1
|
Dai Q, Han P, Qi X, Li F, Li M, Fan L, Zhang H, Zhang X, Yang X. 4-1BB Signaling Boosts the Anti-Tumor Activity of CD28-Incorporated 2 nd Generation Chimeric Antigen Receptor-Modified T Cells. Front Immunol 2020; 11:539654. [PMID: 33281809 PMCID: PMC7691374 DOI: 10.3389/fimmu.2020.539654] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
While chimeric antigen receptor-modified T (CAR-T) cells have shown great success for the treatment of B cell leukemia, their efficacy appears to be compromised in B cell derived lymphoma and solid tumors. Optimization of the CAR design to improve persistence and cytotoxicity is a focus of the current CAR-T study. Herein, we established a novel CAR structure by adding a full length 4-1BB co-stimulatory receptor to a 28Z-based second generation CAR that targets CD20. Our data indicated that this new 2028Z-4-1BB CAR-T cell showed improved proliferation and cytotoxic ability. To further understand the mechanism of action, we found that constitutive 4-1BB sensing significantly reduced the apoptosis of CAR-T cells, enhanced proliferation, and increased NF-κB pathway activation. Consistent with the enhanced proliferation and cytotoxicity in vitro, this new structure of CAR-T cells exhibited robust persistence and anti-tumor activity in a mouse xenograft lymphoma model. This work provides evidence for a new strategy to optimize the function of CAR-T against lymphoma.
Collapse
Affiliation(s)
- Qiang Dai
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Han
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyue Qi
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Fanlin Li
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Min Li
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Lilv Fan
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Huihui Zhang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoqing Zhang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Xuanming Yang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
2
|
Shrestha P, Garrahy I, Rahimian S. Eosinophilic granulomatosis with polyangiitis (EGPA) on remission with a new neuropathy: a rare case of mycophenolate induced primary CNS lymphoproliferative disease. J Community Hosp Intern Med Perspect 2020; 10:600-603. [PMID: 33194139 PMCID: PMC7643737 DOI: 10.1080/20009666.2020.1811066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Introduction Mycophenolate Mofetil (MMF), although a widely used immunosuppressant; an increasing concern of MMF induced Primary Central Nervous System Lymphoma (PCNSL) are being reported. Timely diagnosis and management of MMF induced PCNSL can play a vital role in improved outcomes. Case Presentation Eighty-one-year-old female with history of Eosinophilic Granulomatosis with Polyangiitis (EGPA) presented with word finding difficulty, right-hand weakness and right foot clumsiness. EGPA had been stable with MMF for 6 years. Physical examination revealed weakened right-hand grip, positive right-sided dysdiadokokinesia and right foot drop. MRI-brain identified three enhancing solid lesions – in right parietal, left insular and left mid brain extending into the left thalamus. Brain biopsy revealed a focally dense lymphoid infiltrate with CD20 positive B cells, with large atypical cells resembling Hodgkin Reed-Sternberg cells. With concern for immunosuppression related PCNSL, MMF was stopped. Patient was treated with 8 weeks of rituximab therapy for its least toxic profile and concomitant benefit in EGPA. On a 2 month follow up MRI-brain, near total resolution of the intracranial lesion was observed. Patient still had some residual right lower extremity incoordination, however, strength and speech normalized with resolution of dysdiadokokinesia. Patient was advised to discontinue MMF indefinitely and remains on low dose prednisone daily. Conclusion MMF is an inhibitor of Inosine Monophosphate Dehydrogenase which prevents T- and B-cell proliferation. PCNSL is a potential complication of chronic immunosuppression with this medication. Discontinuation of the drug along with immunosuppressive therapies have been the effective therapeutic options till date.
