1
|
Yang S, Hu Q, Wang X, Qiao S, Qi C, Jin H, Zhong Y. Interferon Regulatory Factor 4: An Alternative Marker for Plasma Cells in Daratumumab-Treated Patients With Multiple Myeloma. Int J Lab Hematol 2025; 47:110-119. [PMID: 39267340 DOI: 10.1111/ijlh.14366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 09/17/2024]
Abstract
INTRODUCTION Anti-CD38 therapeutic modalities (e.g., daratumumab) can impede classical CD38 and CD138 gating use for plasma cell (PC) detection in multiple myeloma (MM) patients with minimal residual disease (MRD). We assessed the applicability of CD229, CD269, and interferon regulatory factor (IRF-4) for PC detection in MM MRD patients. METHODS Bone marrow samples were collected from patients with MM. Through multiparameter flow cytometry, we evaluated the suitability of CD229, CD269, and IRF-4 for distinguishing PCs from other hematopoietic cells and compared their expression pattern on normal PCs (nPCs) and aberrant PCs (aPCs). We also assessed IRF-4 expression stability after sample storage under different conditions. A 10-color MRD antibody panel was used to determine whether IRF-4 is an alternative primary PC-gating marker for MM MRD assessment. RESULTS IRF-4 was expressed specifically on all PCs; its mean fluorescence intensity (MFI) was highest on PCs among all hematopoietic cells. This MFI did not decrease even after sample storage at 4°C or 25°C for 72 h. In all 42 MRD assessment samples, except for samples (n = 10) with no PCs, the use of IRF-4 enabled accurate nPC (n = 12), aPC (n = 13), and nPC + aPC (n = 7) identification. Even samples from daratumumab-treated patients had high IRF-4 MFI, with no difference between pre-treatment and post-treatment (n = 7; p = 0.610). CONCLUSIONS IRF-4 demonstrates high MFI on PCs, and it is not expressed on other leukocytes. In MM patients with MRD, daratumumab treatment does not affect IRF-4 expression. IRF-4 is a promising marker for PC identification in MRD assessment of MM patients undergoing anti-CD38 therapy.
Collapse
Affiliation(s)
- Suwen Yang
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Qianwen Hu
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiaofen Wang
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sai Qiao
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chao Qi
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hong Jin
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yuhong Zhong
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Goto H, Shiraishi Y, Okada S. Continuing progress in radioimmunotherapy for hematologic malignancies. Blood Rev 2025; 69:101250. [PMID: 39609167 DOI: 10.1016/j.blre.2024.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024]
Abstract
Radioimmunotherapy (RIT) involves combining a cytotoxic radionuclide with an antibody (Ab) targeting a tumor antigen. Compared with conventional therapies, RIT improves the therapeutic efficacy of Ab and ameliorates toxicity. This comprehensive review describes the current advancements and future prospects in RIT for treating hematologic malignancies based on recent investigations. Although β-particle RITs targeting CD20 are effective with low toxicity in patients with relapsed/refractory indolent or transformed non-Hodgkin's lymphoma, these treatments have not gained popularity because of the increasing availability of new therapies. RIT using single-domain antibodies is expected to improve tumor penetrance and reduce radiation exposure to non-target organs. To enhance RIT efficacy, α-particle RIT and pretargeted radioimmunotherapy (PRIT) are currently being developed. Alpha-particle RIT demonstrates substantial antitumor activity and reduced bystander effects due to its high linear energy transfer and short particle range. PRIT may increase the tumor-to-whole body dose ratio.
Collapse
Affiliation(s)
- Hiroki Goto
- Division of Radioisotope and Tumor Pathobiology, Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| | - Yoshioki Shiraishi
- Radioisotope Center, Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| |
Collapse
|
3
|
Riccardi F, Tangredi C, Dal Bo M, Toffoli G. Targeted therapy for multiple myeloma: an overview on CD138-based strategies. Front Oncol 2024; 14:1370854. [PMID: 38655136 PMCID: PMC11035824 DOI: 10.3389/fonc.2024.1370854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Multiple myeloma (MM) is an incurable hematological disease characterized by the uncontrolled growth of plasma cells primarily in the bone marrow. Although its treatment consists of the administration of combined therapy regimens mainly based on immunomodulators and proteosome inhibitors, MM remains incurable, and most patients suffer from relapsed/refractory disease with poor prognosis and survival. The robust results achieved by immunotherapy targeting MM-associated antigens CD38 and CD319 (also known as SLAMF7) have drawn attention to the development of new immune-based strategies and different innovative compounds in the treatment of MM, including new monoclonal antibodies, antibody-drug conjugates, recombinant proteins, synthetic peptides, and adaptive cellular therapies. In this context, Syndecan1 (CD138 or SDC1), a transmembrane heparan sulfate proteoglycan that is upregulated in malignant plasma cells, has gained increasing attention in the panorama of MM target antigens, since its key role in MM tumorigenesis, progression and aggressiveness has been largely reported. Here, our aim is to provide an overview of the most important aspects of MM disease and to investigate the molecular functions of CD138 in physiologic and malignant cell states. In addition, we will shed light on the CD138-based therapeutic approaches currently being tested in preclinical and/or clinical phases in MM and discuss their properties, mechanisms of action and clinical applications.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Carmela Tangredi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
4
|
Caers J, Duray E, Vrancken L, Marcion G, Bocuzzi V, De Veirman K, Krasniqi A, Lejeune M, Withofs N, Devoogdt N, Dumoulin M, Karlström AE, D’Huyvetter M. Radiotheranostic Agents in Hematological Malignancies. Front Immunol 2022; 13:911080. [PMID: 35865548 PMCID: PMC9294596 DOI: 10.3389/fimmu.2022.911080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/06/2022] [Indexed: 12/23/2022] Open
Abstract
Radioimmunotherapy (RIT) is a cancer treatment that combines radiation therapy with tumor-directed monoclonal antibodies (Abs). Although RIT had been introduced for the treatment of CD20 positive non-Hodgkin lymphoma decades ago, it never found a broad clinical application. In recent years, researchers have developed theranostic agents based on Ab fragments or small Ab mimetics such as peptides, affibodies or single-chain Abs with improved tumor-targeting capacities. Theranostics combine diagnostic and therapeutic capabilities into a single pharmaceutical agent; this dual application can be easily achieved after conjugation to radionuclides. The past decade has seen a trend to increased specificity, fastened pharmacokinetics, and personalized medicine. In this review, we discuss the different strategies introduced for the noninvasive detection and treatment of hematological malignancies by radiopharmaceuticals. We also discuss the future applications of these radiotheranostic agents.
Collapse
Affiliation(s)
- Jo Caers
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
- *Correspondence: Jo Caers,
| | - Elodie Duray
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Louise Vrancken
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Guillaume Marcion
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Valentina Bocuzzi
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmet Krasniqi
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Margaux Lejeune
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Nadia Withofs
- Department of Nuclear Medicine, CHU de Liège, Liège, Belgium
| | - Nick Devoogdt
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Mireille Dumoulin
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Matthias D’Huyvetter
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
5
|
Yang Z, Chen S, Ying H, Yao W. Targeting syndecan-1: new opportunities in cancer therapy. Am J Physiol Cell Physiol 2022; 323:C29-C45. [PMID: 35584326 PMCID: PMC9236862 DOI: 10.1152/ajpcell.00024.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022]
Abstract
Syndecan-1 (SDC1, CD138) is one of the heparan sulfate proteoglycans and is essential for maintaining normal cell morphology, interacting with the extracellular and intracellular protein repertoire, as well as mediating signaling transduction upon environmental stimuli. The critical role of SDC1 in promoting tumorigenesis and metastasis has been increasingly recognized in various cancer types, implying a promising potential of utilizing SDC1 as a novel target for cancer therapy. This review summarizes the current knowledge on SDC1 structure and functions, including its role in tumor biology. We also discuss the highlights and limitations of current SDC1-targeted therapies as well as the obstacles in developing new therapeutic methods, offering our perspective on the future directions to target SDC1 for cancer treatment.
Collapse
Affiliation(s)
- Zecheng Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shuaitong Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wantong Yao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
6
|
Parakh S, Lee ST, Gan HK, Scott AM. Radiolabeled Antibodies for Cancer Imaging and Therapy. Cancers (Basel) 2022; 14:1454. [PMID: 35326605 PMCID: PMC8946248 DOI: 10.3390/cancers14061454] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/14/2022] [Accepted: 03/07/2022] [Indexed: 12/04/2022] Open
Abstract
Radioimmunoconjugates consist of a monoclonal antibody (mAb) linked to a radionuclide. Radioimmunoconjugates as theranostics tools have been in development with success, particularly in hematological malignancies, leading to approval by the US Food and Drug Administration (FDA) for the treatment of non-Hodgkin's lymphoma. Radioimmunotherapy (RIT) allows for reduced toxicity compared to conventional radiation therapy and enhances the efficacy of mAbs. In addition, using radiolabeled mAbs with imaging methods provides critical information on the pharmacokinetics and pharmacodynamics of therapeutic agents with direct relevance to the optimization of the dose and dosing schedule, real-time antigen quantitation, antigen heterogeneity, and dynamic antigen changes. All of these parameters are critical in predicting treatment responses and identifying patients who are most likely to benefit from treatment. Historically, RITs have been less effective in solid tumors; however, several strategies are being investigated to improve their therapeutic index, including targeting patients with minimal disease burden; using pre-targeting strategies, newer radionuclides, and improved labeling techniques; and using combined modalities and locoregional application. This review provides an overview of the radiolabeled intact antibodies currently in clinical use and those in development.
Collapse
Affiliation(s)
- Sagun Parakh
- Department of Medical Oncology, Heidelberg, VIC 3084, Australia; (S.P.); (H.K.G.)
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
| | - Sze Ting Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Hui K. Gan
- Department of Medical Oncology, Heidelberg, VIC 3084, Australia; (S.P.); (H.K.G.)
