P = .26) on the study compared with CIBMTR controls. B cell activating factor plasma levels were significantly higher after ixazomib dosing in those who remained cGVHD free compared with those developed cGVHD. This study shows that the novel strategy of short-course oral ixazomib following allogeneic HCT is safe but did not demonstrate significant improvement in cGVHD incidence in recipients of MRD and MUD transplantation compared with matched CIBMTR controls. This study is registered at www.clinicaltrials.gov as NCT02250300.
Collapse
Affiliation(s)
- Saurabh Chhabra
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin
| | - Alexis Visotcky
- Department of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Marcelo C Pasquini
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin
| | - Fenlu Zhu
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Xiaoying Tang
- Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin; Department of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mei-Jie Zhang
- Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin; Department of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Robert Thompson
- Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin
| | - Sameem Abedin
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anita D'Souza
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin
| | - Binod Dhakal
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin
| | - William R Drobyski
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Timothy S Fenske
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin
| | - James H Jerkins
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin
| | - J Douglas Rizzo
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin
| | - Lyndsey Runaas
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Wael Saber
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin
| | - Nirav N Shah
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Bronwen E Shaw
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin
| | - Mary M Horowitz
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin
| | - Parameswaran N Hari
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin
| | - Mehdi Hamadani
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Blood and Marrow Transplant & Cellular Therapy Program, Froedtert & Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee Campus, Milwaukee, Wisconsin.
| |
Collapse
|
13
|
Chiu HF, Wen MC, Wu MJ, Chen CH, Yu TM, Chuang YW, Huang ST, Tsai SF, Lo YC, Ho HC, Shu KH. Treatment of chronic active antibody-mediated rejection in renal transplant recipients - a single center retrospective study. BMC Nephrol 2020; 21:6. [PMID: 31906890 PMCID: PMC6945538 DOI: 10.1186/s12882-019-1672-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/25/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Chronic active antibody-mediated rejection is a major etiology of graft loss in renal transplant recipients. However, there is no consensus on the optimal treatment strategies. METHODS Computerized records from Taichung Veterans General Hospital were collected to identify renal transplant biopsies performed in the past 7 years with a diagnosis of chronic active antibody-mediated rejection. The patients were divided into two groups according to treatment strategy: Group 1 received aggressive treatment (double filtration plasmapheresis and one of the followings: rituximab, intravenous immunoglobulin, antithymogycte globulin, bortezomib, or methylprednisolone pulse therapy); and group 2 received supportive treatment. RESULTS From February 2009 to December 2017, a total of 82 patients with biopsy-proven chronic antibody mediated rejection were identified. Kaplan-Meier analysis of death-censored graft survival showed a worse survival in group 2 (P = 0.015 by log-rank test). Adverse event-free survival was lower in group 1, whereas patient survival was not significantly different. Proteinuria and supportive treatment were independent risk factors for graft loss in multivariate analysis. CONCLUSIONS Aggressive treatment was associated with better graft outcome. However, higher incidence of adverse events merit personalized treatment, especially for those with higher risk of infection. Appropriate prophylactic antibiotics are recommended for patients undergoing aggressive treatment.
Collapse
Affiliation(s)
- Hsien-Fu Chiu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Mei-Chin Wen
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Cheng-Hsu Chen
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tung-Min Yu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ya-Wen Chuang
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shih-Ting Huang
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shang-Feng Tsai
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ying-Chih Lo
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hao-Chung Ho
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Kuo-Hsiung Shu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan. .,Division of Nephrology, Department of Internal Medicine, Lin Shin Hospital, No.36, Sec. 3, Hueijhong Rd., Nantun District, Taichung City, 40867, Taiwan.
| |
Collapse
|
14
|
Hu JJ, Nie SM, Gao Y, Yan XS, Huang JX, Li TL, Liu SS, Mao CX, Zhou JJ, Xu YJ, Wang W, Meng FJ, Feng XQ. [The correlations and prognostic value of neutrophil to lymphocyte ratio, immunophenotype and cytogenetic abnormalities in patients with newly diagnosed multiple myeloma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:1044-1046. [PMID: 32023739 PMCID: PMC7342691 DOI: 10.3760/cma.j.issn.0253-2727.2019.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Indexed: 11/17/2022]
Affiliation(s)
- J J Hu
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - S M Nie
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Y Gao
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - X S Yan
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - J X Huang
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - T L Li
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - S S Liu
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - C X Mao
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - J J Zhou
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Y J Xu
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - W Wang
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - F J Meng
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - X Q Feng
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
15
|
A combination regimen of low-dose bortezomib and rapamycin prolonged the graft survival in a murine allogeneic islet transplantation model. Immunol Lett 2019; 216:21-27. [PMID: 31593743 DOI: 10.1016/j.imlet.2019.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 11/21/2022]
Abstract
As the first FDA-approved proteasome inhibitor drug, bortezomib has been used for the treatment of multiple myeloma and lymphoma. However, its effects alone or in combination with other immunosuppressants on allogeneic islet transplantation have not been reported so far. In this study, we showed that the short-term combination treatment of low-dose bortezomib and rapamycin significantly prolonged the survival of islet allografts. Short-term treatment of low-dose (0.05 mg/kg or 0.1 mg/kg) bortezomib reduced the MHC class II expression in dendritic cells (DCs) of alloantigen-sensitized mice, and prolonged the islet allograft survival for up to 50 days in diabetic mice. Notably, when bortezomib was combined with rapamycin, it induced islet-specific immunological tolerance which allowed the acceptance of a second graft without additional immunosuppression. This regimen dramatically reduced the alloantigen-specific IFN-γ-producing T cells in the spleen, and increased regulatory T cells both at the graft site and in the spleen. Therefore, we propose that short-term treatment of low-dose bortezomib and rapamycin could be a new tolerance-promoting immunosuppressive regimen for allogeneic islet transplantation.
Collapse
|
16
|
Phase I study of graft-versus-host disease prophylaxis including bortezomib for allogeneic hematopoietic cell transplantation from unrelated donors with one or two HLA loci mismatches in Japanese patients. Int J Hematol 2019; 110:736-742. [PMID: 31560116 DOI: 10.1007/s12185-019-02743-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 10/25/2022]
Abstract
This phase I study was designed for graft-versus-host disease (GVHD) prophylaxis including bortezomib in allogeneic hematopoietic cell transplantation (allo-HCT) from human leukocyte antigen (HLA)-mismatched unrelated donors in Japanese patients. Patients were administered bortezomib on days 1, 4, and 7, with short-term methotrexate and tacrolimus. Three bortezomib dose levels were prepared (1.0, 1.3, and 1.5 mg/m2). A dose of 1.3 mg/m2 was planned for administration to the initial six patients, and was adjusted if dose-limiting toxicity developed. Five of six patients enrolled for the initial dose had bone marrow donors. Two cases had single-antigen and single-allele mismatches; four had single-antigen mismatch at the A, B, C, and/or DRB1 loci in the GVH direction. All patients achieved neutrophil engraftment and complete donor chimerism. Three patients developed grade II acute GVHD, and none developed grade III-IV GVHD or any dose-limiting toxicity attributable to bortezomib by day 100. Two patients developed late-onset acute GVHD, and two developed chronic GVHD, but all cases were manageable. All patients were alive without relapse after a median follow-up period of 52 months. The optimal dose of bortezomib was determined to be 1.3 mg/m2. Prophylaxis against GVHD using a regimen including bortezomib thus seems feasible for HLA-mismatched unrelated allo-HCT.
Collapse
|
17
|
Caballero-Velázquez T, Calderón-Cabrera C, López-Corral L, Puig N, Marquez-Malaver F, Pérez-López E, García-Calderón C, Rosso-Fernández CM, Caballero Barrigón D, Martín J, Mateos MV, San Miguel J, Pérez-Simón JA. Efficacy of bortezomib to intensify the conditioning regimen and the graft-versus-host disease prophylaxis for high-risk myeloma patients undergoing transplantation. Bone Marrow Transplant 2019; 55:419-430. [PMID: 31551517 DOI: 10.1038/s41409-019-0670-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/15/2019] [Accepted: 07/22/2019] [Indexed: 11/09/2022]
Abstract
This multicenter phase I trial was designed to evaluate the safety and efficacy of bortezomib (Bz) as part of both the conditioning regimen and the graft-versus-host disease (GvHD) prophylaxis. Patients received fludarabine, melphalan and Bz (days -9 and -2). GVHD prophylaxis consisted of Bz (days +1, +4, and +7), sirolimus (Siro) from day -5 and tacrolimus (Tk) from -3 (except the first five patients that did not receive Tk). Twenty-five patients with poor prognostic multiple myeloma were included. Eleven out of the 19 patients had high-risk features. Out of the 21 patients evaluable at day +100, 14 were in CR (67%) and 7 (33%) in PR. Cumulative incidence (CI) of nonrelapse mortality at 1 year was 24%. CI of grades 2-4 and 3-4 acute GvHD was 35% and 10%, respectively; CI of chronic GvHD was 35% and 55% at 1 and 2 years, respectively. Overall and event free survival at 2 years were 64% and 31%, respectively. Bz as part of the conditioning regimen and in the combination with Siro/tacrolimus for GvHD prophylaxis is safe and effective allowing an optimal disease control early after transplant and reducing the risk of GvHD.