Collapse
Affiliation(s)
- Pragya Shrestha
- Internal Medicine Department, Reading Hospital - Tower Health System, Reading, PA, USA.,Precision Population Science Lab, Department of Internal Medicine and Pediatrics, Mayo Clinic, Rochester, MN, USA
| | - Ian Garrahy
- Internal Medicine Department, Reading Hospital - Tower Health System, Reading, PA, USA
| | - Shoja Rahimian
- Internal Medicine Department, Reading Hospital - Tower Health System, Reading, PA, USA
| |
Collapse
|
3
|
Yu Y, Govindarajan R. Intravascular large B-cell lymphoma presenting as an isolated cauda equina-conus medullaris syndrome - A case report. J Spinal Cord Med 2020; 43:556-559. [PMID: 30346252 PMCID: PMC7480634 DOI: 10.1080/10790268.2018.1527083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Context: Intravascular large B-cell lymphoma (IVLBCL) is a rare form of Non-Hodgkin lymphoma. The central nervous system, skin and hematopoietic system are the most commonly affected sites. We report a case of IVLBCL presenting as a combined conus medullaris-cauda equina syndrome. Findings: A 55-year-old Caucasian male presented with signs and symptoms of a combined conus medullaris-cauda equina syndrome confirmed by MRI and needle EMG. PET scan revealed a hyperactive focus in the adrenal gland, and biopsy showed evidence of IVLBCL. Therapy with R-CHOP resulted in improvement of weakness. Conclusion: A high index of suspicion and consideration of IVLBCL in the differential for patients with these isolated spinal cord symptoms and CSF findings may help initiate early and suitable therapy which has been proven to be beneficial.
Collapse
Affiliation(s)
- Yang Yu
- Department of Neurology, University of Missouri School of Medicine, Columbia, Missouri, USA,Correspondence to: Yang Yu, Department of Neurology, University of Missouri School of Medicine, One Hospital Dr., CE507, Columbia, MO65212, USA; Ph: 573-882-8668.
| | - Raghav Govindarajan
- Department of Neurology, University of Missouri School of Medicine, Columbia, Missouri, USA
| |
Collapse
|
4
|
Antonopoulos I, Daoussis D, Lalioti ME, Markatseli TE, Drosos AA, Taraviras S, Andonopoulos AP, Liossis SNC. B cell depletion treatment decreases CD4+IL4+ and CD4+CD40L+ T cells in patients with systemic sclerosis. Rheumatol Int 2019; 39:1889-1898. [PMID: 31227855 DOI: 10.1007/s00296-019-04350-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/15/2019] [Indexed: 01/29/2023]
Abstract
Recent data suggests that rituximab may favorably affect skin fibrosis and lung function in patients with systemic sclerosis. Based on experimental data suggesting a key role of B and T cells in scleroderma we aimed to explore the effect(s) of rituximab treatment on T cell subpopulations. Fifteen patients with scleroderma who received rituximab treatment and six who received standard treatment alone were recruited. Peripheral CD4+IL4+, CD4+INFγ+, CD4+IL17+ and CD4+CD40L+ T cells were assessed using flow cytometry. Using ELISA, serum levels of IL4 were assessed. Skin CD4+IL4+ T cells were assessed with confocal microscopy from skin biopsies. Following rituximab treatment skin CD4+IL4+ T cells obviously decreased as seen with confocal microscopy. Moreover, peripheral CD4+IL4+ T cells decreased significantly compared to those from patients who received standard treatment alone: median (IQR): 14.9 (22.63-12.88) vs 7.87 (12.81-4.9)%, p = 0.005 and 9.43 (19.53-7.50)% vs 14.86 (21.96-6.75)%, p = NS at baseline and 6 months later respectively, whereas there was no difference in serum IL4 levels. Peripheral CD4+CD40L+ T cells also decreased significantly following rituximab treatment compared to those from patients who received standard treatment alone: median (IQR): 17.78 (25.64-14.44)% vs 8.15 (22.85-3.08)%, p = 0.04 and 22.13 (58.77-8.20)% vs 72.11 (73.05-20.45)%, p = NS at baseline and 6 months later respectively. Furthermore, peripheral CD4+INFγ+ and CD4+IL17+ T cells revealed no differences following rituximab treatment. Our study demonstrates a link between rituximab treatment and CD4+IL4+ T cell decrease both in the skin and peripheral blood of patients with SSc.