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC 3010, Australia
| | - Andrew M. Scott
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC 3010, Australia
| |
Collapse
|
7
|
Van Raemdonck K, Umar S, Palasiewicz K, Volin MV, Elshabrawy HA, Romay B, Tetali C, Ahmed A, Amin MA, Zomorrodi RK, Sweiss N, Shahrara S. Interleukin-34 Reprograms Glycolytic and Osteoclastic Rheumatoid Arthritis Macrophages via Syndecan 1 and Macrophage Colony-Stimulating Factor Receptor. Arthritis Rheumatol 2021; 73:2003-2014. [PMID: 33982895 PMCID: PMC8568622 DOI: 10.1002/art.41792] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 04/27/2021] [Indexed: 11/07/2022]
Abstract
OBJECTIVE In rheumatoid arthritis (RA), elevated serum interleukin-34 (IL-34) levels are linked with increased disease severity. IL-34 binds to 2 receptors, macrophage colony-stimulating factor receptor (M-CSFR) and syndecan 1, which are coexpressed in RA macrophages. Expression of both IL-34 and syndecan 1 is strikingly elevated in the RA synovium, yet their mechanisms of action remain undefined. This study was undertaken to investigate the mechanism of action of IL-34 in RA. METHODS To characterize the significance of IL-34 in immunometabolism, its mechanism of action was elucidated in joint macrophages, fibroblasts, and T effector cells using RA and preclinical models. RESULTS Intriguingly, syndecan 1 activated IL-34-induced M-CSFR phosphorylation and reprogrammed RA naive cells into distinctive CD14+CD86+GLUT1+ M34 macrophages that expressed elevated levels of IL-1β, CXCL8, and CCL2. In murine M34 macrophages, the inflammatory phenotype was accompanied by potentiated glycolytic activity, exhibited by transcriptional up-regulation of GLUT1, c-Myc, and hypoxia-inducible factor 1α (HIF-1α) and amplified pyruvate and l-lactate secretion. Local expression of IL-34 provoked arthritis by expanding the glycolytic F4/80-positive, inducible nitric oxide synthase (iNOS)-positive macrophage population, which in turn attracted fibroblasts and polarized Th1/Th17 cells. The cross-talk between murine M34 macrophages and Th1/Th17 cells broadened the inflammatory and metabolic phenotypes, resulting in the expansion of IL-34 pathogenicity. Consequently, IL-34-instigated joint inflammation was alleviated in RAG-/- mice compared to wild-type mice. Syndecan 1 deficiency attenuated IL-34-induced arthritis by interfering with joint glycolytic M34 macrophage and osteoclast remodeling. Similarly, inhibition of glycolysis by 2-deoxy-d-glucose reversed the joint swelling and metabolic rewiring triggered by IL-34 via HIF-1α and c-Myc induction. CONCLUSION IL-34 is a novel endogenous factor that remodels hypermetabolic M34 macrophages and facilitates their cross-regulation with T effector cells to advance inflammatory bone destruction in RA.
Collapse
Affiliation(s)
- Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL 60612, USA
| | - Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL 60612, USA
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL 60612, USA
| | - Michael V. Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA
| | - Hatem A. Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Bianca Romay
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL 60612, USA
| | - Chandana Tetali
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL 60612, USA
| | - Azam Ahmed
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL 60612, USA
| | - M. Asif Amin
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ryan K. Zomorrodi
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL 60612, USA
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL 60612, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL 60612, USA
| |
Collapse
|
8
|
Kiraga Ł, Kucharzewska P, Paisey S, Cheda Ł, Domańska A, Rogulski Z, Rygiel TP, Boffi A, Król M. Nuclear imaging for immune cell tracking in vivo – Comparison of various cell labeling methods and their application. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
9
|
Targeted-Alpha-Therapy Combining Astatine-211 and anti-CD138 Antibody in A Preclinical Syngeneic Mouse Model of Multiple Myeloma Minimal Residual Disease. Cancers (Basel) 2020; 12:cancers12092721. [PMID: 32971984 PMCID: PMC7564412 DOI: 10.3390/cancers12092721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Multiple myeloma is a cancer that remains incurable. Among the many therapies under evaluation, antibodies can be used as vehicles to target and deliver toxic radiation to the tumour cells. Our objective was therefore to investigate the potential of targeted alpha therapy, combining an anti-CD138 mAb with astatine-211, to destroy the residual cells responsible for relapse. We have shown in a mouse model that mimics human disease, that destroying multiple myeloma cells is feasible with low toxicity by injecting an anti-CD138 mAb coupled with astatine-211. This approach could eradicate residual cells after initial treatment and thus prevent recurrence. Abstract Despite therapeutic progress in recent years with the introduction of targeted therapies (daratumumab, elotuzumab), multiple myeloma remains an incurable cancer. The question is therefore to investigate the potential of targeted alpha therapy, combining an anti-CD138 antibody with astatine-211, to destroy the residual cells that cause relapses. A preclinical syngeneic mouse model, consisting of IV injection of 1 million of 5T33 cells in a KaLwRij C57/BL6 mouse, was treated 10 days later with an anti-mCD138 antibody, called 9E7.4, radiolabeled with astatine-211. Four activities of the 211At-9E7.4 radioimmunoconjugate were tested in two independent experiments: 370 kBq (n = 16), 555 kBq (n = 10), 740 kBq (n = 17) and 1100 kBq (n = 6). An isotype control was also tested at 555 kBq (n = 10). Biodistribution, survival rate, hematological parameters, enzymatic hepatic toxicity, histological examination and organ dosimetry were considered. The survival median of untreated mice was 45 days after engraftment. While the activity of 1100 kBq was highly toxic, the activity of 740 kBq offered the best efficacy with 65% of overall survival 150 days after the treatment with no evident sign of toxicity. This work demonstrates the pertinence of treating minimal residual disease of multiple myeloma with an anti-CD138 antibody coupled to astatine-211.
Collapse
|
10
|
Bruins WSC, Zweegman S, Mutis T, van de Donk NWCJ. Targeted Therapy With Immunoconjugates for Multiple Myeloma. Front Immunol 2020; 11:1155. [PMID: 32636838 PMCID: PMC7316960 DOI: 10.3389/fimmu.2020.01155] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
The introduction of proteasome inhibitors (PI) and immunomodulatory drugs (IMiD) has markedly increased the survival of multiple myeloma (MM) patients. Also, the unconjugated monoclonal antibodies (mAb) daratumumab (anti-CD38) and elotuzumab (anti-SLAMF7) have revolutionized MM treatment given their clinical efficacy and safety, illustrating the potential of targeted immunotherapy as a powerful treatment strategy for MM. Nonetheless, most patients eventually develop PI-, IMiD-, and mAb-refractory disease because of the selection of resistant MM clones, which associates with a poor prognosis. Accordingly, these patients remain in urgent need of new therapies with novel mechanisms of action. In this respect, mAbs or mAb fragments can also be utilized as carriers of potent effector moieties to specifically target surface antigens on cells of interest. Such immunoconjugates have the potential to exert anti-MM activity in heavily pretreated patients due to their distinct and pleiotropic mechanisms of action. In addition, the fusion of highly cytotoxic compounds to mAbs decreases the off-target toxicity, thereby improving the therapeutic window. According to the effector moiety, immunoconjugates are classified into antibody-drug conjugates, immunotoxins, immunocytokines, or radioimmunoconjugates. This review will focus on the mechanisms of action, safety and efficacy of several promising immunoconjugates that are under investigation in preclinical and/or clinical MM studies. We will also include a discussion on combination therapy with immunoconjugates, resistance mechanisms, and future developments.
Collapse
Affiliation(s)
- Wassilis S C Bruins
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sonja Zweegman
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Tuna Mutis
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
11
|
Radioimmunotherapy in Combination with Reduced-Intensity Conditioning for Allogeneic Hematopoietic Cell Transplantation in Patients with Advanced Multiple Myeloma. Biol Blood Marrow Transplant 2019; 26:691-697. [PMID: 31730919 DOI: 10.1016/j.bbmt.2019.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 12/19/2022]
Abstract
Radioimmunotherapy (RIT) has the potential to reduce the incidence of relapse after allogeneic hematopoietic cell transplantation (allo-HCT) in patients with advanced-stage multiple myeloma (MM). In this study, we evaluated the efficacy of RIT in combination with chemotherapy-based reduced-intensity conditioning (RIC). RIT was based on the coupling of an anti-CD66 antibody to the beta emitter 188-rhenium (188-re) for targeted bone marrow irradiation. Between 2012 and 2018, 30 patients with MM, most of them heavily pretreated with various therapies including proteasome inhibitors, immunomodulatory drugs, anti-CD38 antibodies, and autologous hematopoietic cell transplantation (auto-HCT), were treated with a RIT-RIC combination before allo-HCT. In addition to a fludarabine plus melphalan- or treosulfan-based RIC, a median dose of 18.1 Gy (interquartile range [IQR], 14.6 to 24.1 Gy) was applied to the bone marrow. After a median duration of follow-up for surviving patients of 2.1 years (IQR, 1.3 to 3.0 years), the 2-year progression-free survival and overall survival rates were 43% (95% confidence interval [CI], 26% to 73%) and 55% (95% CI, 38% to 79%), respectively. The 2-year nonrelapse mortality and cumulative incidence of progression were 17% (95% CI, 3% to 30%) and 46% (95% CI, 25% to 67%), respectively. Renal toxicity and mucositis were the most frequent extramedullary side effects. In conclusion, the addition of RIT to RIC was safe and feasible and resulted in promising outcomes compared with those previously reported for RIC-based allo-HCT without the addition of RIT in patients with relapsed/refractory MM. Nevertheless, despite the addition of RIT, relapse after allo-HCT remained a major determinant of therapeutic failure. Therefore, the development of novel RIT strategies (eg, dual targeting strategies or combinations with adapter chimeric antigen receptor T cell-based therapies) is needed.
Collapse
|
12
|
Role of cell surface proteoglycans in cancer immunotherapy. Semin Cancer Biol 2019; 62:48-67. [PMID: 31336150 DOI: 10.1016/j.semcancer.2019.07.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 12/23/2022]
Abstract
Over the past few decades, understanding how tumor cells evade the immune system and their communication with their tumor microenvironment, has been the subject of intense investigation, with the aim of developing new cancer immunotherapies. The current therapies against cancer such as monoclonal antibodies against checkpoint inhibitors, adoptive T-cell transfer, cytokines, vaccines, and oncolytic viruses have managed to improve the clinical outcome of the patients. However, in some tumor entities, the response is limited and could benefit from the identification of novel therapeutic targets. It is known that tumor-extracellular matrix interplay and matrix remodeling are necessary for anti-tumor and pro-tumoral immune responses. Proteoglycans are dominant components of the extracellular matrix and are a highly heterogeneous group of proteins characterized by the covalent attachment of a specific linear carbohydrate chain of the glycosaminoglycan type. At cell surfaces, these molecules modulate the expression and activity of cytokines, chemokines, growth factors, adhesion molecules, and function as signaling co-receptors. By these mechanisms, proteoglycans influence the behavior of cancer cells and their microenvironment during the progression of solid tumors and hematopoietic malignancies. In this review, we discuss why cell surface proteoglycans are attractive pharmacological targets in cancer, and we present current and recent developments in cancer immunology and immunotherapy utilizing proteoglycan-targeted strategies.
