1
|
The Proteasome Inhibitor Bortezomib Induces Apoptosis and Activation in Gel-Filtered Human Platelets. Int J Mol Sci 2021; 22:ijms22168955. [PMID: 34445660 PMCID: PMC8396574 DOI: 10.3390/ijms22168955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022] Open
Abstract
Bortezomib (BTZ) has demonstrated its efficacy in several hematological disorders and has been associated with thrombocytopenia. There is controversy about the effect of BTZ on human platelets, so we set out to determine its effect on various types of platelet samples. Human platelets were investigated in platelet-rich plasma (PRP) and as gel-filtered platelets (GFPs). Mitochondrial inner membrane potential depolarization and phosphatidylserine (PS) and P-selectin expression levels were studied by flow cytometry, while thrombin generation was measured by a fluorescent method. In PRP, BTZ caused negligible PS expression after 60 min of treatment. However, in GFPs, PS expression was dose- and time-dependently increased in the BTZ-treated groups, as was P-selectin. The percentage of depolarized cells was also higher after BTZ pretreatment at both time points. Peak thrombin and velocity index increased significantly even with the lowest BTZ concentration (p = 0.0019; p = 0.0032) whereas time to peak and start tail parameters decreased (p = 0.0007; p = 0.0034). The difference between PRP and GFP results can be attributed to the presence of plasma proteins in PRP, as the PS-stimulating effect of BTZ could be attenuated by supplementing GFPs with purified human albumin. Overall, BTZ induces a procoagulant platelet phenotype in an experimental setting devoid of plasma proteins.
Collapse
|
2
|
Knops N, Emonds MP, Herman J, Levtchenko E, Mekahli D, Pirenne J, Van Geet C, Dierickx D. Bortezomib for autoimmune hemolytic anemia after intestinal transplantation. Pediatr Transplant 2020; 24:e13700. [PMID: 32166874 DOI: 10.1111/petr.13700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/03/2019] [Accepted: 02/24/2020] [Indexed: 01/19/2023]
Abstract
AIHA is rare in the general population and associated with a mortality of 8%. In contrast, AIHA occurs in up to 12.2% of cases after intestinal transplantation and is associated with mortality up to 50%. Treatment entails a "step-up" approach including corticosteroids, IvIg, plasmapheresis, and rituximab. However, AIHA after transplantation often is refractory to this strategy, contributing to a poor outcome. We describe a child with microvillous inclusion disease who developed AIHA 1 year after multivisceral transplantation that was refractory to standard therapy and was subsequently treated with bortezomib.We observed remission of AIHA within 1 week after the start of bortezomib. Bortezomib was associated with transient diarrhea, leucopenia, and elevated liver enzymes. Three years later, he remains in remission without important complications. Published data on bortezomib for autoimmune cytopenias outside SOT are discussed. This is the first report to support bortezomib as an important therapeutic alternative for AIHA after SOT. The occurrence and treatment of AIHA after SOT, and specifically intestinal transplantation, should be the subject of future registry studies to collect additional experience and explore the optimal therapeutic approach.