Collapse
Affiliation(s)
- Ioannis Antonopoulos
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, University of Patras Medical School, Rion, 26504, Patras, Greece
| | - Dimitrios Daoussis
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, University of Patras Medical School, Rion, 26504, Patras, Greece
| | - Maria-Eleni Lalioti
- Department of Physiology, School of Medicine, University of Patras, Rion, 26504, Patras, Greece
| | - Theodora E Markatseli
- Department of Rheumatology, Ioannina University Hospital, University of Ioannina Medical School, Ioannina, Greece
| | - Alexandros A Drosos
- Department of Rheumatology, Ioannina University Hospital, University of Ioannina Medical School, Ioannina, Greece
| | - Stavros Taraviras
- Department of Physiology, School of Medicine, University of Patras, Rion, 26504, Patras, Greece
| | - Andrew P Andonopoulos
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, University of Patras Medical School, Rion, 26504, Patras, Greece
| | - Stamatis-Nick C Liossis
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, University of Patras Medical School, Rion, 26504, Patras, Greece.
| |
Collapse
|
5
|
Buatois V, Johnson Z, Salgado-Pires S, Papaioannou A, Hatterer E, Chauchet X, Richard F, Barba L, Daubeuf B, Cons L, Broyer L, D'Asaro M, Matthes T, LeGallou S, Fest T, Tarte K, Clarke Hinojosa RK, Genescà Ferrer E, Ribera JM, Dey A, Bailey K, Fielding AK, Eissenberg L, Ritchey J, Rettig M, DiPersio JF, Kosco-Vilbois MH, Masternak K, Fischer N, Shang L, Ferlin WG. Preclinical Development of a Bispecific Antibody that Safely and Effectively Targets CD19 and CD47 for the Treatment of B-Cell Lymphoma and Leukemia. Mol Cancer Ther 2018; 17:1739-1751. [PMID: 29743205 DOI: 10.1158/1535-7163.mct-17-1095] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/15/2018] [Accepted: 05/04/2018] [Indexed: 12/14/2022]
Abstract
CD47, an ubiquitously expressed innate immune checkpoint receptor that serves as a universal "don't eat me" signal of phagocytosis, is often upregulated by hematologic and solid cancers to evade immune surveillance. Development of CD47-targeted modalities is hindered by the ubiquitous expression of the target, often leading to rapid drug elimination and hemotoxicity including anemia. To overcome such liabilities, we have developed a fully human bispecific antibody, NI-1701, designed to coengage CD47 and CD19 selectively on B cells. NI-1701 demonstrates favorable elimination kinetics with no deleterious effects seen on hematologic parameters following single or multiple administrations to nonhuman primates. Potent in vitro and in vivo activity is induced by NI-1701 to kill cancer cells across a plethora of B-cell malignancies and control tumor growth in xenograft mouse models. The mechanism affording maximal tumor growth inhibition by NI-1701 is dependent on the coengagement of CD47/CD19 on B cells inducing potent antibody-dependent cellular phagocytosis of the targeted cells. NI-1701-induced control of tumor growth in immunodeficient NOD/SCID mice was more effective than that achieved with the anti-CD20 targeted antibody, rituximab. Interestingly, a synergistic effect was seen when tumor-implanted mice were coadministered NI-1701 and rituximab leading to significantly improved tumor growth inhibition and regression in some animals. We describe herein, a novel bispecific antibody approach aimed at sensitizing B cells to become more readily phagocytosed and eliminated thus offering an alternative or adjunct therapeutic option to patients with B-cell malignancies refractory/resistant to anti-CD20-targeted therapy. Mol Cancer Ther; 17(8); 1739-51. ©2018 AACR.
