201
|
Phase 2 Study of Daratumumab in Relapsed/Refractory Mantle-Cell Lymphoma, Diffuse Large B-Cell Lymphoma, and Follicular Lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:275-284. [PMID: 30795996 DOI: 10.1016/j.clml.2018.12.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/19/2018] [Accepted: 12/26/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Daratumumab is a CD38 monoclonal antibody approved for treating relapsed/refractory and newly diagnosed multiple myeloma. Preclinical daratumumab studies demonstrated cytotoxic activity and reduced tumor growth in B-cell non-Hodgkin lymphoma (NHL) subtypes, including diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and mantle-cell lymphoma (MCL). PATIENTS AND METHODS This was a phase 2, open-label, multicenter, 2-stage trial. Patients with relapsed/refractory DLBCL, FL, or MCL with ≥ 50% CD38 expression were eligible for stage 1. Daratumumab (16 mg/kg; 28-day cycles) was administered intravenously weekly for 2 cycles, every 2 weeks for 4 cycles, and every 4 weeks thereafter. Overall response rate was the primary end point. Pharmacokinetic and safety were also evaluated. Stage 2 was planned to further assess daratumumab in larger populations of NHL subtypes if futility criteria were not met. The study was registered with ClinicalTrials.gov (NCT02413489). RESULTS The trial screened 138 patients resulting in accrual of 15 patients with DLBCL, 16 with FL, and 5 with MCL. Median CD38 expression across treated patients was 70%. Overall response rate was 6.7%, 12.5%, and not evaluable in DLBCL, FL, and MCL cohorts, respectively. The most common grade 3/4 treatment-emergent adverse event was thrombocytopenia (11.1%), and 4 (11.1%) patients discontinued treatment because of treatment-emergent adverse events. Infusion-related reactions occurred in 72.2% of patients (3 patients with grade 3; no grade 4). CONCLUSION In NHL, the safety and pharmacokinetics of daratumumab were consistent with myeloma studies. Screen-fail rates were high, prespecified futility thresholds were met in 2 cohorts, and the study was terminated. Studies in other hematologic malignancies and amyloidosis are ongoing.
Collapse
|
202
|
Sepehri B, Ghavami R. Design of new CD38 inhibitors based on CoMFA modelling and molecular docking analysis of 4‑amino-8-quinoline carboxamides and 2,4-diamino-8-quinazoline carboxamides. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2019; 30:21-38. [PMID: 30489181 DOI: 10.1080/1062936x.2018.1545695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/05/2018] [Indexed: 06/09/2023]
Abstract
In this study, based on molecular docking analysis and comparative molecular field analysis (CoMFA) modelling of a series of 71 CD38 inhibitors including 4‑amino-8-quinoline carboxamides and 2,4-diamino-8-quinazoline carboxamides, new CD38 inhibitors were designed. The interactions of the molecules with the greatest and the lowest activities with the nicotinamide mononucleotide (NMN) binding site were investigated by molecular docking analysis. A CoMFA model with four partial least squares regression (PLSR) components was developed to predict the CD38 inhibitory activity of the molecules. The r2 values for the training and test sets were 0.89 and 0.82, respectively. The Q2 values for leave-one-out cross-validation (LOO-CV) and leave-many-out cross-validation (LMO-CV) tests on the training set were 0.65 and 0.64, respectively. The CoMFA model was validated by calculating several statistical parameters. CoMFA contour maps were interpreted, and structural features that influence the CD38 inhibitory activity of molecules were determined. Finally, seven new CD38 inhibitors with greater activity with respect to the greatest active molecules were designed.