Collapse
Affiliation(s)
- Noël Knops
- Pediatrics (Pediatric Nephrology and Solid Organ Transplantation), University Hospitals Leuven, Leuven, Belgium.,Department of Development and Regeneration (Woman and Child), KU Leuven, Leuven, Belgium
| | - Marie-Paule Emonds
- Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jean Herman
- Pediatrics (Pediatric Nephrology and Solid Organ Transplantation), University Hospitals Leuven, Leuven, Belgium.,Department of Development and Regeneration (Woman and Child), KU Leuven, Leuven, Belgium
| | - Elena Levtchenko
- Pediatrics (Pediatric Nephrology and Solid Organ Transplantation), University Hospitals Leuven, Leuven, Belgium.,Department of Development and Regeneration (Woman and Child), KU Leuven, Leuven, Belgium
| | - Djalila Mekahli
- Pediatrics (Pediatric Nephrology and Solid Organ Transplantation), University Hospitals Leuven, Leuven, Belgium.,Department of Development and Regeneration (Woman and Child), KU Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Chris Van Geet
- Pediatrics (Pediatric Hemato-Oncology), University Hospitals Leuven, Leuven, Belgium.,Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Daan Dierickx
- Hematology, University Hospitals Leuven, Leuven, Belgium.,Department of Oncology (Laboratorium of Experimental Hematology), KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Yao HW, Wang LC, Tsai HY, Fang YH, Zheng C, Chen SH, Hsu SM. Bortezomib induces HSV-1 lethality in mice with neutrophil deficiency. J Leukoc Biol 2019; 107:105-112. [PMID: 31729784 DOI: 10.1002/jlb.4ab1019-495r] [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: 12/19/2018] [Revised: 10/05/2019] [Accepted: 11/04/2019] [Indexed: 11/08/2022] Open
Abstract
Bortezomib suppressing NF-κB activity is an effective therapy for patients with myeloma or lymphoma. However, this drug can cause adverse effects, neutropenia, and recurrent infections of herpes viruses. Among herpes viruses, HSV-1 can reactivate to induce mortality. The important issues regarding how bortezomib diminishes neutrophils, whether bortezomib can induce HSV-1 reactivation, and how bortezomib exacerbates HSV-1 infection, need investigation. Using the murine model, we found that bortezomib induced HSV-1 reactivation. Bortezomib diminished neutrophil numbers in organs of uninfected and HSV-1-infected mice and turned a nonlethal infection to lethal with elevated tissue viral loads. In vitro results showed that bortezomib and HSV-1 collaborated to enhance the death and apoptosis of mouse neutrophils. The leukocyte deficiency induced by chemotherapies is generally believed to be the cause for aggravating virus infections. Here we show the potential of pathogen to exacerbate chemotherapy-induced leukocyte deficiency.
Collapse
Affiliation(s)
- Hui-Wen Yao
- Department of Microbiology and Immunology and Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Chiu Wang
- Department of Microbiology and Immunology and Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsien-Yang Tsai
- Department of Ophthalmology, Tzu Chi Hospital, Taichung, Taiwan
| | - Yi-Hsuan Fang
- Department of Ophthalmology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Shun-Hua Chen
- Department of Microbiology and Immunology and Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Min Hsu
- Department of Ophthalmology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
4
|
Basler M, Claus M, Klawitter M, Goebel H, Groettrup M. Immunoproteasome Inhibition Selectively Kills Human CD14 + Monocytes and as a Result Dampens IL-23 Secretion. THE JOURNAL OF IMMUNOLOGY 2019; 203:1776-1785. [PMID: 31484727 DOI: 10.4049/jimmunol.1900182] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023]
Abstract
MECL-1 (β2i), LMP2 (β1i), and LMP7 (β5i) are the proteolytically active subunits of the immunoproteasome (IP), a special type of proteasome mainly expressed in hematopoietic cells. Targeting the IP in autoimmune diseases proved to be therapeutically effective in preclinical mouse models. In endotoxin-stimulated human PBMCs, IP inhibition reduces the secretion of several proinflammatory cytokines, with the suppression of IL-23 being the most prominent. In this study, we investigated why the production of IL-23, a key mediator of inflammation in autoimmunity, is blocked when the IP is inhibited in LPS-stimulated human PBMCs. CD14+ monocytes could be identified as the main producers of IL-23 in LPS-stimulated PBMCs. We found that IP inhibition with the irreversible LMP7/LMP2 inhibitor ONX 0914 induced apoptosis in CD14+ monocytes, whereas CD4+, CD3+, CD19+, and CD56+ cells remained unaffected. A high expression of IPs renders monocytes susceptible to IP inhibition, leading to an accumulation of polyubiquitylated proteins and the induction of the unfolded protein response. Similar to IP inhibition, inducers of the unfolded protein response selectively kill CD14+ monocytes in human PBMCs. The blockage of the translation in CD14+ monocytes protects these cells from ONX 0914-induced cell death, indicating that the IP is required to maintain protein turnover in monocytes. Taken together, our data reveal why IP inhibition is particularly effective in the suppression of IL-23-driven autoimmunity.