Collapse
Affiliation(s)
| | - Zoë Johnson
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | | | | | | | | | | | | | - Laura Cons
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | | | - Thomas Matthes
- Medical Faculty, University of Geneva, Genève, Switzerland
| | | | - Thierry Fest
- Rennes 1 University, Inserm U1236, Rennes, France
| | - Karin Tarte
- Rennes 1 University, Inserm U1236, Rennes, France
| | - Robert K Clarke Hinojosa
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Germans Trias Pujol, Badalona, Barcelona, Spain
| | - Eulàlia Genescà Ferrer
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Germans Trias Pujol, Badalona, Barcelona, Spain
| | - José María Ribera
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Germans Trias Pujol, Badalona, Barcelona, Spain
| | - Aditi Dey
- Paul O'Gorman Building, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Katharine Bailey
- Paul O'Gorman Building, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Adele K Fielding
- Paul O'Gorman Building, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Linda Eissenberg
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Julie Ritchey
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Rettig
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | | | | | | | - Limin Shang
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | |
Collapse
|
6
|
Wang J, Zhou M, Xu JY, Chen B, Ouyang J. Combination of BCL-2 and MYC protein expression improves high-risk stratification in diffuse large B-cell lymphoma. Onco Targets Ther 2015; 8:2645-50. [PMID: 26425100 PMCID: PMC4583112 DOI: 10.2147/ott.s86093] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose To evaluate whether the addition of two biological markers (MYC and BCL-2 protein overexpression) improves the stratification of high-risk patients with diffuse large B-cell lymphoma (DLBCL). Method Seven risk factors were identified at diagnosis, and a maximum of 7 points were assigned to each patient. The patients were classified according to four risk groups: low (0–1), low-intermediate (2–3), high-intermediate (4), and high (5–7). Only high-risk patients with DLBCL were included in this analysis. We retrospectively examined 20 cases from 2008 to 2013 at the Nanjing Drum Tower Hospital. Results The median expression of MYC protein was 60%, and 17 of 20 (65%) evaluable cases overexpressed MYC. The median expression of BCL-2 protein was also 60%. Eighteen of 20 (90%) evaluable cases showed BCL-2 overexpression. Additionally, 12 out of 20 cases (60%) demonstrated coexpression of MYC and BCL-2 proteins. The percentages of overall survival and progression-free survival at the median follow-up time (36 months) were 33.3%±16.1% and 16.9%±13.5%, respectively. By comparison, nine, four, and 20 patients were classified as high risk based on the International Prognostic Index (IPI), National Comprehensive Cancer Network(NCCN)-IPI, and revised IPI criteria, respectively. According to the IPI and NCCN-IPI stratification, the risk groups demonstrated closely overlapping survival curves. In addition, four out of 20 cases were identified as low-intermediate risk according to the NCCN-IPI criteria. Conclusion The addition of MYC and BCL-2 protein expression to the IPI could identify a subset of DLBCL patients with high-risk clinicopathological characteristics and poor clinical outcome.
Collapse
Affiliation(s)
- Jing Wang
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Min Zhou
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Jing-Yan Xu
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Bing Chen
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Jian Ouyang
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
7
|
Abstract
The immune system is designed to discriminate between self and tumor tissue. Through genetic recombination, there is fundamentally no limit to the number of tumor antigens that immune cells can recognize. Yet, tumors use a variety of immunosuppressive mechanisms to evade immunity. Insight into how the immune system interacts with tumors is expanding rapidly and has accelerated the translation of immunotherapies into medical breakthroughs. Herein, we appraise novel strategies that exploit the patient's immune system to kill cancer. We review various forms of immune-based therapies, which have shown significant promise in patients with hematologic malignancies, including (i) conventional monoclonal therapies like rituximab; (ii) engineered monoclonal antibodies called bispecific T-cell engagers; (iii) monoclonal antibodies and pharmaceutical drugs that block inhibitory T-cell pathways (i.e. PD-1, CTLA-4, and IDO); and (iv) adoptive cell transfer therapy with T cells engineered to express chimeric antigen receptors or T-cell receptors. We also assess the idea of using these therapies in combination and conclude by suggesting multi-prong approaches to improve treatment outcomes and curative responses in patients.