Collapse
|
13
|
Vitale D, Kumar Katakam S, Greve B, Jang B, Oh ES, Alaniz L, Götte M. Proteoglycans and glycosaminoglycans as regulators of cancer stem cell function and therapeutic resistance. FEBS J 2019; 286:2870-2882. [PMID: 31230410 DOI: 10.1111/febs.14967] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/29/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022]
Abstract
In contrast to the bulk of the tumor, a subset of cancer cells called cancer stem cells (CSC; or tumor-initiating cells) is characterized by self-renewal, unlimited proliferative potential, expression of multidrug resistance proteins, active DNA repair capacity, apoptosis resistance, and a considerable developmental plasticity. Due to these properties, CSCs display increased resistance to chemo- and radiotherapy. Recent findings indicate that aberrant functions of proteoglycans (PGs) and glycosaminoglycans (GAGs) contribute substantially to the CSC phenotype and therapeutic resistance. In this review, we summarize how the diverse functions of the glycoproteins and carbohydrates facilitate acquisition and maintenance of the CSC phenotype, and how this knowledge can be exploited to develop novel anticancer therapies. For example, the large transmembrane chondroitin sulfate PG NG2/CSPG4 marks stem cell (SC) populations in brain tumors. Cell surface heparan sulfate PGs of the syndecan and glypican families modulate the stemness-associated Wnt, hedgehog, and notch signaling pathways, whereas the interplay of hyaluronan in the SC niche with CSC CD44 determines the maintenance of stemness and promotes therapeutic resistance. A better understanding of the molecular mechanisms by which PGs and GAGs regulate CSC function will aid the development of targeted therapeutic approaches which could avoid relapse after an otherwise successful conventional therapy. Chimeric antigen receptor T cells, PG-primed dendritic cells, PG-targeted antibody-drug conjugates, and inhibitory peptides and glycans have already shown highly promising results in preclinical models.
Collapse
Affiliation(s)
- Daiana Vitale
- Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín, Argentina
| | | | - Burkhard Greve
- Department of Radiotherapy - Radiooncology, Münster University Hospital, Germany
| | - Bohee Jang
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Eok-Soo Oh
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Laura Alaniz
- Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín, Argentina
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Germany
| |
Collapse
|
14
|
What is the Best Radionuclide for Immuno-PET of Multiple Myeloma? A Comparison Study Between 89Zr- and 64Cu-Labeled Anti-CD138 in a Preclinical Syngeneic Model. Int J Mol Sci 2019; 20:ijms20102564. [PMID: 31137758 PMCID: PMC6567828 DOI: 10.3390/ijms20102564] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/16/2022] Open
Abstract
Although positron emission tomography (PET) imaging with 18-Fluorodeoxyglucose (18F-FDG) is a promising technique in multiple myeloma (MM), the development of other radiopharmaceuticals seems relevant. CD138 is currently used as a standard marker for the identification of myeloma cells and could be used in phenotype tumor imaging. In this study, we used an anti-CD138 murine antibody (9E7.4) radiolabeled with copper-64 (64Cu) or zirconium-89 (89Zr) and compared them in a syngeneic mouse model to select the optimal tracers for MM PET imaging. Then, 9E7.4 was conjugated to TE2A-benzyl isothiocyanate (TE2A) and desferrioxamine (DFO) chelators for 64Cu and 89Zr labeling, respectively. 64Cu-TE2A-9E7.4 and 89Zr-DFO-9E7.4 antibodies were evaluated by PET imaging and biodistribution studies in C57BL/KaLwRij mice bearing either 5T33-MM subcutaneous tumors or bone lesions and were compared to 18F-FDG-PET imaging. In biodistribution and PET studies, 64Cu-TE2A-9E7.4 and 89Zr-DFO-9E7.4 displayed comparable good tumor uptake of subcutaneous tumors. On the bone lesions, PET imaging with 64Cu-TE2A-9E7.4 and 89Zr-DFO-9E7.4 showed higher uptake than with 18F-FDG-PET. Comparison of both 9E7.4 conjugates revealed higher nonspecific bone uptakes of 89Zr-DFO-9E7.4 than 64Cu-TE2A-9E7.4. Because of free 89Zr’s tropism for bone when using 89Zr-anti-CD138, 64Cu-anti-CD138 antibody had the most optimal tumor-to-nontarget tissue ratios for translation into humans as a specific new imaging radiopharmaceutical agent in MM.
Collapse
|
15
|
Immunotherapeutics in Multiple Myeloma: How Can Translational Mouse Models Help? JOURNAL OF ONCOLOGY 2019; 2019:2186494. [PMID: 31093282 PMCID: PMC6481018 DOI: 10.1155/2019/2186494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/04/2019] [Indexed: 12/30/2022]
Abstract
Multiple myeloma (MM) is usually diagnosed in older adults at the time of immunosenescence, a collection of age-related changes in the immune system that contribute to increased susceptibility to infection and cancer. The MM tumor microenvironment and cumulative chemotherapies also add to defects in immunity over the course of disease. In this review we discuss how mouse models have furthered our understanding of the immune defects caused by MM and enabled immunotherapeutics to progress to clinical trials, but also question the validity of using immunodeficient models for these purposes. Immunocompetent models, in particular the 5T series and Vk⁎MYC models, are increasingly being utilized in preclinical studies and are adding to our knowledge of not only the adaptive immune system but also how the innate system might be enhanced in anti-MM activity. Finally we discuss the concept of immune profiling to target patients who might benefit the most from immunotherapeutics, and the use of humanized mice and 3D culture systems for personalized medicine.
Collapse
|
16
|
Roshal M. Minimal Residual Disease Detection by Flow Cytometry in Multiple Myeloma: Why and How? Semin Hematol 2018; 55:4-12. [PMID: 29759152 DOI: 10.1053/j.seminhematol.2018.02.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 11/11/2022]
Abstract
The outlook for myeloma patients has steadily improved with the introduction of newer drug combinations in recent years. Unlike older therapies that largely achieved only modest levels of neoplastic clone reduction, the newer drug combinations have led to deeper suppression of myeloma clones in most patients. Frequently the neoplastic clones become undetectable with traditional disease evaluation approaches. Recent studies using ultrasensitive disease monitoring have demonstrated that patients with disease undetectable by traditional techniques show wide heterogeneity in disease levels varying by several orders of magnitude. Moreover, measurement of the depth of disease suppression even at very low level has emerged as the most powerful prognostication tool in myeloma. Minimal (or measurable) residual disease (MRD) evaluation has also been proposed as a relevant tool in assessment of drug efficacy and in selection of further therapeutic options. In the face of the robust MRD measurement utility data, it has become critical to develop widely applicable disease monitoring techniques that can be applied to more patients in a variety of clinical setting. Both DNA-based and flow cytometry-based approaches have been successfully developed for this purpose achieving sensitivity approaching 1 neoplastic cell in a million. This review article focuses on the theoretical and practical aspects and challenges of deep MRD monitoring in myeloma by flow cytometry. Challenges of flow cytometric disease monitoring in the era of antigen-directed therapy are also discussed.
Collapse
Affiliation(s)
- Mikhail Roshal
- Hematopathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
17
|
Green DJ, O'Steen S, Lin Y, Comstock ML, Kenoyer AL, Hamlin DK, Wilbur DS, Fisher DR, Nartea M, Hylarides MD, Gopal AK, Gooley TA, Orozco JJ, Till BG, Orcutt KD, Wittrup KD, Press OW. CD38-bispecific antibody pretargeted radioimmunotherapy for multiple myeloma and other B-cell malignancies. Blood 2018; 131:611-620. [PMID: 29158362 PMCID: PMC5805491 DOI: 10.1182/blood-2017-09-807610] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022] Open
Abstract
Pretargeted radioimmunotherapy (PRIT) has demonstrated remarkable efficacy targeting tumor antigens, but immunogenicity and endogenous biotin blocking may limit clinical translation. We describe a new PRIT approach for the treatment of multiple myeloma (MM) and other B-cell malignancies, for which we developed an anti-CD38-bispecific fusion protein that eliminates endogenous biotin interference and immunogenic elements. In murine xenograft models of MM and non-Hodgkin lymphoma (NHL), the CD38-bispecific construct demonstrated excellent blood clearance and tumor targeting. Dosimetry calculations showed a tumor-absorbed dose of 43.8 Gy per millicurie injected dose of 90Y, with tumor-to-normal organ dose ratios of 7:1 for liver and 15:1 for lung and kidney. In therapy studies, CD38-bispecific PRIT resulted in 100% complete remissions by day 12 in MM and NHL xenograft models, ultimately curing 80% of mice at optimal doses. In direct comparisons, efficacy of the CD38 bispecific proved equal or superior to streptavidin (SA)-biotin-based CD38-SA PRIT. Each approach cured at least 75% of mice at the highest radiation dose tested (1200 µCi), whereas at 600- and 1000-µCi doses, the bispecific outperformed the SA approach, curing 35% more mice overall (P < .004). The high efficacy of bispecific PRIT, combined with its reduced risk of immunogenicity and endogenous biotin interference, make the CD38 bispecific an attractive candidate for clinical translation. Critically, CD38 PRIT may benefit patients with unresponsive, high-risk disease because refractory disease typically retains radiation sensitivity. We posit that PRIT might not only prolong survival, but possibly cure MM and treatment-refractory NHL patients.