Collapse
Affiliation(s)
- Michael Basler
- Division of Immunology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany; and .,Biotechnology Institute Thurgau at the University of Konstanz, 8280 Kreuzlingen, Switzerland
| | - Meike Claus
- Division of Immunology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany; and
| | - Moritz Klawitter
- Division of Immunology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany; and
| | - Heike Goebel
- Division of Immunology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany; and
| | - Marcus Groettrup
- Division of Immunology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany; and.,Biotechnology Institute Thurgau at the University of Konstanz, 8280 Kreuzlingen, Switzerland
| |
Collapse
|
5
|
Pletinckx K, Vaßen S, Schlusche I, Nordhoff S, Bahrenberg G, Dunkern TR. Inhibiting the immunoproteasome's β5i catalytic activity affects human peripheral blood-derived immune cell viability. Pharmacol Res Perspect 2019; 7:e00482. [PMID: 31236277 PMCID: PMC6581949 DOI: 10.1002/prp2.482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/01/2018] [Accepted: 04/05/2019] [Indexed: 01/03/2023] Open
Abstract
Small molecule inhibitors selectively targeting the immunoproteasome subunit β5i are currently being developed for the treatment of autoimmune disorders. However, patients carrying loss-of-function mutations in the gene encoding β5i (Psmb8) suffer from the proteasome-associated autoinflammatory syndromes (PRAAS) emphasizing the need to study pharmacological inhibition of immunoproteasome function in human cells. Here, we characterized the immunomodulatory potential of the selective β5i inhibitor ONX 0914 and Bortezomib, a pan-proteasome inhibitor, in human peripheral blood mononuclear cells (PBMCs). Both compounds efficiently blocked pro-inflammatory cytokine secretion in human whole blood and PBMC cultures stimulated with toll-like receptor (TLR) agonists. Furthermore, the compounds inhibited T cell cytokine production induced by recall antigen CMVpp65 or by polyclonal stimulation. The viability of PBMCs, however, was rapidly decreased in the presence of ONX 0914 and Bortezomib demonstrated by decreased residual cytosolic ATP and increased Annexin V surface binding. Interestingly, HLA-DR + monocytes were rapidly depleted from the cultures in the presence of ONX 0914 as a β5i-selective inhibitor and Bortezomib. In conclusion, the anti-inflammatory potential of β5i-selective inhibitors is correlating with a cytotoxicity increase in human PBMC subsets ex vivo. Our results provide important insights into the anti-inflammatory mechanism of action of β5i-inhibitors which currently hold the promise as a novel therapy for autoinflammatory diseases.
Collapse
|
6
|
Chonat S, Arthur CM, Zerra PE, Maier CL, Jajosky RP, Yee MEM, Miller MJ, Josephson CD, Roback JD, Fasano R, Stowell SR. Challenges in preventing and treating hemolytic complications associated with red blood cell transfusion. Transfus Clin Biol 2019; 26:130-134. [PMID: 30979566 DOI: 10.1016/j.tracli.2019.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Red blood cell (RBC) transfusion support represents a critical component of sickle cell disease (SCD) management. However, as with any therapeutic intervention, RBC transfusion is not without risk. Repeat exposure to allogeneic RBCs can result in the development of RBC alloantibodies that can make it difficult to find compatible RBCs for future transfusions and can directly increase the risk of developing acute or delayed hemolytic transfusion reactions, which can be further complicated by hyperhemolysis. Several prophylactic and treatment strategies have been employed in an effort to reduce or prevent hemolytic transfusion reactions. However, conflicting data exist regarding the efficacy of many of these approaches. We will explore the challenges associated with predicting, preventing and treating different types of hemolytic transfusion reactions in patients with SCD in addition to describing future strategies that may aid in the management of the complex transfusion requirements of SCD patients.
Collapse
Affiliation(s)
- Satheesh Chonat
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Connie M Arthur
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, 101, Woodruff Circle, 30322 Atlanta, GA, USA
| | - Patricia E Zerra
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, 101, Woodruff Circle, 30322 Atlanta, GA, USA
| | - Cheryl L Maier
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, 101, Woodruff Circle, 30322 Atlanta, GA, USA
| | - Ryan P Jajosky
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, 101, Woodruff Circle, 30322 Atlanta, GA, USA
| | - Marianne E M Yee
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Maureen J Miller
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, 101, Woodruff Circle, 30322 Atlanta, GA, USA
| | - Cassandra D Josephson
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, 101, Woodruff Circle, 30322 Atlanta, GA, USA
| | - John D Roback
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, 101, Woodruff Circle, 30322 Atlanta, GA, USA
| | - Ross Fasano
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, and Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, 101, Woodruff Circle, 30322 Atlanta, GA, USA.