Collapse
Affiliation(s)
- Michelle H Nelson
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA; Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
8
|
Doumas S, Sakkas L, Panayiotidis P, Wozniak G, Vlychou M, Vassilopoulos G. Favorable outcome in non-Hodgkin lymphoma of the maxillary sinus treated with R-CHOP. Arch Med Sci 2014; 10:406-9. [PMID: 24904680 PMCID: PMC4042037 DOI: 10.5114/aoms.2013.34986] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/15/2012] [Accepted: 07/05/2012] [Indexed: 12/30/2022] Open
Affiliation(s)
- Stergios Doumas
- Maxillofacial Unit, Queen Victoria Hospital, East Grinstead, UK
| | - Leonidas Sakkas
- Department of Pathology, Papanikolaou General Hospital, Thessaloniki, Greece
| | | | | | - Marianna Vlychou
- Department of Radiology, University Hospital of Larissa, Larissa, Greece
| | | |
Collapse
|
9
|
Boland EL, Van Dyken CM, Duckett RM, McCluskey AJ, Poon GMK. Structural complementation of the catalytic domain of pseudomonas exotoxin A. J Mol Biol 2014; 426:645-55. [PMID: 24211469 PMCID: PMC3997303 DOI: 10.1016/j.jmb.2013.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 10/28/2013] [Accepted: 11/04/2013] [Indexed: 12/17/2022]
Abstract
The catalytic moiety of Pseudomonas exotoxin A (domain III or PE3) inhibits protein synthesis by ADP-ribosylation of eukaryotic elongation factor 2. PE3 is widely used as a cytocidal payload in receptor-targeted protein toxin conjugates. We have designed and characterized catalytically inactive fragments of PE3 that are capable of structural complementation. We dissected PE3 at an extended loop and fused each fragment to one subunit of a heterospecific coiled coil. In vitro ADP-ribosylation and protein translation assays demonstrate that the resulting fusions-supplied exogenously as genetic elements or purified protein fragments-had no significant catalytic activity or effect on protein synthesis individually but, in combination, catalyzed the ADP-ribosylation of eukaryotic elongation factor 2 and inhibited protein synthesis. Although complementing PE3 fragments are catalytically less efficient than intact PE3 in cell-free systems, co-expression in live cells transfected with transgenes encoding the toxin fusions inhibits protein synthesis and causes cell death comparably as intact PE3. Complementation of split PE3 offers a direct extension of the immunotoxin approach to generate bispecific agents that may be useful to target complex phenotypes.
Collapse
Affiliation(s)
- Erin L Boland
- Department of Pharmaceutical Sciences, Washington State University, Pullman, WA 99164, USA
| | - Crystal M Van Dyken
- Department of Pharmaceutical Sciences, Washington State University, Pullman, WA 99164, USA
| | - Rachel M Duckett
- Department of Pharmaceutical Sciences, Washington State University, Pullman, WA 99164, USA
| | - Andrew J McCluskey
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Gregory M K Poon
- Department of Pharmaceutical Sciences, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
10
|
Wang K, Jiang Y, Zheng W, Liu Z, Li H, Lou J, Gu M, Wang X. Silencing of human phosphatidylethanolamine-binding protein 4 enhances rituximab-induced death and chemosensitization in B-cell lymphoma. PLoS One 2013; 8:e56829. [PMID: 23451095 PMCID: PMC3581549 DOI: 10.1371/journal.pone.0056829] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/15/2013] [Indexed: 01/10/2023] Open
Abstract
Rituximab is the first line drug to treat non Hodgkin's lymphoma (B-NHL) alone or in combination with chemotherapy. However, 30-40% of B-NHL patients are unresponsive to rituximab or resistant after therapy. Human phosphatidylethanolamine-binding protein 4 (hPEBP4) is a novel member of PEBP family and functions as an anti-apoptotic molecule. In this study, we found hPEBP4 to be expressed in up to 90% of B-cell lymphoma patients, but in only 16.7% of normal lymph nodes. Interestingly, hPEBP4 overexpression inhibited rituximab-mediated complement dependent cytotoxicity (R-CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) in B-NHL cells while downregulation of hPEBP4 augmented the therapeutic efficacy of rituximab both in vitro and in vivo. Furthermore, hPEBP4 silencing sensitized the primary B-acute lymphocytic leukemia (B-ALL) cells to R-CDC. During rituximab-mediated complement dependent cytotoxicity, hPEBP4 was recruited to the cell membrane in a PE-binding domain dependent manner and inhibited R-CDC induced calcium flux and reactive oxygen species (ROS) generation. These events contributed to the decrease of cell death induced by R-CDC in B-cell lymphomas. Meanwhile, hPEBP4 knockdown potentiated the chemosensitization of the rituximab in B-cell lymphoma cells by regulating the expression of Bcl-xl, Cycline E, p21(waf/cip1) and p53 and the activation of caspase-3 and caspase-9. Considering that hPEBP4 conferred cellular resistance to rituximab treatment and was preferentially expressed in lymphoma tissue, it could be a potential valuable target for adjuvant therapy for B-cell lymphoma.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- Antibodies, Monoclonal, Murine-Derived/therapeutic use