Collapse
Affiliation(s)
- T Caballero-Velázquez
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, Seville, Spain
| | - C Calderón-Cabrera
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, Seville, Spain
| | - L López-Corral
- Department of Hematology, Complejo Asistencial Universitario de Salamanca-IBSAL-CIBERONC, Centro de Investigación del Cáncer-IBMCC, Salamanca, Spain
| | - N Puig
- Department of Hematology, Complejo Asistencial Universitario de Salamanca-IBSAL-CIBERONC, Centro de Investigación del Cáncer-IBMCC, Salamanca, Spain
| | - F Marquez-Malaver
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, Seville, Spain
| | - E Pérez-López
- Department of Hematology, Complejo Asistencial Universitario de Salamanca-IBSAL-CIBERONC, Centro de Investigación del Cáncer-IBMCC, Salamanca, Spain
| | - C García-Calderón
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, Seville, Spain
| | - C M Rosso-Fernández
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, Seville, Spain
| | - D Caballero Barrigón
- Department of Hematology, Complejo Asistencial Universitario de Salamanca-IBSAL-CIBERONC, Centro de Investigación del Cáncer-IBMCC, Salamanca, Spain
| | - J Martín
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, Seville, Spain
| | - M V Mateos
- Department of Hematology, Complejo Asistencial Universitario de Salamanca-IBSAL-CIBERONC, Centro de Investigación del Cáncer-IBMCC, Salamanca, Spain
| | - J San Miguel
- Clínica Universidad de Navarra, Centro Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - J A Pérez-Simón
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, Seville, Spain.
| | | |
Collapse
|
18
|
Schmidt M, Altdörfer V, Schnitte S, Fuchs AR, Kropp KN, Maurer S, Müller MR, Salih HR, Rittig SM, Grünebach F, Dörfel D. The Deubiquitinase Inhibitor b-AP15 and Its Effect on Phenotype and Function of Monocyte-Derived Dendritic Cells. Neoplasia 2019; 21:653-664. [PMID: 31132676 PMCID: PMC6538843 DOI: 10.1016/j.neo.2019.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
The ubiquitin-proteasome system is elementary for cellular protein degradation and gained rising attention as a new target for cancer therapy due to promising clinical trials with bortezomib, the first-in class proteasome inhibitor meanwhile approved for multiple myeloma and mantle cell lymphoma. Both bortezomib and next-generation proteasome inhibitors mediate their effects by targeting the 20S core particle of the 26S proteasome. The novel small molecule inhibitor b-AP15 affects upstream elements of the ubiquitin-proteasome cascade by suppressing the deubiquitinase activity of both proteasomal regulatory 19S subunits and showed promising anticancer activity in preclinical models. Nonetheless, effects of inhibitors on the ubiquitin-proteasome system are not exclusively restricted to malignant cells: alteration of natural killer cell-mediated immune responses had already been described for drugs targeting either 19S or 20S proteasomal subunits. Moreover, it has been shown that bortezomib impairs dendritic cell (DC) phenotype and function at different levels. In the present study, we comparatively analyzed effects of bortezomib and b-AP15 on monocyte-derived DCs. In line with previous results, bortezomib exposure impaired maturation, antigen uptake, migration, cytokine secretion and immunostimulation, whereas treatment with b-AP15 had no compromising effects on these DC features. Our findings warrant the further investigation of b-AP15 as an alternative to clinically approved proteasome inhibitors in the therapy of malignancies, especially in the context of combinatorial treatment with DC-based immunotherapies.
Collapse
Affiliation(s)
- Moritz Schmidt
- CCU Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany
| | - Vanessa Altdörfer
- Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, UKT, Germany
| | - Sarah Schnitte
- Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, UKT, Germany
| | - Alexander Rolf Fuchs
- CCU Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany
| | - Korbinian Nepomuk Kropp
- CCU Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany
| | - Stefanie Maurer
- CCU Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany
| | - Martin Rudolf Müller
- Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, UKT, Germany
| | - Helmut Rainer Salih
- CCU Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany; Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, UKT, Germany
| | - Susanne Malaika Rittig
- Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, UKT, Germany; Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Germany
| | - Frank Grünebach
- Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, UKT, Germany
| | - Daniela Dörfel
- CCU Translational Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner site Tübingen, Germany; Department of Medical Oncology, Hematology, Immunology, Rheumatology and Pulmonology, UKT, Germany.
| |
Collapse
|
19
|
Infectious complications and NK cell depletion following daratumumab treatment of Multiple Myeloma. PLoS One 2019; 14:e0211927. [PMID: 30759167 PMCID: PMC6374018 DOI: 10.1371/journal.pone.0211927] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/17/2019] [Indexed: 12/16/2022] Open
Abstract
Treatment with Daratumumab (Dara), a monoclonal anti-CD38 antibody of IgG1 subtype, is effective in patients with multiple myeloma (MM). However, Dara also impairs the cellular immunity, which in turn may lead to higher susceptibility to infections. The exact link between immune impairment and infectious complications is unclear. In this study, we report that nine out of 23 patients (39%) with progressive MM had infectious complications after Dara treatment. Five of these patients had viral infections, two developed with bacterial infections and two with both bacterial and viral infections. Two of the viral infections were exogenous, i.e. acute respiratory syncytial virus (RSV) and human metapneumovirus (hMPV), while five consisted of reactivations, i.e. one herpes simplex (HSV), 1 varicella-zoster (VZV) and three cytomegalovirus (CMV). Infections were solely seen in patients with partial response or worse. Assessment of circulating lymphocytes indicated a selective depletion of NK cells and viral reactivation after Dara treatment, however this finding does not exclude the multiple components of viral immune-surveillance that may get disabled during this monoclonal treatment in this patient cohort. These results suggest that the use of antiviral and antibacterial prophylaxis and screening of the patients should be considered.
Collapse
|
20
|
Beling A, Kespohl M. Proteasomal Protein Degradation: Adaptation of Cellular Proteolysis With Impact on Virus-and Cytokine-Mediated Damage of Heart Tissue During Myocarditis. Front Immunol 2018; 9:2620. [PMID: 30546359 PMCID: PMC6279938 DOI: 10.3389/fimmu.2018.02620] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/24/2018] [Indexed: 12/26/2022] Open
Abstract
Viral myocarditis is an inflammation of the heart muscle triggered by direct virus-induced cytolysis and immune response mechanisms with most severe consequences during early childhood. Acute and long-term manifestation of damaged heart tissue and disturbances of cardiac performance involve virus-triggered adverse activation of the immune response and both immunopathology, as well as, autoimmunity account for such immune-destructive processes. It is a matter of ongoing debate to what extent subclinical virus infection contributes to the debilitating sequela of the acute disease. In this review, we conceptualize the many functions of the proteasome in viral myocarditis and discuss the adaptation of this multi-catalytic protease complex together with its implications on the course of disease. Inhibition of proteasome function is already highly relevant as a strategy in treating various malignancies. However, cardiotoxicity and immune-related adverse effects have proven significant hurdles, representative of the target's wide-ranging functions. Thus, we further discuss the molecular details of proteasome-mediated activity of the immune response for virus-mediated inflammatory heart disease. We summarize how the spatiotemporal flexibility of the proteasome might be tackled for therapeutic purposes aiming to mitigate virus-mediated adverse activation of the immune response in the heart.
Collapse
Affiliation(s)
- Antje Beling
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Site Berlin, Berlin, Germany
| | - Meike Kespohl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
21
|
Perrin-Cocon L, Aublin-Gex A, Diaz O, Ramière C, Peri F, André P, Lotteau V. Toll-like Receptor 4-Induced Glycolytic Burst in Human Monocyte-Derived Dendritic Cells Results from p38-Dependent Stabilization of HIF-1α and Increased Hexokinase II Expression. THE JOURNAL OF IMMUNOLOGY 2018; 201:1510-1521. [PMID: 30037846 DOI: 10.4049/jimmunol.1701522] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 07/03/2018] [Indexed: 12/22/2022]
Abstract
Cell metabolism now appears as an essential regulator of immune cells activation. In particular, TLR stimulation triggers metabolic reprogramming of dendritic cells (DCs) with an increased glycolytic flux, whereas inhibition of glycolysis alters their functional activation. The molecular mechanisms involved in the control of glycolysis upon TLR stimulation are poorly understood for human DCs. TLR4 activation of human monocyte-derived DCs (MoDCs) stimulated glycolysis with an increased glucose consumption and lactate production. Global hexokinase (HK) activity, controlling the initial rate-limiting step of glycolysis, was also increased. TLR4-induced glycolytic burst correlated with a differential modulation of HK isoenzymes. LPS strongly enhanced the expression of HK2, whereas HK3 was reduced, HK1 remained unchanged, and HK4 was not expressed. Expression of the other rate-limiting glycolytic enzymes was not significantly increased. Exploring the signaling pathways involved in LPS-induced glycolysis with various specific inhibitors, we observed that only the inhibitors of p38-MAPK (SB203580) and of HIF-1α DNA binding (echinomycin) reduced both the glycolytic activity and production of cytokines triggered by TLR4 stimulation. In addition, LPS-induced HK2 expression required p38-MAPK-dependent HIF-1α accumulation and transcriptional activity. TLR1/2 and TLR2/6 stimulation increased glucose consumption by MoDCs through alternate mechanisms that are independent of p38-MAPK activation. TBK1 contributed to glycolysis regulation when DCs were stimulated via TLR2/6. Therefore, our results indicate that TLR4-dependent upregulation of glycolysis in human MoDCs involves a p38-MAPK-dependent HIF-1α accumulation, leading to an increased HK activity supported by enhanced HK2 expression.