| | - Sean R Stowell
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, 101, Woodruff Circle, 30322 Atlanta, GA, USA.
| |
Collapse
|
7
|
Dean CL, Maier CL, Chonat S, Chang A, Carden MA, El Rassi F, McLemore ML, Stowell SR, Fasano RM. Challenges in the treatment and prevention of delayed hemolytic transfusion reactions with hyperhemolysis in sickle cell disease patients. Transfusion 2019; 59:1698-1705. [PMID: 30848512 DOI: 10.1111/trf.15227] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/13/2018] [Accepted: 01/08/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Delayed hemolytic transfusion reactions (DHTRs) are serious complications of RBC transfusion that can occur in previously alloimmunized patients. Patients who require episodic transfusions during heightened inflammatory states, such as patients with sickle cell disease (SCD), are particularly prone to alloimmunization and developing DHTRs with hyperhemolysis. While efforts to mitigate these hemolytic episodes via immunosuppressive drugs can be employed, the relative efficacy of various treatment options remains incompletely understood. CASE REPORTS In this study, we explored five patients with SCD and multiple RBC alloantibodies who received various forms of immunosuppressive therapy in an attempt to prevent or treat severe DHTRs. RESULTS The clinical course for these five patients provides insight into the difficulty of effectively treating and preventing DHTRs in patients with SCD with currently available immunosuppressive therapies. CONCLUSION Based on our experience, and the current literature, it is difficult to predict the potential impact of various immunosuppressive therapies when seeking to prevent or treat DHTRs. Future mechanistic studies are needed to identify the optimal treatment options for DHTRs in the presence or absence of distinct alloantibodies in patients with SCD.
Collapse
Affiliation(s)
- Christina L Dean
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Cheryl L Maier
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Satheesh Chonat
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Andres Chang
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Marcus A Carden
- Department of Pediatrics and Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Fuad El Rassi
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Morgan L McLemore
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Sean R Stowell
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ross M Fasano
- Center for Transfusion and Cellular Therapy, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia.,Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| |
Collapse
|
8
|
On the role of the immunoproteasome in transplant rejection. Immunogenetics 2018; 71:263-271. [PMID: 30220008 DOI: 10.1007/s00251-018-1084-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/04/2018] [Indexed: 12/14/2022]
Abstract
The immunoproteasome is expressed in cells of hematopoietic origin and is induced during inflammation by IFN-γ. Targeting the immunoproteasome with selective inhibitors has been shown to be therapeutically effective in pre-clinical models for autoimmune diseases, colitis-associated cancer formation, and transplantation. Immunoproteasome inhibition prevents activation and proliferation of lymphocytes, lowers MHC class I cell surface expression, reduces the expression of cytokines of activated immune cells, and curtails T helper 1 and 17 cell differentiation. This might explain the in vivo efficacy of immunoproteasome inhibition in different pre-clinical disease models for autoimmunity, cancer, and transplantation. In this review, we summarize the effect of immunoproteasome inhibition in different animal models for transplantation.
Collapse
|
9
|
Liang YY, Schwarzinger I, Simonitsch-Klupp I, Agis H, Oehler R. Impaired efferocytosis by monocytes in multiple myeloma. Oncol Lett 2018; 16:409-416. [PMID: 29928429 DOI: 10.3892/ol.2018.8620] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/08/2017] [Indexed: 01/18/2023] Open
Abstract
Efficient clearance of apoptotic cells by efferocytosis is important for tissue homeostasis. Impaired efferocytosis leads to the accumulation of cell debris, which is regarded as a trigger in chronic inflammation and autoimmune diseases. Patients with hematological neoplastic disorders such as multiple myeloma (MM) exhibit high blood levels of apoptotic microparticles. The present study investigated whether these high levels of apoptotic microparticles are associated with insufficient dead cell clearance. Blood samples were collected from patients with MM immediately prior to and 3, 7 and 10 days after the initial cycle of bortezomib-based therapy. In addition, bone marrow aspirates (BMA) were collected prior to and following therapy. Prior to therapy, a 52% reduction in efferocytosis by blood monocytes was observed compared with the healthy controls (P<0.017). This was associated with an elevated number of 7-AAD+ dead cell remnants in the blood flow as well as in BMA. A portion of the blood samples contained active caspase 3. The subsequent bortezomib-based therapy had no effect on efferocytosis, although the quantity of dead cell remnants decreased. This reduction was associated with a decline in cluster of differentiation 8 (CD8)+ and CD4+ T cells and an increase in the number of monocytes. However, of 28 distinct soluble immune-modulating molecules (i.e. chemokines, cytokines and soluble co-stimulators) only C-C motif chemokine ligand 2 (CCL2), CCL24 and sCD27 were affected by bortezomib-based therapy. The levels of all other molecules remained unchanged or were below the detection threshold in all samples. The present study results revealed that the presence of dead cell remnants in the blood and bone morrow of patients with MM is associated with impaired efferocytosis by monocytes; however, its contribution to inflammatory events during MM remains unclear.