Collapse
Affiliation(s)
- Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Shyril O'Steen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Yukang Lin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Melissa L Comstock
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Aimee L Kenoyer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Donald K Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, WA
| | - D Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, WA
| | | | - Margaret Nartea
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Mark D Hylarides
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Ajay K Gopal
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Theodore A Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Johnnie J Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Kelly D Orcutt
- Department of Chemical Engineering and
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - K Dane Wittrup
- Department of Chemical Engineering and
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| |
Collapse
|
18
|
Bailly C, Gouard S, Lacombe M, Remaud-Le Saëc P, Chalopin B, Bourgeois M, Chouin N, Tripier R, Halime Z, Haddad F, Faivre-Chauvet A, Kraeber-Bodéré F, Chérel M, Bodet-Milin C. Comparison of Immuno-PET of CD138 and PET imaging with 64CuCl 2 and 18F-FDG in a preclinical syngeneic model of multiple myeloma. Oncotarget 2018; 9:9061-9072. [PMID: 29507674 PMCID: PMC5823645 DOI: 10.18632/oncotarget.23886] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 11/10/2017] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Although recent data from the literature suggest that PET imaging with [18]-Fluorodeoxyglucose (18F-FDG) is a promising technique in multiple myeloma (MM), the development of other radiopharmaceuticals seems relevant. CD138 is currently used as a standard marker in many laboratories for the identification and purification of myeloma cells, and could be used in phenotype tumor imaging. In this study, we evaluated a 64Cu-labeled anti-CD138 murine antibody (64Cu-TE2A-9E7.4) and a metabolic tracer (64CuCl2) for PET imaging in a MM syngeneic mouse model. EXPERIMENTAL DESIGN AND RESULTS 64Cu-TE2A-9E7.4 antibody and 64CuCl2 were evaluated via PET imaging and biodistribution studies in C57BL / KaLwRij mice bearing either 5T33-MM subcutaneous tumors or bone lesions. These results were compared to 18F-FDG-PET imaging. Autoradiography and histology of representative tumors were secondly conducted. In biodistribution and PET studies, 64Cu-TE2A-9E7.4 displayed good tumor uptake of subcutaneous and intra-medullary lesions, greater than that demonstrated with 18F-FDG-PET. In control experiments, only low-level, non-specific uptake of 64Cu-labeled isotype IgG was observed in tumors. Similarly, low activity concentrations of 64CuCl2 were accumulated in MM lesions. Histopathologic analysis of the immuno-PET-positive lesions revealed the presence of plasma cell infiltrates within the bone marrow. CONCLUSIONS 64Cu-labeled anti-CD138 antibody can detect subcutaneous MM tumors and bone marrow lesions with high sensitivity, outperforming 18F-FDG-PET and 64CuCl2 in this preclinical model. These data support 64Cu-anti-CD138 antibody as a specific and promising new imaging radiopharmaceutical agent in MM.
Collapse
Affiliation(s)
- Clément Bailly
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
| | - Sébastien Gouard
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
| | - Marie Lacombe
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nuclear Medicine Department, ICO-René Gauducheau Cancer Center, Saint-Herblain, France
| | - Patricia Remaud-Le Saëc
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
| | - Benjamin Chalopin
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
| | - Mickaël Bourgeois
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
- Groupement d’Intérêt Public ARRONAX, Saint-Herblain, France
| | - Nicolas Chouin
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
- AMaROC, Oniris, Ecole Nationale Vétérinaire, Agroalimentaire et de l'Alimentation de Nantes-Atlantique, Nantes, France
| | - Raphaël Tripier
- CNRS-UMR6521, University of Bretagne Occidentale, Brest, France
| | - Zakaria Halime
- CNRS-UMR6521, University of Bretagne Occidentale, Brest, France
| | - Ferid Haddad
- Groupement d’Intérêt Public ARRONAX, Saint-Herblain, France
| | - Alain Faivre-Chauvet
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
| | - Françoise Kraeber-Bodéré
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
- Nuclear Medicine Department, ICO-René Gauducheau Cancer Center, Saint-Herblain, France
| | - Michel Chérel
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
- Nuclear Medicine Department, ICO-René Gauducheau Cancer Center, Saint-Herblain, France
- Groupement d’Intérêt Public ARRONAX, Saint-Herblain, France
| | - Caroline Bodet-Milin
- Nuclear Medicine Department, University Hospital, Nantes, France
- Nantes-Angers Cancer Research Center CRCINA, University of Nantes, INSERM UMR1232, CNRS-ERL6001, Nantes, France
| |
Collapse
|
19
|
Qin Y, Zhu Y, Luo F, Chen C, Chen X, Wu M. Killing two birds with one stone: dual blockade of integrin and FGF signaling through targeting syndecan-4 in postoperative capsular opacification. Cell Death Dis 2017; 8:e2920. [PMID: 28703800 PMCID: PMC5550862 DOI: 10.1038/cddis.2017.315] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/03/2017] [Accepted: 06/05/2017] [Indexed: 12/12/2022]
Abstract
The most common complication after cataract surgery is postoperative capsular opacification, which includes anterior capsular opacification (ACO) and posterior capsular opacification (PCO). Increased adhesion of lens epithelial cells (LECs) to the intraocular lens material surface promotes ACO formation, whereas proliferation and migration of LECs to the posterior capsule lead to the development of PCO. Cell adhesion is mainly mediated by the binding of integrin to extracellular matrix proteins, while cell proliferation and migration are regulated by fibroblast growth factor (FGF). Syndecan-4 (SDC-4) is a co-receptor for both integrin and FGF signaling pathways. Therefore, SDC-4 may be an ideal therapeutic target for the prevention and treatment of postoperative capsular opacification. However, how SDC-4 contributes to FGF-mediated proliferation, migration, and integrin-mediated adhesion of LECs is unclear. Here, we found that downregulation of SDC-4 inhibited FGF signaling through the blockade of ERK1/2 and PI3K/Akt/mTOR activation, thus suppressing cell proliferation and migration. In addition, downregulation of SDC-4 suppressed integrin-mediated cell adhesion through inhibiting focal adhesion kinase (FAK) phosphorylation. Moreover, SDC-4 knockout mice exhibited normal lens morphology, but had significantly reduced capsular opacification after injury. Finally, SDC-4 expression level was increased in the anterior capsule LECs of age-related cataract patients. Taken together, we for the first time characterized the key regulatory role of SDC-4 in FGF and integrin signaling in human LECs, and provided the basis for future pharmacological interventions of capsular opacification.
Collapse
Affiliation(s)
- Yingyan Qin
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yi Zhu
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Furong Luo
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Chuan Chen
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xiaoyun Chen
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Mingxing Wu
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|
20
|
Ali AM, Dehdashti F, DiPersio JF, Cashen AF. Radioimmunotherapy-based conditioning for hematopoietic stem cell transplantation: Another step forward. Blood Rev 2016; 30:389-99. [PMID: 27174151 DOI: 10.1016/j.blre.2016.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 04/16/2016] [Accepted: 04/26/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Alaa M Ali
- Department of Internal Medicine, Washington University School of Medicine, 660 S Euclid Avenue, Campus 8058, St. Louis, MO 63110, USA.
| | - Farrokh Dehdashti
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA.
| | - John F DiPersio
- Department of Internal Medicine, Washington University School of Medicine, 660 S Euclid Avenue, Campus 8058, St. Louis, MO 63110, USA.
| | - Amanda F Cashen
- Department of Internal Medicine, Washington University School of Medicine, 660 S Euclid Avenue, Campus 8058, St. Louis, MO 63110, USA.
| |
Collapse
|
21
|
Diab M, Nguyen F, Berthaud M, Maurel C, Gaschet J, Verger E, Ibisch C, Rousseau C, Chérel M, Abadie J, Davodeau F. Production and characterization of monoclonal antibodies specific for canine CD138 (syndecan-1) for nuclear medicine preclinical trials on spontaneous tumours. Vet Comp Oncol 2016; 15:932-951. [PMID: 27076401 DOI: 10.1111/vco.12233] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/17/2015] [Accepted: 03/18/2016] [Indexed: 12/31/2022]
Abstract
We isolated 11 antibodies specific for canine CD138 (cCD138) to validate the interest of CD138 antigen targeting in dogs with spontaneous mammary carcinoma. The affinity of the monoclonal antibodies in the nanomolar range is suitable for immunohistochemistry and nuclear medicine applications. Four distinct epitopes were recognized on cCD138 by this panel of antibodies. CD138 expression in canine healthy tissues is comparable to that reported in humans. CD138 is frequently expressed in canine mammary carcinomas corresponding to the human triple negative breast cancer subtype, with cytoplasmic and membranous expression. In canine diffuse large B-cell lymphoma, CD138 expression is associated with the 'non-germinal center' phenotype corresponding to the most aggressive subtype in humans. This homology of CD138 expression between dogs and humans confirms the relevance of tumour-bearing dogs as spontaneous models for nuclear medicine applications, especially for the evaluation of new tumour targeting strategies for diagnosis by phenotypic imaging and radio-immunotherapy.