Collapse
Affiliation(s)
- Laure Perrin-Cocon
- Centre International de Recherche en Infectiologie, Biologie Cellulaire des Infections Virales, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Hospices Civils de Lyon, Université de Lyon, Lyon, France; and
| | - Anne Aublin-Gex
- Centre International de Recherche en Infectiologie, Biologie Cellulaire des Infections Virales, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Hospices Civils de Lyon, Université de Lyon, Lyon, France; and
| | - Olivier Diaz
- Centre International de Recherche en Infectiologie, Biologie Cellulaire des Infections Virales, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Hospices Civils de Lyon, Université de Lyon, Lyon, France; and
| | - Christophe Ramière
- Centre International de Recherche en Infectiologie, Biologie Cellulaire des Infections Virales, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Hospices Civils de Lyon, Université de Lyon, Lyon, France; and
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy
| | - Patrice André
- Centre International de Recherche en Infectiologie, Biologie Cellulaire des Infections Virales, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Hospices Civils de Lyon, Université de Lyon, Lyon, France; and
| | - Vincent Lotteau
- Centre International de Recherche en Infectiologie, Biologie Cellulaire des Infections Virales, INSERM, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Hospices Civils de Lyon, Université de Lyon, Lyon, France; and
| |
Collapse
|
22
|
Mori Y, Yoshimoto G, Yuda JI, Hayashi M, Odawara J, Kuriyama T, Sugio T, Miyawaki K, Kamezaki K, Kato K, Takenaka K, Iwasaki H, Maeda T, Miyamoto T, Akashi K. Previous exposure to bortezomib is linked to a lower risk of engraftment syndrome after autologous hematopoietic stem cell transplantation. Leuk Lymphoma 2018; 60:271-273. [PMID: 29741437 DOI: 10.1080/10428194.2018.1466295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Yasuo Mori
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Goichi Yoshimoto
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Jun-Ichiro Yuda
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Masayasu Hayashi
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Jun Odawara
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Takuro Kuriyama
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Takeshi Sugio
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Kohta Miyawaki
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Kenjiro Kamezaki
- b Center for Cellular and Molecular Medicine , Kyushu University Hospital , Fukuoka , Japan
| | - Koji Kato
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Katsuto Takenaka
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Hiromi Iwasaki
- b Center for Cellular and Molecular Medicine , Kyushu University Hospital , Fukuoka , Japan
| | - Takahiro Maeda
- b Center for Cellular and Molecular Medicine , Kyushu University Hospital , Fukuoka , Japan
| | - Toshihiro Miyamoto
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Koichi Akashi
- a Medicine and Biosystemic Science , Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan.,b Center for Cellular and Molecular Medicine , Kyushu University Hospital , Fukuoka , Japan
| |
Collapse
|
23
|
Tang AC, Rahavi SM, Fung SY, Lu HY, Yang H, Lim CJ, Reid GS, Turvey SE. Combination therapy with proteasome inhibitors and TLR agonists enhances tumour cell death and IL-1β production. Cell Death Dis 2018; 9:162. [PMID: 29415982 PMCID: PMC5833743 DOI: 10.1038/s41419-017-0194-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 12/14/2022]
Abstract
Proteasome inhibitors have emerged as an effective therapy for the treatment of haematological malignancies; however, their efficacy can be limited by the development of tumour resistance mechanisms. Novel combination strategies including the addition of TLR adjuvants to increase cell death and augment immune responses may help enhance their effectiveness. Although generally thought to inhibit inflammatory responses and NF-κB activation, we found that under specific conditions proteasome inhibitors can promote inflammatory responses by mediating IL-1β maturation and secretion after TLR stimulation. This was dependent on the timing of proteasome inhibition relative to TLR stimulation where reversal of treatment order could alternatively increase or inhibit IL-1β secretion (P < 0.001). TLR stimulation combined with proteasome inhibition enhanced cell death in vitro and delayed tumour development in vivo in NOD SCID mice (P < 0.01). However, unlike IL-1β secretion, cell death occurred similarly regardless of treatment order and was only partially caspase dependent, possessing characteristics of both apoptosis and necrosis as indicated by activation of caspase-1, 3, 8 and RIP3 phosphorylation. Although stimulation of various TLRs was capable of driving IL-1β production, TLR4 stimulation was the most effective at increasing cell death in THP-1 and U937 cells. TLR4 stimulation and proteasome inhibition independently activated the RIP3 necroptotic pathway and ultimately reduced the effectiveness of caspase/necroptosis inhibitors in mitigating overall levels of cell death. This strategy of combining TLR stimulation with proteasome inhibition may improve the ability of proteasome inhibitors to generate immunogenic cell death and increase anti-tumour activity.
Collapse
Affiliation(s)
- Anthony C Tang
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Seyed M Rahavi
- Experimental Medicine Program, University of British Columbia, BC Children's Hospital, Vancouver, BC, Canada
| | - Shan-Yu Fung
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Henry Y Lu
- Experimental Medicine Program, University of British Columbia, BC Children's Hospital, Vancouver, BC, Canada.,Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Hong Yang
- Department of Respiratory Medicine, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chinten J Lim
- Experimental Medicine Program, University of British Columbia, BC Children's Hospital, Vancouver, BC, Canada.,Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Gregor S Reid
- Experimental Medicine Program, University of British Columbia, BC Children's Hospital, Vancouver, BC, Canada.,Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Stuart E Turvey
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada. .,Experimental Medicine Program, University of British Columbia, BC Children's Hospital, Vancouver, BC, Canada. .,Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
24
|
Koreth J, Kim HT, Lange PB, Poryanda SJ, Reynolds CG, Rai SC, Armand P, Cutler CS, Ho VT, Glotzbecker B, Yusuf R, Nikiforow S, Chen YB, Dey B, McMasters M, Ritz J, Blazar BR, Soiffer RJ, Antin JH, Alyea EP. Bortezomib-based immunosuppression after reduced-intensity conditioning hematopoietic stem cell transplantation: randomized phase II results. Haematologica 2018; 103:522-530. [PMID: 29326124 PMCID: PMC5830392 DOI: 10.3324/haematol.2017.176859] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/10/2018] [Indexed: 11/16/2022] Open
Abstract
Aprior phase I/II trial of bortezomib/tacrolimus/methotrexate prophylaxis after human leukocyte antigen (HLA)-mismatched reduced intensity conditioning allogeneic hematopoietic stem cell transplantation documented low acute graft-versus-host disease incidence, with promising overall and progression-free survival. We performed an open-label three-arm 1:1:1 phase II randomized controlled trial comparing grade II–IV acute graft-versus-host disease between conventional tacrolimus/methotrexate (A) versus bortezomib/tacrolimus/methotrexate (B), and versus bortezomib/sirolimus/tacrolimus (C), in reduced intensity conditioning allogeneic transplantation recipients lacking HLA-matched related donors. The primary endpoint was grade II–IV acute graft-versus-host disease incidence rate by day +180. One hundred and thirty-eight patients (A 46, B 45, C 47) with a median age of 64 years (range: 24–75), varying malignant diagnoses and disease risk (low 14, intermediate 96, high/very high 28) received 7–8/8 HLA-mismatched (40) or matched unrelated donor (98) grafts. Median follow up in survivors was 30 months (range: 14–46). Despite early immune reconstitution differences, day +180 grade II-IV acute graft-versus-host disease rates were similar (A 32.6%, B 31.1%, C 21%; P=0.53 for A vs. B, P=0.16 for A vs. C). The 2-year non-relapse mortality incidence was similar (A 14%, B 16%, C 6.4%; P=0.62), as were relapse (A 32%, B 32%, C 38%; P=0.74), chronic graft-versus-host disease (A 59%, B 60% C 55%; P=0.66), progression-free survival (A 54%, B 52%, C 55%; P=0.95), and overall survival (A 61%, B 62%, C 62%; P=0.98). Overall, the bortezomib-based regimens evaluated did not improve outcomes compared with tacrolimus/methotrexate therapy. clinicaltrials.gov Identifier: 01754389
Collapse
Affiliation(s)
- John Koreth
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Haesook T Kim
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard School of Public Health, Boston, MA
| | - Paulina B Lange
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Samuel J Poryanda
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Carol G Reynolds
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Sharmila Chamling Rai
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Philippe Armand
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Corey S Cutler
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Vincent T Ho
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Brett Glotzbecker
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Rushdia Yusuf
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Sarah Nikiforow
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Yi-Bin Chen
- Massachusetts General Hospital Cancer Center, Boston, MA
| | | | | | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Bruce R Blazar
- University of Minnesota Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, Minneapolis, MN, USA
| | - Robert J Soiffer
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Joseph H Antin
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Edwin P Alyea
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| |
Collapse
|
25
|
Malek E, El-Jurdi N, Kröger N, de Lima M. Allograft for Myeloma: Examining Pieces of the Jigsaw Puzzle. Front Oncol 2017; 7:287. [PMID: 29322027 PMCID: PMC5732220 DOI: 10.3389/fonc.2017.00287] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/13/2017] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) cure remains elusive despite the availability of newer anti-myeloma agents. Patients with high-risk disease often suffer from early relapse and short survival. Allogeneic hematopoietic cell transplantation (allo-HCT) is an “immune-based” therapy that has the potential to offer long-term remission in a subgroup of patients, at the expense of high rates of transplant-related morbidity and mortality. Donor lymphocyte infusion (DLI) upon disease relapse after allo-HCT is able to generate an anti-myeloma response suggestive of a graft-versus-myeloma effect. Allo-HCT provides a robust platform for additional immune-based therapy upon relapse including DLI and, maintenance with immunomodulatory drugs and immunosuppressive therapy. There have been conflicting findings from randomized prospective trials questioning the role of allo-HCT. However, to this date, allo-HCT remains the only potential curable treatment for MM and its therapeutic role needs to be better defined especially for patients with high-risk disease. This review examines different aspects of this treatment and summarizes ongoing attempts at improving its therapeutic index.