Collapse
Affiliation(s)
- Ying Yu Liang
- Department of Surgery and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ilse Schwarzinger
- Department of Laboratory Medicine, Medical University of Vienna, A-1090 Vienna, Austria
| | | | - Hermine Agis
- Department of Internal Medicine I and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Rudolf Oehler
- Department of Surgery and Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
10
|
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: 2.5] [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
|
11
|
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: 1.6] [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
|
12
|
Type-2 cannabinoid receptor regulates proliferation, apoptosis, differentiation, and OPG/RANKL ratio of MC3T3-E1 cells exposed to Titanium particles. Mol Cell Biochem 2014; 399:131-41. [DOI: 10.1007/s11010-014-2240-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 10/01/2014] [Indexed: 12/30/2022]
|
13
|
Trabanelli S, Očadlíková D, Ciciarello M, Salvestrini V, Lecciso M, Jandus C, Metz R, Evangelisti C, Laury-Kleintop L, Romero P, Prendergast GC, Curti A, Lemoli RM. The SOCS3-independent expression of IDO2 supports the homeostatic generation of T regulatory cells by human dendritic cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:1231-40. [PMID: 24391212 DOI: 10.4049/jimmunol.1300720] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Dendritic cells (DCs) are professional APCs that have a role in the initiation of adaptive immune responses and tolerance. Among the tolerogenic mechanisms, the expression of the enzyme IDO1 represents an effective tool to generate T regulatory cells. In humans, different DC subsets express IDO1, but less is known about the IDO1-related enzyme IDO2. In this study, we found a different pattern of expression and regulation between IDO1 and IDO2 in human circulating DCs. At the protein level, IDO1 is expressed only in circulating myeloid DCs (mDCs) and is modulated by PGE2, whereas IDO2 is expressed in both mDCs and plasmacytoid DCs and is not modulated by PGE2. In healthy subjects, IDO1 expression requires the presence of PGE2 and needs continuous transcription and translation, whereas IDO2 expression is constitutive, independent from suppressor of cytokine signaling 3 activity. Conversely, in patients suffering from inflammatory arthritis, circulating DCs express both IDO1 and IDO2. At the functional level, both mDCs and plasmacytoid DCs generate T regulatory cells through an IDO1/IDO2-dependent mechanism. We conclude that, in humans, whereas IDO1 provides an additional mechanism of tolerance induced by proinflammatory mediators, IDO2 is stably expressed in steady-state conditions and may contribute to the homeostatic tolerogenic capacity of DCs.
Collapse
Affiliation(s)
- Sara Trabanelli
- Department of Specialistic, Diagnostic, and Experimental Medicine, Institute of Hematology "Seràgnoli," University of Bologna, 40138 Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Protection against titanium particle-induced inflammatory osteolysis by the proteasome inhibitor bortezomib in vivo. Inflammation 2013; 35:1378-91. [PMID: 22391745 DOI: 10.1007/s10753-012-9451-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Wear particle-induced vascularized granulomatous inflammation and subsequent inflammatory osteolysis is the most common cause of aseptic loosening after total joint replacement (TJR); however, the precise mechanism by which this occurs is unclear. This study investigates the effects of the proteasome inhibitor bortezomib (Bzb) on the expression of key biochemical markers of bone metabolism and vascularised granulomatous tissues, such as receptor activator of nuclear factor-κB ligand (RANKL), osteoprotegerin (OPG), vascular endothelial growth factor (VEGF) and tumor necrosis factor receptor-associated factor 6 (TRAF6). In addition, the effect of Bzb on apoptosis of CD68+ cells was examined. A total of 32 female BALB/C mice were randomly divided into four groups. After implantation of calvaria bone from syngeneic littermates, titanium (Ti) particles were injected into established air pouches for all mice (excluding negative controls) to provoke inflammatory osteolysis. Subsequently, Bzb was administered at a ratio of 0, 0.1, or 0.5 mg/kg on day 1, 4, 8, and 11 post-surgery to alleviate this response. All of the air pouches were harvested 14 days after the surgical procedure and were processed for molecular and histological analysis. The results demonstrated that Ti injection elevated the expression of RANKL, OPG, VEGF, and TRAF6 at both the gene and protein levels, increased counts of infiltrated cells and thickness of air pouch membranes, and elevated the apoptosis index (AI) of CD68+ cells. Bzb treatment significantly improved Ti particle-induced implanted bone osteolysis, attenuated vascularised granulomatous tissues and elevated AI of CD68+ cells. Therefore, the proteasome pathway may represent an effective therapeutic target for the prevention and treatment of aseptic loosening.