Collapse
Affiliation(s)
- M Diab
- Nantes-Angers Cancer Research Center CRCNA/INSERM UMR892, Nantes, France
| | - F Nguyen
- ONIRIS Nantes Atlantic National College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - M Berthaud
- Nantes-Angers Cancer Research Center CRCNA/INSERM UMR892, Nantes, France
| | - C Maurel
- Nantes-Angers Cancer Research Center CRCNA/INSERM UMR892, Nantes, France
| | - J Gaschet
- Nantes-Angers Cancer Research Center CRCNA/INSERM UMR892, Nantes, France
| | - E Verger
- Nantes-Angers Cancer Research Center CRCNA/INSERM UMR892, Nantes, France
| | - C Ibisch
- ONIRIS Nantes Atlantic National College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - C Rousseau
- ICO Integrated Center for Oncology, Nantes, France
| | - M Chérel
- Nantes-Angers Cancer Research Center CRCNA/INSERM UMR892, Nantes, France.,ICO Integrated Center for Oncology, Nantes, France
| | - J Abadie
- ONIRIS Nantes Atlantic National College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - F Davodeau
- Nantes-Angers Cancer Research Center CRCNA/INSERM UMR892, Nantes, France
| |
Collapse
|
22
|
Bodet-Milin C, Kraeber-Bodéré F, Eugène T, Guérard F, Gaschet J, Bailly C, Mougin M, Bourgeois M, Faivre-Chauvet A, Chérel M, Chevallier P. Radioimmunotherapy for Treatment of Acute Leukemia. Semin Nucl Med 2016; 46:135-46. [DOI: 10.1053/j.semnuclmed.2015.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
23
|
Guo B, Chen M, Han Q, Hui F, Dai H, Zhang W, Zhang Y, Wang Y, Zhu H, Han W. CD138-directed adoptive immunotherapy of chimeric antigen receptor (CAR)-modified T cells for multiple myeloma. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jocit.2014.11.001] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
24
|
Wang L, Zhang S, Ou J, Bai H. [Cytotoxity of pomalidomide combined CAR-T cell for multiple myeloma cell RPMI8226 and U266]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2016; 36:497-500. [PMID: 26134016 PMCID: PMC7343067 DOI: 10.3760/cma.j.issn.0253-2727.2015.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
目的 观察CD138-CAR-T细胞对人多发性骨髓瘤(MM)细胞株RPMI8226和U266细胞的杀伤作用,探讨泊马度胺对CD138-CAR-T细胞及其杀伤作用的影响。 方法 采用CFSE/7-AAD双标法检测CD138-CAR-T细胞及其联合泊马度胺对RPMI8226、U266细胞的杀伤活性。ELISA法检测CD138-CAR-T细胞分泌IFN-γ的变化。 结果 CD138-CAR-T细胞作用18 h后,对RPMI8226、U266细胞的杀伤活性分别为(55.2±3.9)%、(85.1±2.4)%,对照组分别为(7.0±1.5)%、(12.5±2.1)%,差异均有统计学意义(P值均<0.01);与CD138-CAR-T细胞组比较,CD138-CAR-T细胞联合泊马度胺(2.5 µg/ml)作用18 h后,对RPMI8226、U266细胞的杀伤活性差异无统计学意义(P值均>0.05)。与CD138-CAR-T细胞组比较,CD138-CAR-T细胞和MM细胞共培养组IFN-γ分泌水平显著增高;与共培养组比较,加入泊马度胺后能显著促进IFN-γ的释放,差异均有统计学意义(P值均<0.01)。 结论 CD138-CAR-T细胞对MM细胞及耐药细胞株均有明显的杀伤作用;其与MM细胞共培养能促进后者IFN-γ的分泌;泊马度胺能促进CD138-CAR-T细胞分泌IFN-γ。
Collapse
Affiliation(s)
- Lei Wang
- Department of Hematology, Center of Hematologic Diseases of Chinese PLA; Lanzhou Military Area General Hospital, Lanzhou 730050, China
| | - Shuting Zhang
- Department of Hematology, Center of Hematologic Diseases of Chinese PLA; Lanzhou Military Area General Hospital, Lanzhou 730050, China
| | - Jianfeng Ou
- Department of Hematology, Center of Hematologic Diseases of Chinese PLA; Lanzhou Military Area General Hospital, Lanzhou 730050, China
| | - Hai Bai
- Department of Hematology, Center of Hematologic Diseases of Chinese PLA; Lanzhou Military Area General Hospital, Lanzhou 730050, China
| |
Collapse
|
25
|
Fichou N, Gouard S, Maurel C, Barbet J, Ferrer L, Morgenstern A, Bruchertseifer F, Faivre-Chauvet A, Bigot-Corbel E, Davodeau F, Gaschet J, Chérel M. Single-Dose Anti-CD138 Radioimmunotherapy: Bismuth-213 is More Efficient than Lutetium-177 for Treatment of Multiple Myeloma in a Preclinical Model. Front Med (Lausanne) 2015; 2:76. [PMID: 26582128 PMCID: PMC4631990 DOI: 10.3389/fmed.2015.00076] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/19/2015] [Indexed: 11/21/2022] Open
Abstract
Objectives Radioimmunotherapy (RIT) has emerged as a potential treatment option for multiple myeloma (MM). In humans, a dosimetry study recently showed the relevance of RIT using an antibody targeting the CD138 antigen. The therapeutic efficacy of RIT using an anti-CD138 antibody coupled to 213Bi, an α-emitter, was also demonstrated in a preclinical MM model. Since then, RIT with β-emitters has shown efficacy in treating hematologic cancer. In this paper, we investigate the therapeutic efficacy of RIT in the 5T33 murine MM model using a new anti-CD138 monoclonal antibody labeled either with 213Bi for α-RIT or 177Lu for β-RIT. Methods A new monoclonal anti-CD138 antibody, 9E7.4, was generated by immunizing a rat with a murine CD138-derived peptide. Antibody specificity was validated by flow cytometry, biodistribution, and α-RIT studies. Then, a β-RIT dose-escalation assay with the 177Lu-radiolabeled 9E7.4 mAb was performed in KalwRij C57/BL6 mice 10 days after i.v. engraftment with 5T33 MM cells. Animal survival and toxicological parameters were assessed to define the optimal activity. Results α-RIT performed with 3.7 MBq of 213Bi-labeled 9E7.4 anti-CD138 mAb increased median survival to 80 days compared to 37 days for the untreated control and effected cure in 45% of animals. β-RIT performed with 18.5 MBq of 177Lu-labeled 9E7.4 mAb was well tolerated and significantly increased mouse survival (54 vs. 37 days in the control group); however, no mice were cured with this treatment. Conclusion This study revealed the advantages of α-RIT in the treatment of MM in a preclinical model where β-RIT shows almost no efficacy.
Collapse
Affiliation(s)
- Nolwenn Fichou
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France
| | - Sébastien Gouard
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France
| | - Catherine Maurel
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France
| | - Jacques Barbet
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France
| | - Ludovic Ferrer
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France ; Institut de Cancérologie de l'Ouest , Saint-Herblain , France
| | | | | | - Alain Faivre-Chauvet
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France ; Nuclear Medicine Department, CHU Nantes , Nantes , France
| | - Edith Bigot-Corbel
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France
| | - François Davodeau
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France
| | - Joëlle Gaschet
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France
| | - Michel Chérel
- Centre Régional de Recherche en Cancérologie Nantes/Angers (CRCNA) - UMR 892 INSERM, Université de Nantes , Nantes , France ; CNRS 6299, Université de Nantes , Nantes , France ; Université de Nantes , Nantes , France ; Institut de Cancérologie de l'Ouest , Saint-Herblain , France
| |
Collapse
|
26
|
Akl MR, Nagpal P, Ayoub NM, Prabhu SA, Gliksman M, Tai B, Hatipoglu A, Goy A, Suh KS. Molecular and clinical profiles of syndecan-1 in solid and hematological cancer for prognosis and precision medicine. Oncotarget 2015; 6:28693-715. [PMID: 26293675 PMCID: PMC4745686 DOI: 10.18632/oncotarget.4981] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/11/2015] [Indexed: 12/18/2022] Open
Abstract
Syndecan-1 (SDC1, CD138) is a key cell surface adhesion molecule essential for maintaining cell morphology and interaction with the surrounding microenvironment. Deregulation of SDC1 contributes to cancer progression by promoting cell proliferation, metastasis, invasion and angiogenesis, and is associated with relapse through chemoresistance. SDC1 expression level is also associated with responses to chemotherapy and with prognosis in multiple solid and hematological cancers, including multiple myeloma and Hodgkin lymphoma. At the tissue level, the expression levels of SDC1 and the released extracellular domain of SDC1 correlate with tumor malignancy, phenotype, and metastatic potential for both solid and hematological tumors in a tissue-specific manner. The SDC1 expression profile varies among cancer types, but the differential expression signatures between normal and cancer cells in epithelial and stromal compartments are directly associated with aggressiveness of tumors and patient's clinical outcome and survival. Therefore, relevant biomarkers of SDC signaling may be useful for selecting patients that would most likely respond to a particular therapy at the time of diagnosis or perhaps for predicting relapse. In addition, the reciprocal expression signature of SDC between tumor epithelial and stromal compartments may have synergistic value for patient selection and the prediction of clinical outcome.
Collapse
Affiliation(s)
- Mohamed R. Akl
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Poonam Nagpal
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Sathyen A. Prabhu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Matthew Gliksman
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Betty Tai
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Ahmet Hatipoglu
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Andre Goy
- Lymphoma Division, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - K. Stephen Suh
- Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
27
|
Targeted Therapies in Adult B-Cell Malignancies. BIOMED RESEARCH INTERNATIONAL 2015; 2015:217593. [PMID: 26425544 PMCID: PMC4575712 DOI: 10.1155/2015/217593] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/03/2015] [Accepted: 05/05/2015] [Indexed: 12/17/2022]
Abstract
B-lymphocytes are programmed for the production of immunoglobulin (Ig) after antigen presentation, in the context of T-lymphocyte control within lymphoid organs. During this differentiation/activation process, B-lymphocytes exhibit different restricted or common surface markers, activation of cellular pathways that regulate cell cycle, metabolism, proteasome activity, and protein synthesis. All molecules involved in these different cellular mechanisms are potent therapeutic targets. Nowadays, due to the progress of the biology, more and more targeted drugs are identified, a situation that is correlated with an extended field of the targeted therapy. The full knowledge of the cellular machinery and cell-cell communication allows making the best choice to treat patients, in the context of personalized medicine. Also, focus should not be restricted to the immediate effects observed as clinical endpoints, that is, response rate, survival markers with conventional statistical methods, but it should consider the prediction of different clinical consequences due to other collateral drug targets, based on new methodologies. This means that new reflection and new bioclinical follow-up have to be monitored, particularly with the new drugs used with success in B-cell malignancies. This review discussed the principal aspects of such evident bioclinical progress.