- Apoptosis/drug effects
- Apoptosis/genetics
- Cell Line, Tumor
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Silencing
- Humans
- Immunohistochemistry
- In Vitro Techniques
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Confocal
- Phosphatidylethanolamine Binding Protein/genetics
- Phosphatidylethanolamine Binding Protein/metabolism
- Reactive Oxygen Species/metabolism
- Rituximab
Collapse
Affiliation(s)
- Kai Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yu Jiang
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Weiyan Zheng
- Department of Hematology, First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Zhiyong Liu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hui Li
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Jianzhou Lou
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Meidi Gu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaojian Wang
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- * E-mail:
| |
Collapse
|
11
|
Li J, Chen F, Cona MM, Feng Y, Himmelreich U, Oyen R, Verbruggen A, Ni Y. A review on various targeted anticancer therapies. Target Oncol 2012; 7:69-85. [PMID: 22350489 DOI: 10.1007/s11523-012-0212-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 01/30/2012] [Indexed: 12/11/2022]
Abstract
Translational oncology aims to translate laboratory research into new anticancer therapies. Contrary to conventional surgery, chemotherapy, and radiotherapy, targeted anticancer therapy (TAT) refers to systemic administration of drugs with particular mechanisms that specifically act on well-defined targets or biologic pathways that, when activated or inactivated, may cause regression or destruction of the malignant process, meanwhile with minimized adverse effects on healthy tissues. In this article, we intend to first give a brief review on various known TAT approaches that are deemed promising for clinical applications in the current trend of personalized medicine, and then we will introduce our newly developed approach namely small molecular sequential dual targeting theragnostic strategy as a generalized class of TAT for the management of most solid malignancies, which, after optimization, is expected to help improve overall cancer treatability and curability.
Collapse
Affiliation(s)
- Junjie Li
- Section of Radiology, Department of Diagnostic Sciences, Faculty of Medicine, University of Leuven, Herestraat 49, BE-3000, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Sinha A, Nast CC, Hristea I, Vo AA, Jordan SC. Resolution of clinical and pathologic features of C1q nephropathy after rituximab therapy. Clin Exp Nephrol 2010; 15:164-70. [PMID: 21107633 DOI: 10.1007/s10157-010-0377-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 10/26/2010] [Indexed: 01/22/2023]
Abstract
C1q nephropathy is a rare idiopathic glomerulopathy characterized by mesangial deposition of immunoglobulin and complement with C1q dominance or co-dominance, and the absence of clinical and laboratory evidence of systemic lupus erythematosus. Its clinical course is unpredictable and the response to corticosteroid or cytotoxic treatment is variable. Here, we report two cases of C1q nephropathy, one in a child and one in an adult, both presenting with impaired renal function and massive proteinuria. Both patients failed to respond to immunosuppressive medications; however, rituximab, an anti-CD20 antibody, was effective in preserving renal function in one patient and eliminating the need for hemodialysis in the other. In one patient, histologic regression of abnormalities was documented over 3 years post-treatment. Both patients have remained off other immunosuppressive medication for a prolonged period with stable renal function. These cases are, to our knowledge, the first reported successful treatment of C1q nephropathy with rituximab.
Collapse
Affiliation(s)
- Aditi Sinha
- Transplant Immunotherapy Program, Division of Nephrology, Cedars Sinai Medical Center, 8635 West 3rd Street, Suite 590 W, Los Angeles, CA 90048, USA
| | | | | | | | | |
Collapse
|