Collapse
|
15
|
Targeting multiple-myeloma-induced immune dysfunction to improve immunotherapy outcomes. Clin Dev Immunol 2012; 2012:196063. [PMID: 22567028 PMCID: PMC3332181 DOI: 10.1155/2012/196063] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Revised: 01/12/2012] [Accepted: 01/29/2012] [Indexed: 12/16/2022]
Abstract
Multiple myeloma (MM) is a plasma cell malignancy associated with high levels of monoclonal (M) protein in the blood and/or serum. MM can occur de novo or evolve from benign monoclonal gammopathy of undetermined significance (MGUS). Current translational research into MM focuses on the development of combination therapies directed against molecularly defined targets and that are aimed at achieving durable clinical responses. MM cells have a unique ability to evade immunosurveillance through several mechanisms including, among others, expansion of regulatory T cells (Treg), reduced T-cell cytotoxic activity and responsiveness to IL-2, defects in B-cell immunity, and induction of dendritic cell (DC) dysfunction. Immune defects could be a major cause of failure of the recent immunotherapy trials in MM. This article summarizes our current knowledge on the molecular determinants of immune evasion in patients with MM and highlights how these pathways can be targeted to improve patients' clinical outcome.
Collapse
|
16
|
Mast cells promote the growth of Hodgkin's lymphoma cell tumor by modifying the tumor microenvironment that can be perturbed by bortezomib. Leukemia 2012; 26:2269-76. [PMID: 22430634 DOI: 10.1038/leu.2012.81] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hodgkin's lymphoma is frequently associated with mast cell infiltration that correlates directly with disease severity, but the mechanisms underlying this relationship remain unclear. Here, we report that mast cells promote the growth of Hodgkin's tumor by modifying the tumor microenvironment. A transplantation assay shows that primary murine mast cells accelerate tumor growth by established Hodgkin's cell lines, and promote marked neovascularization and fibrosis. Both mast cells and Hodgkin's cells were sensitive to bortezomib, but mast cells were more resistant to bortezomib. However, bortezomib inhibited degranulation, PGE(2)-induced rapid release of CCL2, and continuous release of vascular endothelial growth factor-A from mast cells even at the concentration that did not induce cell death. Bortezomib-treated mast cells lost the ability to induce neovasculization and fibrosis, and did not promote the growth of Hodgkin tumor in vivo. These results provide further evidence supporting causal relationships between inflammation and tumor growth, and demonstrate that bortezomib can target the tumor microenvironment.
Collapse
|
17
|
The humanized anti-HLA-DR moAb, IMMU-114, depletes APCs and reduces alloreactive T cells: implications for preventing GVHD. Bone Marrow Transplant 2011; 47:967-80. [PMID: 22020022 DOI: 10.1038/bmt.2011.203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In contrast to the conventional immunosuppressive agents and nonselective T-cell-depleting antibodies, selective depletion of donor alloreactive T cells and/or host APCs, particularly DCs, represents a novel approach that can effectively control GVHD with less or no impairment of T-cell-mediated antiviral and GVL immunity. Here we report that IMMU-114, a humanized anti-human leukocyte antigen-DR (HLA-DR) moAb, efficiently depleted human PBMCs of all APCs, including B cells, monocytes, myeloid DC type-1 (mDC1), mDC2 and plasmacytoid DCs (pDCs). Early and late apoptosis of mDC1, mDC2 and pDCs, and late apoptosis of all APC subsets, were increased by IMMU-114 treatment. Although IMMU-114 had little, if any, effect on the survival and apoptosis of non-B lymphocytes (>80% of which are T cells and ∼1-2% of T cells express HLA-DR), it selectively inhibited the proliferation of purified HLA-DR(+) T cells rather than HLA-DR(-) T cells. As a consequence, IMMU-114 treatment resulted in suppressed T-cell proliferation and reduced CD25(+) alloreactive T cells in allogeneic MLRs. Given the critical roles of APCs and alloreactive T cells in the pathogenesis of GVHD, these results suggest that IMMU-114 may have therapeutic potential against GVHD.