Collapse
Affiliation(s)
- Ehsan Malek
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Najla El-Jurdi
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcos de Lima
- Stem Cell Transplant Program, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
26
|
Allogeneic Hematopoietic Cell Transplantation for Myeloma: When and in Whom Does It Work. Curr Hematol Malig Rep 2017; 12:126-135. [PMID: 28285435 DOI: 10.1007/s11899-017-0374-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The growing list of available therapies for patients with multiple myeloma has resulted in tremendously high response rates and prolonged survival. However, the cure remains elusive. A continued effort at developing strategies to utilize all available treatment modalities in the most effective manner is needed. Allogeneic hematopoietic cell transplantation (allo-HCT) is a robust platform, associated with high response rates, and provides a unique foundation on which immune therapies and novel agents can be employed to improve clinical outcomes. Patients with high-risk myeloma and those relapsing after novel agent-based therapies or early after an autologous HCT should be considered for allo-HCT, ideally in a clinical trial setting. Results from several ongoing studies are expected to provide important information that will help determine the place of allo-HCT in the myeloma treatment algorithm.
Collapse
|
27
|
ONX-0914, a selective inhibitor of immunoproteasome, ameliorates experimental autoimmune myasthenia gravis by modulating humoral response. J Neuroimmunol 2017; 311:71-78. [PMID: 28844501 DOI: 10.1016/j.jneuroim.2017.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/29/2017] [Accepted: 08/18/2017] [Indexed: 11/23/2022]
Abstract
Accumulating evidence shows that the immunoproteasome participates in the immune response, beyond its initial role in the protein degradation. Here, we tested the effects of the selective immunoproteasome inhibitor, ONX-0914, on experimental autoimmune myasthenia gravis (EAMG). We found that ONX-0914 ameliorated the severity of ongoing EAMG by reducing the autoantibody affinity, accompanied with decreased Tfh cells and antigen presenting cells. Also it reduced the percentage of Th17 cells and inhibited the secretion of IL-17. Our data indicated ONX-0914 may bring benefit for MG therapy.
Collapse
|
28
|
Karpova YD, Bozhok GA, Alabedal’karim NM, Lyupina YV, Astakhova TM, Legach EI, Sharova NP. Proteasomes and transplantology: Current state of the problem and the search for promising trends. BIOL BULL+ 2017. [DOI: 10.1134/s1062359017030049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
29
|
Al-Homsi AS, Cole K, Muilenburg M, Goodyke A, Abidi M, Duffner U, Williams S, Parker J, Abdel-Mageed A. Calcineurin and mTOR Inhibitor-Free Post-Transplantation Cyclophosphamide and Bortezomib Combination for Graft-versus-Host Disease Prevention after Peripheral Blood Allogeneic Hematopoietic Stem Cell Transplantation: A Phase I/II Study. Biol Blood Marrow Transplant 2017; 23:1651-1657. [PMID: 28549771 DOI: 10.1016/j.bbmt.2017.05.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/19/2017] [Indexed: 12/11/2022]
Abstract
Graft-versus-host disease (GVHD) hampers the utility of allogeneic hematopoietic stem cell transplantation (AHSCT). The purpose of this study was to determine the feasibility, safety, and efficacy of a novel combination of post-transplantation cyclophosphamide (PTC) and bortezomib for the prevention of GVHD. Patients undergoing peripheral blood AHSCT for hematological malignancies after reduced-intensity conditioning with grafts from HLA-matched related or unrelated donors were enrolled in a phase I/II clinical trial. Patients received a fixed dose of PTC and an increasing dose of bortezomib in 3 cohorts, from .7 to 1 and then to 1.3 mg/m2, administered 6 hours after graft infusion and 72 hours thereafter, during phase I. The study was then extended at the higher dose in phase II for a total of 28 patients. No graft failure and no unexpected grade ≥3 nonhematologic toxicities were encountered. The median times to neutrophil and platelet engraftment were 16 and 27 days, respectively. Day +100 treatment-related mortality was 3.6% (95% confidence interval [CI], .2% to 15.7%). The cumulative incidences of grades II to IV and grades III and IV acute GVHD were 35.9% (95% CI, 18.6% to 53.6%) and 11.7% (95% CI, 2.8% to 27.5%), respectively. The incidence of chronic GVHD was 27% (95% CI, 11.4% to 45.3%). Progression-free survival, overall survival, and GVHD and relapse-free survival rates were 50% (95% CI, 30.6% to 66.6%), 50.8% (95% CI, 30.1% to 68.2%), and 37.7% (95% CI, 20.1% to 55.3%), respectively. Immune reconstitution, measured by CD3, CD4, and CD8 recovery, was prompt. The combination of PTC and bortezomib for the prevention of GVHD is feasible, safe, and yields promising results. The combination warrants further examination in a multi-institutional trial.
Collapse
Affiliation(s)
- A Samer Al-Homsi
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan; Michigan State University College of Human Medicine, Grand Rapids, Michigan.
| | - Kelli Cole
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan
| | - Marlee Muilenburg
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan
| | - Austin Goodyke
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan
| | - Muneer Abidi
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan; Michigan State University College of Human Medicine, Grand Rapids, Michigan
| | - Ulrich Duffner
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan; Michigan State University College of Human Medicine, Grand Rapids, Michigan
| | - Stephanie Williams
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan; Michigan State University College of Human Medicine, Grand Rapids, Michigan
| | - Jessica Parker
- Office of Research Administration, Spectrum Health, Grand Rapids, Michigan
| | - Aly Abdel-Mageed
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan; Michigan State University College of Human Medicine, Grand Rapids, Michigan
| |
Collapse
|
30
|
Valković T, Gačić V, Ivandić J, Petrov B, Dobrila-Dintinjana R, Dadić-Hero E, Načinović-Duletić A. Infections in Hospitalised Patients with Multiple Myeloma: Main Characteristics and Risk Factors. Turk J Haematol 2017; 32:234-42. [PMID: 26376590 PMCID: PMC4563199 DOI: 10.4274/tjh.2013.0173] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Multiple myeloma is a common haematological malignancy and immune dysfunction is the hallmark of the disease. It leads to an increased infection risk, which is still a major cause of mortality. The infection spectrum and characteristics have evolved with the introduction of novel agents. An understanding of risk factors that increase susceptibility to infections is critical in fighting them. This retrospective investigation aimed to establish the incidence and main characteristics of infections in non-transplanted hospitalised myeloma patients in our department over a 3-year period, as well as factors associated with infections. MATERIALS AND METHODS A total of 240 hospitalised patients with multiple myeloma (120 males and 120 females; average age: 69 years, range: 41-89 years) who were diagnosed or treated in our department from January 2008 to December 2010 were included in this study and their data were retrospectively analysed. RESULTS Infections were identified in 17.9% of hospitalised patients. The most common pathogen found was Pseudomonas aeruginosa. The frequency of gram-positive and gram-negative pathogens was similar. In 37.2% of cases, the agent was not isolated. The most common sites of infections were the urinary system and the blood (septicemia). The frequency of infection increased with duration of disease and the rate of reinfection was 41.9%. The patients treated with bortezomib had the highest infection occurrence. Fatal outcome occurred in 9.3% of cases. CONCLUSION The factors associated with infections in this investigation were female sex, 3B clinical stage of disease, increased serum creatinine and ferritin levels, neutropenia, poor general condition, and presence of catheters. Myeloma patients with one or more of these mentioned risk factors should be monitored with particular care in order to decrease the incidence and severity of infective complications.