Collapse
|
28
|
Pojero F, Flores-Montero J, Sanoja L, Pérez JJ, Puig N, Paiva B, Bottcher S, van Dongen JJM, Orfao A. Utility of CD54, CD229, and CD319 for the identification of plasma cells in patients with clonal plasma cell diseases. CYTOMETRY PART B-CLINICAL CYTOMETRY 2015; 90:91-100. [PMID: 26130131 DOI: 10.1002/cyto.b.21269] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 05/22/2015] [Accepted: 06/25/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Multiparameter flow cytometry (MFC) identification and characterization of plasma cells (PCs) is a useful tool to support diagnosis, prognostication, and monitoring of PC diseases (PCD). Currently, the number of MFC markers suited for the identification of PC remains limited. Moreover, antibody therapies against PC-associated markers further compromise the utility of the most widely used reagents (e.g., CD38). Despite markers other than CD38 and CD138 are recognized as potentially useful PC-identification markers, no study has comparatively evaluated their performance in combination with CD38 and CD138. Here we compared the utility of CD229, CD54, and CD319 for the identification of normal and aberrant PCs. METHODS Bone marrow (BM) samples from 5 healthy controls, two noninfiltrated nonHodgkin lymphoma cases and 46 PCD patients plus 3 extraosseous plasmocytomas, and normal peripheral blood (PB) specimens, were studied. RESULTS Our results showed adequate performance of all three markers once combined with CD38. In contrast, when combined with CD138 for the identification of PC, only CD229 provided a good discrimination between PCs and all other cells for all BM and PB samples analyzed; in contrast, CD54 and CD319 showed limited utility for the identification of PCs, mainly because of significant overlap of the staining for these two markers on PCs and other myeloid cells in the sample. CONCLUSIONS From the three markers evaluated, CD229 may be considered as the most reliable marker to replace CD38 or CD138 for the identification of PCs in patients undergoing anti-CD38 or anti-CD138 therapy, until a better alternative is available.
Collapse
Affiliation(s)
- Fanny Pojero
- Centro de Investigación del Cáncer (Instituto de Biología Molecular y Celular del Cáncer, CSIC-USAL), Instituto Biosanitario de Salamanca (IBSAL), Servicio General de Citometría y Departamento De Medicina (NUCLEUS), Universidad de Salamanca (Salamanca), Spain.,Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Universita' degli Studi di Palermo, Palermo, Italy
| | - Juan Flores-Montero
- Centro de Investigación del Cáncer (Instituto de Biología Molecular y Celular del Cáncer, CSIC-USAL), Instituto Biosanitario de Salamanca (IBSAL), Servicio General de Citometría y Departamento De Medicina (NUCLEUS), Universidad de Salamanca (Salamanca), Spain
| | - Luzalba Sanoja
- Centro de Investigación del Cáncer (Instituto de Biología Molecular y Celular del Cáncer, CSIC-USAL), Instituto Biosanitario de Salamanca (IBSAL), Servicio General de Citometría y Departamento De Medicina (NUCLEUS), Universidad de Salamanca (Salamanca), Spain
| | - José Juan Pérez
- Departmento de Hematología, Hospital Universitario de Salamanca, IBSAL; IBMCC (USAL-CSIC), Salamanca, Spain
| | - Noemí Puig
- Departmento de Hematología, Hospital Universitario de Salamanca, IBSAL; IBMCC (USAL-CSIC), Salamanca, Spain
| | - Bruno Paiva
- Clínica Universidad de Navarra; Centro de Investigaciones Médicas Aplicadas (CIMA), Pamplona, Spain
| | - Sebastian Bottcher
- Second Department of Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Jacques J M van Dongen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam (Erasmus MC), Rotterdamthe, Netherlands
| | - Alberto Orfao
- Centro de Investigación del Cáncer (Instituto de Biología Molecular y Celular del Cáncer, CSIC-USAL), Instituto Biosanitario de Salamanca (IBSAL), Servicio General de Citometría y Departamento De Medicina (NUCLEUS), Universidad de Salamanca (Salamanca), Spain
| | | |
Collapse
|
29
|
Theocharis AD, Skandalis SS, Neill T, Multhaupt HAB, Hubo M, Frey H, Gopal S, Gomes A, Afratis N, Lim HC, Couchman JR, Filmus J, Sanderson RD, Schaefer L, Iozzo RV, Karamanos NK. Insights into the key roles of proteoglycans in breast cancer biology and translational medicine. Biochim Biophys Acta Rev Cancer 2015; 1855:276-300. [PMID: 25829250 DOI: 10.1016/j.bbcan.2015.03.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/27/2015] [Accepted: 03/24/2015] [Indexed: 12/18/2022]
Abstract
Proteoglycans control numerous normal and pathological processes, among which are morphogenesis, tissue repair, inflammation, vascularization and cancer metastasis. During tumor development and growth, proteoglycan expression is markedly modified in the tumor microenvironment. Altered expression of proteoglycans on tumor and stromal cell membranes affects cancer cell signaling, growth and survival, cell adhesion, migration and angiogenesis. Despite the high complexity and heterogeneity of breast cancer, the rapid evolution in our knowledge that proteoglycans are among the key players in the breast tumor microenvironment suggests their potential as pharmacological targets in this type of cancer. It has been recently suggested that pharmacological treatment may target proteoglycan metabolism, their utilization as targets for immunotherapy or their direct use as therapeutic agents. The diversity inherent in the proteoglycans that will be presented herein provides the potential for multiple layers of regulation of breast tumor behavior. This review summarizes recent developments concerning the biology of selected proteoglycans in breast cancer, and presents potential targeted therapeutic approaches based on their novel key roles in breast cancer.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Hinke A B Multhaupt
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Mario Hubo
- University of Frankfurt, Institute of Pharmacology and Toxicology, Theodor-Stern Kai 7, Frankfurt 60590, Germany
| | - Helena Frey
- University of Frankfurt, Institute of Pharmacology and Toxicology, Theodor-Stern Kai 7, Frankfurt 60590, Germany
| | - Sandeep Gopal
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Angélica Gomes
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Nikos Afratis
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Hooi Ching Lim
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - John R Couchman
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Jorge Filmus
- Department of Biological Sciences, Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Canada
| | - Ralph D Sanderson
- University of Alabama at Birmingham, Department of Pathology, UAB Comprehensive Cancer Center, 1720 2nd Ave. S, WTI 602B, Birmingham, AL 35294, USA
| | - Liliana Schaefer
- University of Frankfurt, Institute of Pharmacology and Toxicology, Theodor-Stern Kai 7, Frankfurt 60590, Germany
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
30
|
Theocharis AD, Gialeli C, Bouris P, Giannopoulou E, Skandalis SS, Aletras AJ, Iozzo RV, Karamanos NK. Cell-matrix interactions: focus on proteoglycan-proteinase interplay and pharmacological targeting in cancer. FEBS J 2014; 281:5023-42. [PMID: 25333340 PMCID: PMC5036392 DOI: 10.1111/febs.12927] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/04/2014] [Accepted: 07/09/2014] [Indexed: 01/10/2023]
Abstract
Proteoglycans are major constituents of extracellular matrices, as well as cell surfaces and basement membranes. They play key roles in supporting the dynamic extracellular matrix by generating complex structural networks with other macromolecules and by regulating cellular phenotypes and signaling. It is becoming evident, however, that proteolytic enzymes are required partners for matrix remodeling and for modulating cell signaling via matrix constituents. Proteinases contribute to all stages of diseases, particularly cancer development and progression, and contextually participate in either the removal of damaged products or in the processing of matrix molecules and signaling receptors. The dynamic interplay between proteoglycans and proteolytic enzymes is a crucial biological step that contributes to the pathophysiology of cancer and inflammation. Moreover, proteoglycans are implicated in the expression and secretion of proteolytic enzymes and often modulate their activities. In this review, we describe the emerging biological roles of proteoglycans and proteinases, with a special emphasis on their complex interplay. We critically evaluate this important proteoglycan-proteinase interactome and discuss future challenges with respect to targeting this axis in the treatment of cancer.
Collapse
Affiliation(s)
- Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Chrisostomi Gialeli
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Panagiotis Bouris
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Efstathia Giannopoulou
- Clinical Oncology Laboratory, Division of Oncology, University Hospital of Patras, Patras Medical School, Patras 26110, Greece
| | - Spyros S. Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Alexios J. Aletras
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece
| |
Collapse
|
31
|
Kraeber-Bodéré F, Bodet-Milin C, Rousseau C, Eugène T, Pallardy A, Frampas E, Carlier T, Ferrer L, Gaschet J, Davodeau F, Gestin JF, Faivre-Chauvet A, Barbet J, Chérel M. Radioimmunoconjugates for the treatment of cancer. Semin Oncol 2014; 41:613-22. [PMID: 25440606 DOI: 10.1053/j.seminoncol.2014.07.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Radioimmunotherapy (RIT) has been developed for more than 30 years. Two products targeting the CD20 antigen are approved in the treatment of non-Hodgkin B-cell lymphoma (NHBL): iodine 131-tositumomab and yttrium 90-ibritumomab tiuxetan. RIT can be integrated in clinical practice for the treatment of patients with relapsed or refractory follicular lymphoma (FL) or as consolidation after induction chemotherapy. High-dose treatment, RIT in first-line treatment, fractionated RIT, and use of new humanized monoclonal antibodies (MAbs), in particular targeting CD22, showed promising results in NHBL. In other hemopathies, such as multiple myeloma, efficacy has been demonstrated in preclinical studies. In solid tumors, more resistant to radiation and less accessible to large molecules such as MAbs, clinical efficacy remains limited. However, pretargeting methods have shown clinical efficacy. Finally, new beta emitters such as lutetium 177, with better physical properties will further improve the safety of RIT and alpha emitters, such as bismuth 213 or astatine 211, offer the theoretical possibility to eradicate the last microscopic clusters of tumor cells, in the consolidation setting. Personalized treatments, based on quantitative positron emission tomography (PET), pre-therapeutic imaging, and dosimetry procedures, also could be applied to adapt injected activity to each patient.