Collapse
|
18
|
Breccia M, Alimena G. NF-κB as a potential therapeutic target in myelodysplastic syndromes and acute myeloid leukemia. Expert Opin Ther Targets 2011; 14:1157-76. [PMID: 20858024 DOI: 10.1517/14728222.2010.522570] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IMPORTANCE OF THE FIELD The inactive NF-κB-inhibitor of NF-κB (IκB) complex is activated by stimuli including pro-inflammatory cytokines, mitogens, growth factors and stress-inducing agents. The release of NF-κB facilitates its translocation to the nucleus, where it promotes cell survival by initiating transcription of genes encoding stress-response enzymes, cell-adhesion molecules, pro-inflammatory cytokines and anti-apoptotic proteins. NF-κB and associated regulatory factors (IκB kinase subunits and bcl-3) are implicated in hematological and solid tumour malignancies. NF-κB appears to be involved in cell proliferation control, apoptosis control, angiogenesis promotion and possibly regulation of diffusion of metastases. There are several reports that inhibition of NF-κB as a therapeutic target may have a role in tumour cell death or growth inhibition. AREA COVERED IN THIS REVIEW We review data about inhibition of NF-κB in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). We describe the molecular mechanisms underlying NF-κB deregulation in these haematological malignancies. WHAT THE READER WILL GAIN Constitutive activation of NF-κB in the nucleus has been reported in some varieties of MDS/AML. The in vitro and in vivo results of NF-κB inhibition in myeloid malignancies are highlighted. TAKE HOME MESSAGE NF-κB selective inhibitory drugs may be useful, either as single agents or associated with conventional chemotherapy.
Collapse
Affiliation(s)
- Massimo Breccia
- Sapienza University, Department of Human Biotechnologies and Hematology, Rome, Italy.
| | | |
Collapse
|
19
|
Ritchie DS, Quach H, Fielding K, Neeson P. Drug-mediated and cellular immunotherapy in multiple myeloma. Immunotherapy 2010; 2:243-55. [PMID: 20635931 DOI: 10.2217/imt.10.9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Multiple myeloma is an immunologically relevant disease, which subverts and suppresses immunity, but that may also be amenable to immunological control. Novel drug and cell-based therapies provide an opportunity for the design of antimyeloma immunotherapy. Reversing the immunosuppression associated myeloma remains a substantial challenge. The minimal residual disease setting achieved by autologous stem cell transplant or highly efficacious induction therapy may reverse this immunoparesis and provide a setting for induction of antimyeloma T-cell responses. Adoptive cytotoxic T-lymphocyte/NK therapy and comprehensive treatment with immunomodulatory drug therapy represent means by which antimyeloma immune responses may be promoted. In addition, apoptosis-inducing therapies may prime endogenous antigen presentation via immunogenic cell death, which again may be enhanced by the addition of immunomodulatory drug therapy.
Collapse
Affiliation(s)
- David S Ritchie
- Department of Haematology & Medical Oncology, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia.