Collapse
Affiliation(s)
| | | | - Jelena Ivandić
- University Hospital Centre Rijeka, Clinic of Gynaecology and Obstetrics, Rijeka, Croatia Phone: +0038551421426 E-mail:
| | | | | | | | | |
Collapse
|
31
|
Dendritic cells in hematological malignancies. Crit Rev Oncol Hematol 2016; 108:86-96. [DOI: 10.1016/j.critrevonc.2016.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/05/2016] [Accepted: 10/18/2016] [Indexed: 01/17/2023] Open
|
32
|
Paulus A, Akhtar S, Caulfield TR, Samuel K, Yousaf H, Bashir Y, Paulus SM, Tran D, Hudec R, Cogen D, Jiang J, Edenfield B, Novak A, Ansell SM, Witzig T, Martin P, Coleman M, Roy V, Ailawadhi S, Chitta K, Linder S, Chanan-Khan A. Coinhibition of the deubiquitinating enzymes, USP14 and UCHL5, with VLX1570 is lethal to ibrutinib- or bortezomib-resistant Waldenstrom macroglobulinemia tumor cells. Blood Cancer J 2016; 6:e492. [PMID: 27813535 PMCID: PMC5148058 DOI: 10.1038/bcj.2016.93] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/21/2016] [Accepted: 08/22/2016] [Indexed: 01/30/2023] Open
Abstract
The survival of Waldenstrom macroglobulinemia (WM) tumor cells hinges on aberrant B-cell receptor (BCR) and MYD88 signaling. WM cells upregulate the proteasome function to sustain the BCR-driven growth while maintaining homeostasis. Clinically, two treatment strategies are used to disrupt these complementary yet mutually exclusive WM survival pathways via ibrutinib (targets BTK/MYD88 node) and bortezomib (targets 20 S proteasome). Despite the success of both agents, WM patients eventually become refractory to treatment, highlighting the adaptive plasticity of WM cells and underscoring the need for development of new therapeutics. Here we provide a comprehensive preclinical report on the anti-WM activity of VLX1570, a novel small-molecule inhibitor of the deubiquitinating enzymes (DUBs), ubiquitin-specific protease 14 (USP14) and ubiquitin carboxyl-terminal hydrolase isozyme L5 (UCHL5). Both DUBs reside in the 19 S proteasome cap and their inhibition by VLX1570 results in rapid and tumor-specific apoptosis in bortezomib- or ibrutinib-resistant WM cells. Notably, treatment of WM cells with VLX1570 downregulated BCR-associated elements BTK, MYD88, NFATC, NF-κB and CXCR4, the latter whose dysregulated function is linked to ibrutinib resistance. VLX1570 administered to WM-xenografted mice resulted in decreased tumor burden and prolonged survival (P=0.0008) compared with vehicle-treated mice. Overall, our report demonstrates significant value in targeting USP14/UCHL5 with VLX1570 in drug-resistant WM and carries a high potential for clinical translation.
Collapse
Affiliation(s)
- A Paulus
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.,Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - S Akhtar
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - T R Caulfield
- Department of Molecular Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - K Samuel
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - H Yousaf
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Y Bashir
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - S M Paulus
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - D Tran
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - R Hudec
- Department of Molecular Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - D Cogen
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - J Jiang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | - B Edenfield
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - A Novak
- Department of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - S M Ansell
- Department of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - T Witzig
- Department of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - P Martin
- Department of Medicine, Weill Cornell Medical College, Cornell, NY, USA
| | - M Coleman
- Department of Medicine, Weill Cornell Medical College, Cornell, NY, USA
| | - V Roy
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - S Ailawadhi
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - K Chitta
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - S Linder
- Institute for Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - A Chanan-Khan
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.,Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
33
|
Al-Homsi AS, Feng Y, Duffner U, Al Malki MM, Goodyke A, Cole K, Muilenburg M, Abdel-Mageed A. Bortezomib for the prevention and treatment of graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Exp Hematol 2016; 44:771-777. [PMID: 27224851 DOI: 10.1016/j.exphem.2016.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/06/2016] [Indexed: 01/13/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation is the standard treatment for a variety of benign and malignant conditions. However, graft-versus-host disease (GvHD) continues to present a major barrier to the success and wide applicability of this procedure. Although current GvHD prevention and treatment regimens exclusively target T cells, bortezomib, a reversible proteasome inhibitor, possesses unique immune regulatory activities that span a wide variety of cellular processes of T and dendritic cells essential for the development of GvHD. Herein, we review the current understanding of the effects of bortezomib in vitro and in animal models and summarize the clinical data relevant to its use in the prevention and treatment of GvHD. We conclude with an outline of the remaining challenges and opportunities to optimize bortezomib's potential role in this setting.
Collapse
Affiliation(s)
- Ahmad Samer Al-Homsi
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, MI, USA; Michigan State University College of Human Medicine, Grand Rapids, MI, USA.
| | - Yuxin Feng
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, MI, USA
| | - Ulrich Duffner
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, MI, USA; Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Monzr M Al Malki
- Department of Hematology and Hematopoietic Stem Cell Transplantation, City of Hope, Duarte, CA, USA
| | - Austin Goodyke
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, MI, USA
| | - Kelli Cole
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, MI, USA
| | - Marlee Muilenburg
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, MI, USA
| | - Aly Abdel-Mageed
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, MI, USA; Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| |
Collapse
|
34
|
Drug-induced modulation of T lymphocytes as a potential mechanism of susceptibility to infections in patients with multiple myeloma during bortezomib therapy. Cell Biochem Biophys 2016; 71:457-64. [PMID: 25343940 PMCID: PMC4289524 DOI: 10.1007/s12013-014-0224-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bortezomib is effective in the therapy of multiple myeloma (MM), but causes infections that are different from those associated with conventional chemotherapy. It is important to identify the risk factors that facilitate infections associated with bortezomib therapy. In the present report, we sought to (1) define the features of the infections associated with this therapy and (2) identify the immune mechanisms responsible for the observed susceptibility to these infections. We first retrospectively analyzed the clinical data of 143 patients who had received bortezomib therapy for MM. We then prospectively assessed the modulation of T lymphocyte status during this therapy, and evaluated potential relationships between infections and T lymphocyte changes. The infection rates peaked during the first cycle of bortezomib therapy (47.6 %) in patients with MM (p < 0.05 vs. subsequent cycles). Bortezomib therapy was associated with higher incidence rates of viral and fungal infections (15.8 %, p < 0.05 vs. conventional chemotherapy). In addition, patients with the IgG immunophenotype showed higher bacterial and viral infection rates (respectively, p = 0.008 and 0.009). The T lymphocyte numbers significantly decreased after bortezomib therapy (p < 0.05), and the same was true for the Th1/Th2 ratio (p < 0.01). Patients with MM who have decreased lymphocyte counts, while on bortezomib therapy are more likely to develop bacterial or viral infections. In addition, an imbalance in T lymphocyte subsets is also associated with bacterial or viral infections in these patients.
Collapse
|
35
|
Malek E, de Lima M, Letterio JJ, Kim BG, Finke JH, Driscoll JJ, Giralt SA. Myeloid-derived suppressor cells: The green light for myeloma immune escape. Blood Rev 2016; 30:341-8. [PMID: 27132116 DOI: 10.1016/j.blre.2016.04.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/28/2016] [Accepted: 04/01/2016] [Indexed: 01/04/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous, immature myeloid cell population with the ability to suppress innate and adaptive immune responses that promote tumor growth. MDSCs are increased in patients with multiple myeloma (MM) and have bidirectional interaction with tumors within the MM microenvironment. MM-MDSCs promote MM tumor growth and induce immune suppression; conversely, MM cells induce MDSC development and survival. Although the role of MDSCs in infections, inflammatory diseases and solid tumors has been extensively characterized, their tumor-promoting and immune-suppressive role in MM and the MM microenvironment is only beginning to emerge. The presence and activation of MDSCs in MM patients has been well documented; however, the direct actions and functional consequences of MDSCs on cancer cells is poorly defined. Immunosuppressive MDSCs play an important role in tumor progression primarily because of their capability to promote immune-escape, angiogenesis, drug resistance and metastasis. However, their role in the bone marrow (BM), the primary MM site, is poorly understood. MM remains an incurable malignancy, and it is likely that the BM microenvironment protects MM against chemotherapy agents and the host immune system. A growing body of evidence suggests that host immune cells with a suppressive phenotype contribute to a myeloma immunosuppressive network. Among the known suppressor cells, MDSCs and T regulatory cells (Tregs) have been found to be significantly increased in myeloma patients and their levels correlate with disease stage and clinical outcome. Furthermore, it has been shown that MDSC can mediate suppression of myeloma-specific T-cell responses through the induction of T-cell anergy and Treg development in the MM microenvironment. Here, we review clinical correlations and the preclinical proof-of-principle data on the role of MDSCs in myeloma immunotolerance and highlight the mechanistically relevant MDSC-targeted compounds and their potential utility in a new approach for anti-myeloma therapy.
Collapse
Affiliation(s)
- Ehsan Malek
- University Hospitals Case Medical Center, Seidman Cancer Center, Cleveland, OH, USA.
| | - Marcos de Lima
- University Hospitals Case Medical Center, Seidman Cancer Center, Cleveland, OH, USA
| | - John J Letterio
- Department of Pediatrics, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA; The Angie Fowler Adolescent & Young Adult Cancer Institute, Rainbow Babies & Children's Hospital, University Hospitals, Cleveland, OH, USA
| | - Byung-Gyu Kim
- Department of Pediatrics, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA; The Angie Fowler Adolescent & Young Adult Cancer Institute, Rainbow Babies & Children's Hospital, University Hospitals, Cleveland, OH, USA
| | - James H Finke
- Taussig Cancer Institute, Glickman Urological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James J Driscoll
- Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sergio A Giralt
- Adult Bone Marrow Transplant Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
36
|
Renteria AS, Levine JE, Ferrara JLM. Therapeutic targets and emerging treatment options in gastrointestinal acute graft-versus-host disease. Expert Opin Orphan Drugs 2016; 4:469-484. [PMID: 30057862 DOI: 10.1517/21678707.2016.1166949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction Graft-versus-host disease (GVHD) continues to be the major lethal complication of allogeneic hematopoietic stem cell transplantation (HCT) but the standard of care, high dose steroids, has not changed in 40 years. Approximately 50% of GVHD patients will develop steroid refractory disease, typically involving the gastrointestinal (GI) tract, which has a very poor prognosis. Newly developed GVHD biomarker-based risk scores provide the first opportunity to treat patients at the onset of symptoms according to risk of steroid failure. Furthermore, improvements in our understanding of the pathobiology of GVHD, its different signaling pathways, involved cytokines, and the role of post-translational and epigenetic modifications, has identified new therapeutic targets for clinical trials. Areas covered This manuscript summarizes the pathophysiology, diagnosis, staging, current and new targeted therapies for GVHD, with an emphasis on GI GVHD. A literature search on PubMed was undertaken and the most relevant references included. Expert Opinion The standard treatment for GVHD, high dose steroids, offers less than optimal outcomes as well as significant toxicities. Better treatments, especially for GI GVHD, are needed to reduce non-relapse mortality after allogeneic HCT. The identification of high risk patients through a biomarker-defined scoring system offers a personalized approach to a disease that still requires significant research attention.