Collapse
Affiliation(s)
- Françoise Kraeber-Bodéré
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France; Department of Nuclear Medicine, CHU de Nantes, Nantes, France; Department of Nuclear Medicine, ICO-René Gauducheau, Saint-Herblain, France.
| | - Caroline Bodet-Milin
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France; Department of Nuclear Medicine, CHU de Nantes, Nantes, France
| | - Caroline Rousseau
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France; Department of Nuclear Medicine, ICO-René Gauducheau, Saint-Herblain, France
| | - Thomas Eugène
- Department of Nuclear Medicine, CHU de Nantes, Nantes, France
| | | | - Eric Frampas
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France; Department of Radiology, CHU de Nantes, Nantes, France
| | - Thomas Carlier
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France; Department of Nuclear Medicine, CHU de Nantes, Nantes, France
| | - Ludovic Ferrer
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France; Department of Nuclear Medicine, ICO-René Gauducheau, Saint-Herblain, France
| | - Joëlle Gaschet
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France
| | - François Davodeau
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France
| | - Jean-François Gestin
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France
| | - Alain Faivre-Chauvet
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France; Department of Nuclear Medicine, ICO-René Gauducheau, Saint-Herblain, France
| | - Jacques Barbet
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France; GIP Arronax, Saint-Herblain, France. This work has been in part supported by a grant from the French National Agency for Research called "Investissements d'Avenir" n°ANR-11-LABX-0018-01 and by a grant from the Pays de la Loire Regional Council called "NucSan"
| | - Michel Chérel
- Centre de Recherche en Cancérologie de Nantes-Angers, Inserm, Université de Nantes, Nantes, France; Department of Nuclear Medicine, ICO-René Gauducheau, Saint-Herblain, France
| |
Collapse
|
32
|
Bae J, Prabhala R, Voskertchian A, Brown A, Maguire C, Richardson P, Dranoff G, Anderson KC, Munshi NC. A multiepitope of XBP1, CD138 and CS1 peptides induces myeloma-specific cytotoxic T lymphocytes in T cells of smoldering myeloma patients. Leukemia 2014; 29:218-29. [PMID: 24935722 PMCID: PMC4237716 DOI: 10.1038/leu.2014.159] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/24/2014] [Accepted: 04/02/2014] [Indexed: 02/07/2023]
Abstract
We evaluated a cocktail of HLA-A2-specific peptides including heteroclitic XBP1 US184-192 (YISPWILAV), heteroclitic XBP1 SP367-375 (YLFPQLISV), native CD138260-268 (GLVGLIFAV) and native CS1239-247 (SLFVLGLFL), for their ability to elicit multipeptide-specific cytotoxic T lymphocytes (MP-CTLs) using T cells from smoldering multiple myeloma (SMM) patients. Our results demonstrate that MP-CTLs generated from SMM patients' T cells show effective anti-MM responses including CD137 (4-1BB) upregulation, CTL proliferation, interferon-γ production and degranulation (CD107a) in an HLA-A2-restricted and peptide-specific manner. Phenotypically, we observed increased total CD3(+)CD8(+) T cells (>80%) and cellular activation (CD69(+)) within the memory SMM MP-CTL (CD45RO(+)/CD3(+)CD8(+)) subset after repeated multipeptide stimulation. Importantly, SMM patients could be categorized into distinct groups by their level of MP-CTL expansion and antitumor activity. In high responders, the effector memory (CCR7(-)CD45RO(+)/CD3(+)CD8(+)) T-cell subset was enriched, whereas the remaining responders' CTL contained a higher frequency of the terminal effector (CCR7(-)CD45RO(-)/CD3(+)CD8(+)) subset. These results suggest that this multipeptide cocktail has the potential to induce effective and durable memory MP-CTL in SMM patients. Therefore, our findings provide the rationale for clinical evaluation of a therapeutic vaccine to prevent or delay progression of SMM to active disease.
Collapse
Affiliation(s)
- J Bae
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - R Prabhala
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA [3] VA Boston Healthcare System, Boston, MA, USA
| | - A Voskertchian
- Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - A Brown
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - C Maguire
- Tufts University School of Medicine, Boston, MA, USA
| | - P Richardson
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - G Dranoff
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - K C Anderson
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA
| | - N C Munshi
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA [2] Harvard Medical School, Boston, MA, USA [3] VA Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
33
|
Gouard S, Pallardy A, Gaschet J, Faivre-Chauvet A, Bruchertseifer F, Morgenstern A, Maurel C, Matous E, Kraeber-Bodéré F, Davodeau F, Chérel M. Comparative analysis of multiple myeloma treatment by CD138 antigen targeting with bismuth-213 and Melphalan chemotherapy. Nucl Med Biol 2014; 41 Suppl:e30-5. [DOI: 10.1016/j.nucmedbio.2014.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 02/28/2014] [Indexed: 11/25/2022]
|
34
|
Abstract
Monoclonal antibody therapy has revolutionized cancer treatment by significantly improving patient survival both in solid tumors and hematologic malignancies. Recent technological advances have increased the effectiveness of immunotherapy leading to its broader application in diverse treatment settings. Immunoconjugates (ICs) consist of a cytotoxic effector covalently linked to a monoclonal antibody that enables the targeted delivery of its therapeutic payload to tumors based on cell-surface receptor recognition. ICs are classified into 3 groups based on their effector type: immunotoxins (protein toxin), radioimmunoconjugates (radionuclide), and antibody drug conjugates (small-molecule drug). Optimization of each individual component of an IC (antibody, linker, and effector) is essential for therapeutic efficacy. Clinical trials have been conducted to investigate the effectiveness of ICs in hematologic malignancies both as monotherapy and in multiagent regimens in relapsed/refractory disease as well as frontline settings. These studies have yielded encouraging results particularly in lymphoma. ICs comprise an exciting group of therapeutics that promise to play an increasingly important role in the management of hematologic malignancies.
Collapse
|
35
|
Barbouri D, Afratis N, Gialeli C, Vynios DH, Theocharis AD, Karamanos NK. Syndecans as modulators and potential pharmacological targets in cancer progression. Front Oncol 2014; 4:4. [PMID: 24551591 PMCID: PMC3910246 DOI: 10.3389/fonc.2014.00004] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/09/2014] [Indexed: 12/17/2022] Open
Abstract
Extracellular matrix (ECM) components form a dynamic network of key importance for cell function and properties. Key macromolecules in this interplay are syndecans (SDCs), a family of transmembrane heparan sulfate proteoglycans (HSPGs). Specifically, heparan sulfate (HS) chains with their different sulfation pattern have the ability to interact with growth factors and their receptors in tumor microenvironment, promoting the activation of different signaling cascades that regulate tumor cell behavior. The affinity of HS chains with ligands is altered during malignant conditions because of the modification of chain sequence/sulfation pattern. Furthermore, matrix degradation enzymes derived from the tumor itself or the tumor microenvironment, like heparanase and matrix metalloproteinases, ADAM as well as ADAMTS are involved in the cleavage of SDCs ectodomain at the HS and protein core level, respectively. Such released soluble SDCs "shed SDCs" in the ECM interact in an autocrine or paracrine manner with the tumor or/and stromal cells. Shed SDCs, upon binding to several matrix effectors, such as growth factors, chemokines, and cytokines, have the ability to act as competitive inhibitors for membrane proteoglycans, and modulate the inflammatory microenvironment of cancer cells. It is notable that SDCs and their soluble counterparts may affect either the behavior of cancer cells and/or their microenvironment during cancer progression. The importance of these molecules has been highlighted since HSPGs have been proposed as prognostic markers of solid tumors and hematopoietic malignancies. Going a step further down the line, the multi-actions of SDCs in many levels make them appealing as potential pharmacological targets, either by targeting directly the tumor or indirectly the adjacent stroma.
Collapse
Affiliation(s)
- Despoina Barbouri
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Nikolaos Afratis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Chrisostomi Gialeli
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Demitrios H. Vynios
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
36
|
Green DJ, Orgun NN, Jones JC, Hylarides MD, Pagel JM, Hamlin DK, Wilbur DS, Lin Y, Fisher DR, Kenoyer AL, Frayo SL, Gopal AK, Orozco JJ, Gooley TA, Wood BL, Bensinger WI, Press OW. A preclinical model of CD38-pretargeted radioimmunotherapy for plasma cell malignancies. Cancer Res 2013; 74:1179-89. [PMID: 24371230 DOI: 10.1158/0008-5472.can-13-1589] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The vast majority of patients with plasma cell neoplasms die of progressive disease despite high response rates to novel agents. Malignant plasma cells are very radiosensitive, but the potential role of radioimmunotherapy (RIT) in the management of plasmacytomas and multiple myeloma has undergone only limited evaluation. Furthermore, CD38 has not been explored as a RIT target despite its uniform high expression on malignant plasma cells. In this report, both conventional RIT (directly radiolabeled antibody) and streptavidin-biotin pretargeted RIT (PRIT) directed against the CD38 antigen were assessed as approaches to deliver radiation doses sufficient for multiple myeloma cell eradication. PRIT demonstrated biodistributions that were markedly superior to conventional RIT. Tumor-to-blood ratios as high as 638:1 were seen 24 hours after PRIT, whereas ratios never exceeded 1:1 with conventional RIT. (90)Yttrium absorbed dose estimates demonstrated excellent target-to-normal organ ratios (6:1 for the kidney, lung, liver; 10:1 for the whole body). Objective remissions were observed within 7 days in 100% of the mice treated with doses ranging from 800 to 1,200 μCi of anti-CD38 pretargeted (90)Y-DOTA-biotin, including 100% complete remissions (no detectable tumor in treated mice compared with tumors that were 2,982% ± 2,834% of initial tumor volume in control animals) by day 23. Furthermore, 100% of animals bearing NCI-H929 multiple myeloma tumor xenografts treated with 800 μCi of anti-CD38 pretargeted (90)Y-DOTA-biotin achieved long-term myeloma-free survival (>70 days) compared with none (0%) of the control animals.
Collapse
Affiliation(s)
- Damian J Green
- Authors' Affiliations: Clinical Research Division, Fred Hutchinson Cancer Research Center; Departments of Medicine, Radiation Oncology, and Laboratory Medicine, University of Washington, Seattle; and Dade Moeller Health Group, Richland, Washington
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lückerath K, Lapa C, Spahmann A, Jörg G, Samnick S, Rosenwald A, Einsele H, Knop S, Buck AK. Targeting paraprotein biosynthesis for non-invasive characterization of myeloma biology. PLoS One 2013; 8:e84840. [PMID: 24376850 PMCID: PMC3871597 DOI: 10.1371/journal.pone.0084840] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 11/19/2013] [Indexed: 12/20/2022] Open
Abstract
Purpose Multiple myeloma is a hematologic malignancy originating from clonal plasma cells. Despite effective therapies, outcomes are highly variable suggesting marked disease heterogeneity. The role of functional imaging for therapeutic management of myeloma, such as positron emission tomography with 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG-PET), remains to be determined. Although some studies already suggested a prognostic value of 18F-FDG-PET, more specific tracers addressing hallmarks of myeloma biology, e.g. paraprotein biosynthesis, are needed. This study evaluated the amino acid tracers L-methyl-[11C]-methionine (11C-MET) and [18F]-fluoroethyl-L-tyrosine (18F-Fet) for their potential to image myeloma and to characterize tumor heterogeneity. Experimental Design To study the utility of 11C-MET, 18F-Fet and 18F-FDG for myeloma imaging, time activity curves were compared in various human myeloma cell lines (INA-6, MM1.S, OPM-2) and correlated to cell-biological characteristics, such as marker gene expression and immunoglobulin levels. Likewise, patient-derived CD138+ plasma cells were characterized regarding uptake and biomedical features. Results Using myeloma cell lines and patient-derived CD138+ plasma cells, we found that the relative uptake of 11C-MET exceeds that of 18F-FDG 1.5- to 5-fold and that of 18F-Fet 7- to 20-fold. Importantly, 11C-MET uptake significantly differed between cell types associated with worse prognosis (e.g. t(4;14) in OPM-2 cells) and indolent ones and correlated with intracellular immunoglobulin light chain and cell surface CD138 and CXCR4 levels. Direct comparison of radiotracer uptake in primary samples further validated the superiority of 11C-MET. Conclusion These data suggest that 11C-MET might be a versatile biomarker for myeloma superior to routine functional imaging with 18F-FDG regarding diagnosis, risk stratification, prognosis and discrimination of tumor subtypes.