| | | | | | | |
Collapse
|
20
|
Vogelbacher R, Meister S, Guckel E, Starke C, Wittmann S, Stief A, Voll R, Daniel C, Hugo C. Bortezomib and sirolimus inhibit the chronic active antibody-mediated rejection in experimental renal transplantation in the rat. Nephrol Dial Transplant 2010; 25:3764-73. [DOI: 10.1093/ndt/gfq230] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
21
|
Koreth J, Alyea EP, Murphy WJ, Welniak LA. Proteasome inhibition and allogeneic hematopoietic stem cell transplantation: a review. Biol Blood Marrow Transplant 2010; 15:1502-12. [PMID: 19896073 DOI: 10.1016/j.bbmt.2009.07.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 07/16/2009] [Indexed: 11/17/2022]
Abstract
The proteasome and its associated ubiquitin protein modification system have proved to be an important therapeutic target in the treatment of multiple myeloma and other cancers. In addition to direct antitumor effects, proteasome inhibition also exerts strong effects on nonneoplastic immune cells. This indicates that proteasome inhibition, through the use of agents like bortezomib, could be used therapeutically to modulate immune responses. In this review we explore the emerging data, both preclinical and clinical, highlighting the importance of proteasome targeting of immunologic responses, primarily in the context of allogeneic hematopoietic stem cell transplantation (HSCT), both for the control of transplant-related toxicities like acute and chronic graft-versus-host disease (aGVHD, cGHVHD), and for improved malignant disease control after allogeneic HSCT.
Collapse
Affiliation(s)
- John Koreth
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachustts, USA
| | | | | | | |
Collapse
|
22
|
Van Herck JL, De Meyer GRY, Martinet W, Bult H, Vrints CJ, Herman AG. Proteasome inhibitor bortezomib promotes a rupture-prone plaque phenotype in ApoE-deficient mice. Basic Res Cardiol 2009; 105:39-50. [PMID: 19693627 DOI: 10.1007/s00395-009-0054-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 07/08/2009] [Accepted: 07/22/2009] [Indexed: 11/27/2022]
Abstract
The ubiquitin-proteasome system is involved in the development and progression of atherosclerosis. The aim of this study was to investigate whether plaque composition is affected by proteasome function. In vitro, the potent and selective proteasome inhibitor bortezomib induced apoptosis in both cultured smooth muscle cells (SMCs) and activated macrophages. This effect was associated with increased expression of C/EBP homologous protein and cleavage of caspase-12, indicative of endoplasmic reticulum stress. The sensitivity to the proapoptotic effects of proteasome inhibition correlated with the protein synthesis rate. Proteasome inhibition in explanted atherosclerotic plaques of ApoE-deficient mice resulted in a significant decrease in SMCs and macrophages, indicating that both cell types in the atherosclerotic plaque were susceptible to the proapoptotic effects of proteasome inhibition. In vivo proteasome inhibition in ApoE-deficient mice did not affect plaque size or composition of early atherosclerotic plaques, but resulted in a significant decrease in collagen content as well as a significant enlargement of the necrotic core in advanced atherosclerotic plaques. In conclusion, our results indicate that an impaired proteasome function promotes features of a more rupture-prone plaque phenotype.
Collapse
Affiliation(s)
- Jozef Leo Van Herck
- Division of Cardiology, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium.
| | | | | | | | | | | |
Collapse
|
23
|
Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome. Blood 2009; 114:3439-47. [PMID: 19671918 DOI: 10.1182/blood-2009-05-223677] [Citation(s) in RCA: 263] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Carfilzomib is a proteasome inhibitor in clinical development that primarily targets the chymotrypsin-like (CT-L) subunits in both the constitutive proteasome (c20S) and the immunoproteasome (i20S). To investigate the impact of inhibiting the CT-L activity with carfilzomib, we set out to quantitate the levels of CT-L subunits beta5 from the c20S and LMP7 from the i20S in normal and malignant hematopoietic cells. We found that the i20S is a major form of the proteasome expressed in cells of hematopoietic origin, including multiple myeloma (MM) CD138+ tumor cells. Although specific inhibition of either LMP7 or beta5 alone was insufficient to produce an antitumor response, inhibition of all proteasome subunits was cytotoxic to both hematologic tumor cells and peripheral blood mononuclear cells. However, selective inhibition of both beta5 and LMP7 was sufficient to induce an antitumor effect in MM, non-Hodgkin lymphoma, and leukemia cells while minimizing the toxicity toward nontransformed cells. In MM tumor cells, CT-L inhibition alone was sufficient to induce proapoptotic sequelae, including proteasome substrate accumulation, Noxa and caspase 3/7 induction, and phospho-eIF2alpha suppression. These data support a hypothesis that hematologic tumor cells are uniquely sensitive to CT-L inhibition and provide a mechanistic understanding of the clinical safety profile and antitumor activity of proteasome inhibitors.
Collapse
|