Collapse
Affiliation(s)
- Anne S Renteria
- Blood and Marrow Transplantation Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John E Levine
- Blood and Marrow Transplantation Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James L M Ferrara
- Hematologic Malignancies Translational Research Center, Blood and Marrow Transplantation Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
37
|
Guillerey C, Nakamura K, Vuckovic S, Hill GR, Smyth MJ. Immune responses in multiple myeloma: role of the natural immune surveillance and potential of immunotherapies. Cell Mol Life Sci 2016; 73:1569-89. [PMID: 26801219 PMCID: PMC11108512 DOI: 10.1007/s00018-016-2135-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/07/2016] [Indexed: 02/06/2023]
Abstract
Multiple myeloma (MM) is a tumor of terminally differentiated B cells that arises in the bone marrow. Immune interactions appear as key determinants of MM progression. While myeloid cells foster myeloma-promoting inflammation, Natural Killer cells and T lymphocytes mediate protective anti-myeloma responses. The profound immune deregulation occurring in MM patients may be involved in the transition from a premalignant to a malignant stage of the disease. In the last decades, the advent of stem cell transplantation and new therapeutic agents including proteasome inhibitors and immunoregulatory drugs has dramatically improved patient outcomes, suggesting potentially key roles for innate and adaptive immunity in disease control. Nevertheless, MM remains largely incurable for the vast majority of patients. A better understanding of the complex interplay between myeloma cells and their immune environment should pave the way for designing better immunotherapies with the potential of very long term disease control. Here, we review the immunological microenvironment in myeloma. We discuss the role of naturally arising anti-myeloma immune responses and their potential corruption in MM patients. Finally, we detail the numerous promising immune-targeting strategies approved or in clinical trials for the treatment of MM.
Collapse
Affiliation(s)
- Camille Guillerey
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD, 4006, Australia
- School of Medicine, The University of Queensland, Herston Road, Herston, QLD, 4072, Australia
| | - Kyohei Nakamura
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD, 4006, Australia
| | - Slavica Vuckovic
- School of Medicine, The University of Queensland, Herston Road, Herston, QLD, 4072, Australia
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Mark J Smyth
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD, 4006, Australia.
- School of Medicine, The University of Queensland, Herston Road, Herston, QLD, 4072, Australia.
| |
Collapse
|
38
|
Shanker A, Pellom ST, Dudimah DF, Thounaojam MC, de Kluyver RL, Brooks AD, Yagita H, McVicar DW, Murphy WJ, Longo DL, Sayers TJ. Bortezomib Improves Adoptive T-cell Therapy by Sensitizing Cancer Cells to FasL Cytotoxicity. Cancer Res 2015; 75:5260-72. [PMID: 26494122 PMCID: PMC4681610 DOI: 10.1158/0008-5472.can-15-0794] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 09/23/2015] [Indexed: 01/15/2023]
Abstract
Cancer immunotherapy shows great promise but many patients fail to show objective responses, including in cancers that can respond well, such as melanoma and renal adenocarcinoma. The proteasome inhibitor bortezomib sensitizes solid tumors to apoptosis in response to TNF-family death ligands. Because T cells provide multiple death ligands at the tumor site, we investigated the effects of bortezomib on T-cell responses in immunotherapy models involving low-avidity antigens. Bortezomib did not affect lymphocyte or tissue-resident CD11c(+)CD8(+) dendritic cell counts in tumor-bearing mice, did not inhibit dendritic cell expression of costimulatory molecules, and did not decrease MHC class I/II-associated antigen presentation to cognate T cells. Rather, bortezomib activated NF-κB p65 in CD8(+) T cells, stabilizing expression of T-cell receptor CD3ζ and IL2 receptor-α, while maintaining IFNγ secretion to improve FasL-mediated tumor lysis. Notably, bortezomib increased tumor cell surface expression of Fas in mice as well as human melanoma tissue from a responsive patient. In renal tumor-bearing immunodeficient Rag2(-/-) mice, bortezomib treatment after adoptive T-cell immunotherapy reduced lung metastases and enhanced host survival. Our findings highlight the potential of proteasome inhibitors to enhance antitumor T-cell function in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee. Host-Tumor Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee. School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee.
| | - Samuel T Pellom
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee. School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee. Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, Nashville, Tennessee
| | - Duafalia F Dudimah
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee
| | - Menaka C Thounaojam
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, Tennessee
| | - Rachel L de Kluyver
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland
| | - Alan D Brooks
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland. Basic Sciences Program, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Daniel W McVicar
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland
| | - William J Murphy
- Division of Hematology/Oncology, Departments of Dermatology and Internal Medicine, University of California School of Medicine, Davis, California
| | - Dan L Longo
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Thomas J Sayers
- Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland. Basic Sciences Program, Leidos Biomedical Research, Inc., Frederick, Maryland.
| |
Collapse
|
39
|
Kim JW, Min CK, Mun YC, Park Y, Kim BS, Nam SH, Koh Y, Kwon JH, Choe PG, Park WB, Kim I. Varicella-zoster virus-specific cell-mediated immunity and herpes zoster development in multiple myeloma patients receiving bortezomib- or thalidomide-based chemotherapy. J Clin Virol 2015; 73:64-69. [DOI: 10.1016/j.jcv.2015.10.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/25/2015] [Accepted: 10/21/2015] [Indexed: 10/22/2022]
|
40
|
Koreth J, Kim HT, Lange PB, Bindra B, Reynolds CG, Chammas MJ, Armand P, Cutler CS, Ho VT, Glotzbecker B, Nikiforow S, Ritz J, Blazar BR, Soiffer RJ, Antin JH, Alyea EP. A Bortezomib-Based Regimen Offers Promising Survival and Graft-versus-Host Disease Prophylaxis in Myeloablative HLA-Mismatched and Unrelated Donor Transplantation: A Phase II Trial. Biol Blood Marrow Transplant 2015; 21:1907-13. [PMID: 26055298 DOI: 10.1016/j.bbmt.2015.05.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/30/2015] [Indexed: 11/25/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) recipients lacking HLA-matched related donors have increased graft-versus-host disease (GVHD) and nonrelapse mortality (NRM). Bortezomib added to reduced-intensity conditioning can offer benefit in T cell-replete HLA-mismatched HSCT and may also benefit myeloablative conditioning (MAC) transplants. We conducted a phase II trial of short-course bortezomib plus standard tacrolimus/methotrexate after busulfan/fludarabine MAC in 34 patients with predominantly myeloid malignancies. Fourteen (41%) received 8/8 HLA-matched unrelated donor (MUD) and 20 (59%) received 7/8 HLA-mismatched related/unrelated donor peripheral blood stem cell grafts. Median age was 49 years (range, 21 to 60), and median follow-up was 25 months (range, 11 to 36). The regimen was well tolerated. No dose modifications were required. Neutrophil and platelet engraftment occurred at a median of 14 (range, 10 to 33) and 17 (range, 10 to 54) days, respectively. Median 30-day donor chimerism was 99% (range, 90 to 100), and 100-day grades II to IV and III to IV acute GVHD incidence was 32% and 12% respectively. One-year chronic GVHD incidence was 50%. Two-year cumulative incidence of both NRM and relapse was 16%. Two-year progression-free and overall survival rates were 70% and 71%, respectively. Outcomes were comparable to an 8/8 MUD MAC cohort (n = 45). Immune reconstitution was robust. Bortezomib-based MAC HSCT is well tolerated, with HLA-mismatched outcomes comparable with 8/8 MUD MAC HSCT, and is suitable for randomized evaluation. (clinicaltrials.gov: NCT01323920.).