Collapse
Affiliation(s)
- Katharina Lückerath
- University Wuerzburg, Medical Center, Department of Nuclear Medicine, Wuerzburg, Germany
- * E-mail:
| | - Constantin Lapa
- University Wuerzburg, Medical Center, Department of Nuclear Medicine, Wuerzburg, Germany
| | - Annika Spahmann
- University Wuerzburg, Medical Center, Department of Nuclear Medicine, Wuerzburg, Germany
| | - Gerhard Jörg
- University Wuerzburg, Medical Center, Department of Nuclear Medicine, Wuerzburg, Germany
| | - Samuel Samnick
- University Wuerzburg, Medical Center, Department of Nuclear Medicine, Wuerzburg, Germany
| | | | - Herrmann Einsele
- University Wuerzburg, Medical Center, Department of Hematology and Oncology, Wuerzburg, Germany
| | - Stefan Knop
- University Wuerzburg, Medical Center, Department of Hematology and Oncology, Wuerzburg, Germany
| | - Andreas K. Buck
- University Wuerzburg, Medical Center, Department of Nuclear Medicine, Wuerzburg, Germany
| |
Collapse
|
38
|
Jiang H, Zhang W, Shang P, Zhang H, Fu W, Ye F, Zeng T, Huang H, Zhang X, Sun W, Man-Yuen Sze D, Yi Q, Hou J. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Mol Oncol 2013; 8:297-310. [PMID: 24388357 DOI: 10.1016/j.molonc.2013.12.001] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 12/13/2022] Open
Abstract
Reprogramming of NK cells with a chimeric antigen receptor (CAR) proved an effective strategy to increase NK cell reactivity and recognition specificity toward tumor cells. To enhance the cytotoxicity of NK cells against CD138-positive multiple myeloma (MM) cells, we generated genetically modified NK-92MI cells carrying a CAR that consists of an anti-CD138 single-chain variable fragment (scFv) fused to the CD3ζ chain as a signaling moiety. The genetic modification through a lentiviral vector did not affect the intrinsic cytolytic activity of NK-92MI toward human erythroleukemic cell line K562 cells or CD138-negative targets. However, these retargeted NK-92MI (NK-92MI-scFv) displayed markedly enhanced cytotoxicity against CD138-positive human MM cell lines (RPMI8226, U266 and NCI-H929) and primary MM cells at various effector-to-target ratios (E:T) as compared to the empty vector-transfected NK-92MI (NK-92MI-mock). In line with the enhanced cytotoxicity of NK-92MI-scFv, significant elevations in the secretion of granzyme B, interferon-γ and proportion of CD107a expression were also found in NK-92MI-scFv in response to CD138-positive targets compared with NK-92MI-mock. Most importantly, the enhancement in the cytotoxicity of NK-92MI-scFv did not attenuate with 10Gy-irradiation that sufficiently blocked cell proliferation. Moreover, the irradiated NK-92MI-scFv exerted definitely intensified anti-tumor activity toward CD138-positive MM cells than NK-92MI-mock in the xenograft NOD-SCID mouse model. This study provides the rationale and feasibility for adoptive immunotherapy with CD138-specific CAR-modified NK cells in CD138-positive plasmacytic malignancies, which potentially further improves remission quality and prolongs the remission duration of patients with MM after upfront chemotherapy.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Rd, Shanghai 200003, China
| | - Wenhao Zhang
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Rd, Shanghai 200003, China
| | - Peipei Shang
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Rd, Shanghai 200003, China
| | - Hui Zhang
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Rd, Shanghai 200003, China
| | - Weijun Fu
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Rd, Shanghai 200003, China
| | - Fei Ye
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Rd, Shanghai 200003, China
| | - Tianmei Zeng
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Rd, Shanghai 200003, China
| | - Hejing Huang
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Rd, Shanghai 200003, China
| | - Xueguang Zhang
- Institute of Biotechnology and Clinical Immunology, Research Laboratory of Jiangsu Province, Soochow University, Suzhou 215007, China
| | - Wanping Sun
- Institute of Biotechnology and Clinical Immunology, Research Laboratory of Jiangsu Province, Soochow University, Suzhou 215007, China
| | - Daniel Man-Yuen Sze
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Region
| | - Qing Yi
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, and Center for Cancer Immunology Research, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Jian Hou
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Rd, Shanghai 200003, China.
| |
Collapse
|
39
|
Imaging and radioimmunotherapy of multiple myeloma with anti-idiotypic Nanobodies. Leukemia 2013; 28:444-7. [DOI: 10.1038/leu.2013.292] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Chérel M, Gouard S, Gaschet J, Saï-Maurel C, Bruchertseifer F, Morgenstern A, Bourgeois M, Gestin JF, Bodéré FK, Barbet J, Moreau P, Davodeau F. 213Bi Radioimmunotherapy with an Anti-mCD138 Monoclonal Antibody in a Murine Model of Multiple Myeloma. J Nucl Med 2013; 54:1597-604. [DOI: 10.2967/jnumed.112.111997] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
41
|
Heel K, Tabone T, Röhrig KJ, Maslen PG, Meehan K, Grimwade LF, Erber WN. Developments in the immunophenotypic analysis of haematological malignancies. Blood Rev 2013; 27:193-207. [PMID: 23845589 DOI: 10.1016/j.blre.2013.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunophenotyping is the method by which antibodies are used to detect cellular antigens in clinical samples. Although the major role is in the diagnosis and classification of haematological malignancies, applications have expanded over the past decade. Immunophenotyping is now used extensively for disease staging and monitoring, to detect surrogate markers of genetic aberrations, to identify potential immuno-therapeutic targets and to aid prognostic prediction. This expansion in applications has resulted from developments in antibodies, methodology, automation and data handling. In this review we describe recent advances in both the technology and applications for the analysis of haematological malignancies. We highlight the importance of the expanding repertoire of testing capability for diagnostic, prognostic and therapeutic applications. The impact and significance of immunophenotyping in the assessment of haematological neoplasms are evident.
Collapse
Affiliation(s)
- Kathy Heel
- Pathology and Laboratory Medicine, University of Western Australia, Crawley, WA 6009, Australia.
| | | | | | | | | | | | | |
Collapse
|
42
|
Allegra A, Penna G, Alonci A, Russo S, Greve B, Innao V, Minardi V, Musolino C. Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma. Eur J Haematol 2013; 90:441-68. [DOI: 10.1111/ejh.12107] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2013] [Indexed: 12/12/2022]
Affiliation(s)
| | - Giuseppa Penna
- Division of Haematology; University of Messina; Messina; Italy
| | - Andrea Alonci
- Division of Haematology; University of Messina; Messina; Italy
| | - Sabina Russo
- Division of Haematology; University of Messina; Messina; Italy
| | - Bruna Greve
- Division of Haematology; University of Messina; Messina; Italy
| | - Vanessa Innao
- Division of Haematology; University of Messina; Messina; Italy
| | - Viviana Minardi
- Division of Haematology; University of Messina; Messina; Italy
| | | |
Collapse
|
43
|
Orecchia P, Conte R, Balza E, Petretto A, Mauri P, Mingari MC, Carnemolla B. A novel human anti-syndecan-1 antibody inhibits vascular maturation and tumour growth in melanoma. Eur J Cancer 2013; 49:2022-33. [PMID: 23352437 DOI: 10.1016/j.ejca.2012.12.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 12/17/2012] [Accepted: 12/20/2012] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Syndecan-1 is a cell membrane protein that, after its shedding by heparanase enzymes, is accumulated in the extracellular matrix of some tumours, e.g. myeloma and lung carcinoma, where it modulates several key processes of tumourigenesis such as cancer cell proliferation and apoptosis, angiogenesis and metastasis. Few studies have focused on syndecan-1 in malignant melanoma, a tumour for which new therapeutic targets are desperately needed. We aimed to investigate the role of syndecan-1 in melanoma and to evaluate the potential therapeutic efficacy of a novel fully human anti-syndecan-1 recombinant antibody in this deadly disease. METHODS The OC-46F2 recombinant antibody was generated by selecting a human antibody phage display library on human melanoma cells and by its expression in mammalian cells. The specific antigen recognised by the antibody was identified by mass spectrometry. Murine models of human melanoma and ovarian carcinoma were used in the pre-clinical in vivo experiments. RESULTS The fully human antibody OC-46F2, specific for the extracellular domain of syndecan-1, inhibited vascular maturation and tumour growth in an experimental human melanoma model. The therapeutic efficacy of this antibody was also demonstrated in an experimental ovarian carcinoma model. A co-distribution of syndecan-1 with vascular endothelial growth factor receptor 2 (VEGFR2) observed in the intratumour melanoma microenvironment was absent in the tumours from mice treated with OC-46F2 scFv. CONCLUSION These findings highlight the role of syndecan-1 as a potential therapeutic target in melanoma and ovarian carcinoma and provide a new tool able to block vessel maturation, one of the mechanisms that underpin the angiogenic process essential for solid tumour growth.
Collapse
Affiliation(s)
- Paola Orecchia
- Laboratory of Immunology, IRCCS AOU San Martino-IST, Genoa, Italy
| | | | | | | | | | | | | |
Collapse
|