Collapse
Affiliation(s)
- John Koreth
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - Haesook T Kim
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute and Harvard School of Public Health, Boston, Massachusetts
| | - Paulina B Lange
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Bhavjot Bindra
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Carol G Reynolds
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Marie J Chammas
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Philippe Armand
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Corey S Cutler
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Vincent T Ho
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Brett Glotzbecker
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Sarah Nikiforow
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, University of Minnesota Masonic Cancer Center and Department of Pediatrics, Minneapolis, Minnesota
| | - Robert J Soiffer
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Joseph H Antin
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Edwin P Alyea
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
41
|
Hasegawa T, Aisa Y, Shimazaki K, Ito C, Nakazato T. Cytomegalovirus reactivation in patients with multiple myeloma. Eur J Haematol 2015; 96:78-82. [DOI: 10.1111/ejh.12551] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Tetsuo Hasegawa
- Department of Hematology; Yokohama Municipal Citizen's Hospital; Yokohama Japan
| | - Yoshinobu Aisa
- Department of Hematology; Yokohama Municipal Citizen's Hospital; Yokohama Japan
| | - Kengo Shimazaki
- Department of Hematology; Yokohama Municipal Citizen's Hospital; Yokohama Japan
| | - Chisako Ito
- Department of Hematology; Yokohama Municipal Citizen's Hospital; Yokohama Japan
| | - Tomonori Nakazato
- Department of Hematology; Yokohama Municipal Citizen's Hospital; Yokohama Japan
| |
Collapse
|
42
|
Al-Homsi AS, Cole K, Bogema M, Duffner U, Williams S, Mageed A. Short Course of Post-Transplantation Cyclophosphamide and Bortezomib for Graft-versus-Host Disease Prevention after Allogeneic Peripheral Blood Stem Cell Transplantation Is Feasible and Yields Favorable Results: A Phase I Study. Biol Blood Marrow Transplant 2015; 21:1315-20. [PMID: 25765556 DOI: 10.1016/j.bbmt.2015.02.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/09/2015] [Indexed: 10/23/2022]
Abstract
An effective graft-versus-host disease (GVHD) preventative approach that preserves the graft-versus-tumor effect after allogeneic hematopoietic stem cell transplantation (HSCT) remains elusive. Standard GVHD prophylactic regimens suppress T cells indiscriminately and are suboptimal. Conversely, post-transplantation high-dose cyclophosphamide selectively destroys proliferating alloreactive T cells, allows the expansion of regulatory T cells, and induces long-lasting clonal deletion of intrathymic antihost T cells. It has been successfully used to prevent GVHD after allogeneic HSCT. Bortezomib has antitumor activity on a variety of hematological malignancies and exhibits a number of favorable immunomodulatory effects that include inhibition of dendritic cells. Therefore, an approach that combines post-transplantation cyclophosphamide and bortezomib seems attractive. Herein, we report the results of a phase I study examining the feasibility and safety of high-dose post-transplantation cyclophosphamide in combination with bortezomib in patients undergoing allogeneic peripheral blood HSCT from matched siblings or unrelated donors after reduced-intensity conditioning. Cyclophosphamide was given at a fixed dose (50 mg/kg on days +3 and +4). Bortezomib dose was started at .7 mg/m2, escalated up to 1.3 mg/m2, and was administered on days 0 and +3. Patients receiving grafts from unrelated donors also received rabbit antithymocyte globulin. The combination was well tolerated and allowed prompt engraftment in all patients. The incidences of acute GVHD grades II to IV and grades III and IV were 20% and 6.7%, respectively. With a median follow-up of 9.1 months (range, 4.3 to 26.7), treatment-related mortality was 13.5% with predicted 2-year disease-free survival and overall survival of 55.7% and 68%, respectively. The study suggests that the combination of post-transplantation cyclophosphamide and bortezomib is feasible and may offer an effective and practical GVHD prophylactic regimen. The combination, therefore, merits further examination.
Collapse
Affiliation(s)
- Ahmad-Samer Al-Homsi
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan; Michigan State University, College of Human Medicine, Grand Rapids, Michigan.
| | - Kelli Cole
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan
| | - Marlee Bogema
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan
| | - Ulrich Duffner
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan; Michigan State University, College of Human Medicine, Grand Rapids, Michigan
| | - Stephanie Williams
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan; Michigan State University, College of Human Medicine, Grand Rapids, Michigan
| | - Aly Mageed
- Blood and Marrow Transplantation Program, Spectrum Health, Grand Rapids, Michigan; Michigan State University, College of Human Medicine, Grand Rapids, Michigan
| |
Collapse
|
43
|
Rund D. Bortezomib followed by autologous stem cell transplant for patients with hepatitis B positive multiple myeloma: a cautionary note. Leuk Lymphoma 2015; 56:1579-80. [PMID: 25629989 DOI: 10.3109/10428194.2015.1004671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Deborah Rund
- Hebrew University Hadassah Medical Organization , Ein Kerem, Jerusalem , Israel
| |
Collapse
|
44
|
Švajger U, Gobec M, Obreza A, Mlinarič-Raščan I. Novel N-amidinopiperidine-based proteasome inhibitor preserves dendritic cell functionality and rescues their Th1-polarizing capacity in Ramos-conditioned tumor environment. Cancer Immunol Immunother 2015; 64:15-27. [PMID: 25253531 PMCID: PMC11029559 DOI: 10.1007/s00262-014-1608-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 09/07/2014] [Indexed: 01/05/2023]
Abstract
The tumor microenvironment represents a burden that hampers the proper activation of immune cells, including the dendritic cells (DCs). It is, therefore, desired that the important characteristics of a given anticancer drug candidate be seen as consisting not solely of its antitumor properties, but that it also lacks potential side effects that could additionally constrain the development and function of immune cells associated with tumor immunity. We have previously identified compounds with a N-amidinopiperidine scaffold that selectively induce apoptosis in Burkitt's lymphoma cells through proteasome inhibition. Here, we demonstrate that SPI-15 affected neither the viability of DCs nor their differentiation. In addition, the compound had no significant effect on their cytokine secretion or allostimulatory capacity. Moreover, DC functionality in the context of tumor microenvironment was also unaffected, as demonstrated by experiments performed on DCs differentiated in Ramos-conditioned media in the presence or absence of SPI-15. The cytokine profile and functional assays revealed that SPI-15 rescues DC differentiation from the immunosuppressive environment produced by Ramos cells; this was seen by their reacquired ability to induce IFN-γ-secretion from naïve CD4(+)CD45RA(+) T cells and the consequently induced Th1-effector differentiation. Herein, we present novel characteristics of an N-amidinopiperidine-based protease inhibitor whose anticancer properties are not associated with the immunosuppression of DCs. We propose future studies toward the design of structurally similar compounds with the aim of developing potent anticancer drugs with minimal negative effects on crucial factors involved in tumor immunity.
Collapse
Affiliation(s)
- Urban Švajger
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000, Ljubljana, Slovenia,
| | | | | | | |
Collapse
|
45
|
May LJ, Yeh J, Maeda K, Tyan DB, Chen S, Kaufman BD, Bernstein D, Rosenthal DN, Hollander SA. HLA desensitization with bortezomib in a highly sensitized pediatric patient. Pediatr Transplant 2014; 18:E280-2. [PMID: 25174602 DOI: 10.1111/petr.12347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/30/2014] [Indexed: 11/28/2022]
Abstract
The proteasome inhibitor bortezomib has been used with variable success in the treatment of AMR following heart transplant. There is limited experience with this agent as a pretransplant desensitizing therapy. We report a case of successful HLA desensitization with a bortezomib-based protocol prior to successful heart transplantation. A nine-yr-old boy with dilated cardiomyopathy, not initially sensitized to HLA (cPRA of zero), required three days of ECMO, followed by implantation of a Heartmate II LVAD. Within six wk, the patient developed de novo class I IgG and C1q complement-fixing HLA antibodies with a cPRA of 100%. Two doses of IVIG (2 g/kg) failed to reduce antibody levels, although two courses of a novel desensitization protocol consisting of rituximab (375 mg/m(2) ), bortezomib (1.3 mg/m(2) × 5 doses), and plasmapheresis reduced his cPRA to 0% and 87% by the C1q and IgG assays, respectively. He underwent heart transplantation nearly two months later. The patient is now >one yr post-transplant, is free of both AMR and ACR, and has no detectable donor-specific antibodies by IgG or C1q. Proteasome inhibition with bortezomib and plasmapheresis may be an effective therapy for HLA desensitization pretransplant.
Collapse
Affiliation(s)
- Lindsay J May
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Marchesi F, Pimpinelli F, Dessanti ML, Gumenyuk S, Palombi F, Pisani F, Romano A, Spadea A, Maschio M, Ensoli F, Mengarelli A. Evaluation of risk of symptomatic cytomegalovirus reactivation in myeloma patients treated with tandem autologous stem cell transplantation and novel agents: a single-institution study. Transpl Infect Dis 2014; 16:1032-8. [PMID: 25369809 DOI: 10.1111/tid.12309] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 08/18/2014] [Accepted: 08/23/2014] [Indexed: 01/12/2023]
Abstract
The introduction of proteasome inhibitors and/or immunomodulators in the treatment of myeloma has led to an increase in viral infections, particularly in the Herpesviridae family. Previous studies about the risk of cytomegalovirus (CMV) reactivation after autologous stem cell transplantation (ASCT) have examined the clinical outcome after the first ASCT; however, only 1 study to date has investigated the risk of CMV reactivation after a second transplantation. To address this issue, we performed a retrospective chart review on 78 consecutive myeloma patients (median age 56 years) who underwent a tandem non-CD34(+) selected ASCT after induction treatment with either conventional chemotherapy (n = 42) or with novel agents (n = 36), respectively. All subjects had been mobilized and conditioned with cyclophosphamide plus granulocyte colony-stimulating factor and melphalan alone, respectively. CMV DNA load in the blood has been determined by polymerase chain reaction in the case of a clinical suspicion of CMV reactivation; therefore, routine monitoring was not performed. Considering the outcome of both the first and the second transplantations, we observed a total of 13 episodes of symptomatic CMV reactivation (13/156, 8%), in 12 subjects (12/78, 15%), all successfully treated. Eight subjects experienced a CMV reactivation after the first ASCT (8/78, 10%); however, only 1 of them (1/8, 12%) experienced a CMV reactivation after the second transplantation. Conversely, 4 CMV reactivations (6%) were observed after the second transplantation in the group of 70 patients who did not experience a CMV reactivation after the first ASCT. No statistically significant difference was observed between first and second ASCT (8/78, 10% vs. 5/78, 6%; P = 0.767). Univariate analysis showed that a pre-transplant treatment with novel agents was the only baseline factor significantly associated with the occurrence of post-ASCT CMV symptomatic reactivation after the first transplant (odds ratio [OR]: 9.897; 95% confidence interval [CI]: 1.154-84.840; P = 0.021) but not after the second transplant (OR: 5.125; 95% CI: 0.546-48.119; P = 0.115). No end-organ disease or primary infection was documented. Our data suggest that second transplantation does not increase the risk of CMV reactivation in our patient population, when compared with the first one, and confirm the role of a pre-transplant treatment with novel agents as a risk factor for CMV symptomatic reactivation.
Collapse
Affiliation(s)
- F Marchesi
- Hematology and Stem Cell Transplantation Unit, Regina Elena National Cancer Institute, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ciombor KK, Feng Y, Benson AB, Su Y, Horton L, Short SP, Kauh JSW, Staley C, Mulcahy M, Powell M, Amiri KI, Richmond A, Berlin J. Phase II trial of bortezomib plus doxorubicin in hepatocellular carcinoma (E6202): a trial of the Eastern Cooperative Oncology Group. Invest New Drugs 2014; 32:1017-27. [PMID: 24890858 PMCID: PMC4171216 DOI: 10.1007/s10637-014-0111-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/09/2014] [Indexed: 12/27/2022]
Abstract
PURPOSE To evaluate the efficacy and tolerability of bortezomib in combination with doxorubicin in patients with advanced hepatocellular carcinoma, and to correlate pharmacodynamic markers of proteasome inhibition with response and survival. EXPERIMENTAL DESIGN This phase II, open-label, multicenter study examined the efficacy of bortezomib (1.3 mg/m(2) IV on d1, 4, 8, 11) and doxorubicin (15 mg/m(2) IV on d1, 8) in 21-day cycles. The primary endpoint was objective response rate. RESULTS Best responses in 38 treated patients were 1 partial response (2.6 %), 10 (26.3 %) stable disease, and 17 (44.7 %) progressive disease; 10 patients were unevaluable. Median PFS was 2.2 months. Median OS was 6.1 months. The most common grade 3 to 4 toxicities were hypertension, glucose intolerance, ascites, ALT elevation, hyperglycemia and thrombosis/embolism. Worse PFS was seen in patients with elevated IL-6, IL-8, MIP-1α and EMSA for NF-κB at the start of treatment. Worse OS was seen in patients with elevated IL-8 and VEGF at the start of treatment. Patients had improved OS if a change in the natural log of serum MIP-1α/CCL3 was seen after treatment. RANTES/CCL5 levels decreased significantly with treatment. CONCLUSIONS The combination of doxorubicin and bortezomib was well-tolerated in patients with hepatocellular carcinoma, but the primary endpoint was not met. Exploratory analyses of markers of proteasome inhibition suggest a possible prognostic and predictive role and should be explored further in tumor types for which bortezomib is efficacious.
Collapse
Affiliation(s)
- Kristen K Ciombor
- Division of Medical Oncology, Department of Internal Medicine, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, A445A Starling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43212, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hosoba S, Jaye DL, Cohen C, Roback JD, Waller EK. Successful treatment of severe immune hemolytic anemia after allogeneic stem cell transplantation with bortezomib: report of a case and review of literature. Transfusion 2014; 55:259-64. [PMID: 25156334 PMCID: PMC4342762 DOI: 10.1111/trf.12815] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/22/2014] [Accepted: 06/26/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Immune hemolytic anemia is a well-known complication after allogeneic hematopoietic stem cell transplantation (HSCT). Posttransplant hemolytic anemia results in increased red blood cell transfusions and medical sequelae including iron overload. CASE REPORT We present a case report of immune hemolytic anemia that occurred after allogeneic HSCT from an ABO major-mismatched, HLA-matched unrelated donor. The patient had high anti-donor A type antibodies that were unresponsive to treatment with steroids and rituximab, resulting in persistent transfusion dependence. A detailed time course of anti-A titers, plasma cell content of the marrow, and B-cell content of the blood is presented. Treatment with bortezomib, a protease inhibitor, eliminated residual host-type plasma cells secreting anti-A and restored normal donor-derived erythropoiesis. CONCLUSION This report, and a review of literature for treatment of immune hemolytic anemia after allogeneic HSCT, supports the utility of bortezomib as plasma cell-targeted therapy in this setting.
Collapse
Affiliation(s)
- Sakura Hosoba
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | | | | | | | | |
Collapse
|
49
|
Herrera AF, Kim HT, Bindra B, Jones KT, Alyea EP, Armand P, Cutler CS, Ho VT, Nikiforow S, Blazar BR, Ritz J, Antin JH, Soiffer RJ, Koreth J. A phase II study of bortezomib plus prednisone for initial therapy of chronic graft-versus-host disease. Biol Blood Marrow Transplant 2014; 20:1737-43. [PMID: 25017765 DOI: 10.1016/j.bbmt.2014.06.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 06/30/2014] [Indexed: 12/21/2022]
Abstract
Chronic graft-versus-host disease (GVHD) induces significant morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Corticosteroids are standard initial therapy, despite limited efficacy and long-term toxicity. Based on our experience using bortezomib as effective acute GVHD prophylaxis, we hypothesized that proteasome-inhibition would complement the immunomodulatory effects of corticosteroids to improve outcomes in chronic GVHD (cGVHD). We undertook a single-arm phase II trial of bortezomib plus prednisone for initial therapy of cGVHD. Bortezomib was administered at 1.3 mg/m(2) i.v. on days 1, 8, 15, and 22 of each 35-day cycle for 3 cycles (15 weeks). Prednisone was dosed at .5 to 1 mg/kg/day, with a suggested taper after cycle 1. All 22 enrolled participants were evaluable for toxicity; 20 were evaluable for response. Bortezomib plus prednisone therapy was well tolerated, with 1 occurrence of grade 3 sensory peripheral neuropathy possibly related to bortezomib. The overall response rate at week 15 in evaluable participants was 80%, including 2 (10%) complete and 14 (70%) partial responses. The organ-specific complete response rate was 73% for skin, 53% for liver, 75% for gastrointestinal tract, and 33% for joint, muscle, or fascia involvement. The median prednisone dose decreased from 50 mg/day to 20 mg/day at week 15 (P < .001). The combination of bortezomib and prednisone for initial treatment of cGVHD is feasible and well tolerated. We observed a high response rate to combined bortezomib and prednisone therapy; however, in this single-arm study, we could not directly measure the impact of bortezomib. Proteasome inhibition may offer benefit in the treatment of cGVHD and should be further evaluated.
Collapse
Affiliation(s)
- Alex F Herrera
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Haesook T Kim
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Bhavjot Bindra
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kyle T Jones
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Edwin P Alyea
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Philippe Armand
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Corey S Cutler
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Vincent T Ho
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sarah Nikiforow
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Bruce R Blazar
- Blood and Marrow Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joseph H Antin
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Robert J Soiffer
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - John Koreth
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
50
|
Chen C, Meng Y, Wang L, Wang HX, Tian C, Pang GD, Li HH, Du J. Ubiquitin-activating enzyme E1 inhibitor PYR41 attenuates angiotensin II-induced activation of dendritic cells via the IκBa/NF-κB and MKP1/ERK/STAT1 pathways. Immunology 2014; 142:307-19. [PMID: 24456201 DOI: 10.1111/imm.12255] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/24/2013] [Accepted: 01/17/2014] [Indexed: 12/26/2022] Open
Abstract
The activation of dendritic cells (DCs) is necessary to initiate immune responses. Angiotensin II (Ang II) can enhance the maturation and activation of DCs, but the mechanisms are still unclear. Ubiquitin-activating enzyme (E1/Uba1) is the common first step in ubiquitylation, which decides whether or not the modified protein is ultimately degraded by the proteasome. This study aimed to investigate the role of E1 in Ang II-induced activation of DCs and the underlying mechanisms. First, we showed that Ang II stimulation significantly up-regulated E1 expression in DCs. Moreover, Ang II treatment markedly induced phenotypic maturation, the secretion of cytokines and the immunostimulatory capacity of DCs. In contrast, inhibition of E1 by a small molecule inhibitor, 4 [4-(5-nitro-furan-2-ylmethylene)-3, 5-dioxo-pyrazolidin-1-yl]-benzoic acid ethyl ester (PYR41), markedly attenuated these effects. Mechanistically, PYR41 treatment markedly decreased K63-linked ubiquitination of tumour necrosis factor receptor-associated factor 6 and nuclear factor-κB essential modulator, inhibited proteasomal degradation of nuclear factor-κB inhibitor α and mitogen-activated protein kinase phosphatase 1 thereby resulting in activation of nuclear factor-κB, extracellular signal-regulated kinase 1/2 and signal transducer and activator of transcription 1 signalling pathways in DCs induced by Ang II. Taken together, our results demonstrate a novel role of E1 in Ang II-induced activation of DCs, and inhibition of E1 activity might be a potential therapeutic target for DC-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Chen Chen
- Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodelling-related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|