Collapse
Affiliation(s)
- B Sepehri
- a Department of Chemistry, Faculty of Science , University of Kurdistan , Sanandaj , Iran
| | - R Ghavami
- a Department of Chemistry, Faculty of Science , University of Kurdistan , Sanandaj , Iran
| |
Collapse
|
203
|
Goldschmidt H, Ashcroft J, Szabo Z, Garderet L. Navigating the treatment landscape in multiple myeloma: which combinations to use and when? Ann Hematol 2019; 98:1-18. [PMID: 30470875 PMCID: PMC6334731 DOI: 10.1007/s00277-018-3546-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 11/06/2018] [Indexed: 12/31/2022]
Abstract
Multiple myeloma is one of the most common hematological malignancies, affecting mainly elderly patients. The treatment landscape for the management of this disease has evolved significantly over the past 15 years, and a vast array of therapeutics is now available, including immunomodulatory drugs, proteasome inhibitors, histone deacetylase inhibitors, and monoclonal antibodies. As a result, deciding which drugs to use and when, and whether these should be used in a particular order or combination, can be challenging. Although combination regimens are often associated with deeper responses and better long-term outcomes than monotherapy, and are becoming the standard of care, they may result in significant incremental toxicity; hence, a sequential approach may be more appropriate for some patients. In particular, treatment choices can vary depending on whether the patient has newly diagnosed multiple myeloma, is eligible for transplant, has relapsed and/or refractory multiple myeloma, or is considered to have high-risk disease. In this review, we discuss factors to be taken into account when making treatment decisions in each of these settings. We also briefly discuss possible therapeutic strategies involving agents that may become available in the future.
Collapse
Affiliation(s)
- Hartmut Goldschmidt
- Internal Medicine V and National Center for Tumor Diseases (NCT), University Clinic Heidelberg, 69120, Heidelberg, Germany.
| | - John Ashcroft
- Department of Haematology, Mid Yorkshire Hospitals NHS Trust, Wakefield, UK
| | - Zsolt Szabo
- Clinical Development, Amgen (Europe) GmbH, Zug, Switzerland
| | - Laurent Garderet
- INSERM, UMR_S 938, Proliferation and Differentiation of Stem Cells, Paris, 75012, France
- AP-HP, Hôpital Saint Antoine, Département d'hématologie et de thérapie cellulaire, Sorbonne Université, Paris 6, Paris, France
| |
Collapse
|
204
|
Cell-based immunotherapy approaches for multiple myeloma. Br J Cancer 2018; 120:38-44. [PMID: 30518815 PMCID: PMC6325139 DOI: 10.1038/s41416-018-0346-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 12/31/2022] Open
Abstract
Despite the arrival of novel therapies, multiple myeloma (MM) remains incurable and new treatment options are needed. Chimeric antigen receptor (CAR) T cells are genetically modified T cells that express a CAR directed against specific tumour antigens. CAR T cells are able to kill target tumour cells and may result in long-lasting immune responses in vivo. The rapid development of CAR technologies has led to clinical trials in haematological cancers including MM, and CAR T cells might evolve into a standard treatment in the next few years. Only small patient cohorts with relapsed or refractory disease have so far been investigated, but promising preliminary results with high response rates have been obtained in phase I clinical trials with B cell maturation antigen (BCMA), CD19, CD38 and κ-light-chain CAR T cells. Additional preclinical studies on CD38 and SLAMF7-CAR T cells in MM treatment yielded preclinical results that merit further investigation. Beyond the T cell approach, recent studies have focussed on CAR natural killer (NK) cells in order to increase the reactivity of these effector cells. Finally, to investigate the targeting of intracellular antigens, cellular therapies based on engineered T cell receptors (TCRs) are in development. In this review, we discuss results from preclinical and early-phase clinical trials testing the feasibility and safety of CAR T cell administration in MM, as well as early studies into approaches that utilise CAR NK cell and genetically modified TCRs.
Collapse
|
205
|
Schütze K, Petry K, Hambach J, Schuster N, Fumey W, Schriewer L, Röckendorf J, Menzel S, Albrecht B, Haag F, Stortelers C, Bannas P, Koch-Nolte F. CD38-Specific Biparatopic Heavy Chain Antibodies Display Potent Complement-Dependent Cytotoxicity Against Multiple Myeloma Cells. Front Immunol 2018; 9:2553. [PMID: 30524421 PMCID: PMC6262402 DOI: 10.3389/fimmu.2018.02553] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
CD38 is overexpressed by multiple myeloma cells and has emerged as a target for therapeutic antibodies. Nanobodies are soluble single domain antibody fragments derived from the VHH variable domain of heavy chain antibodies naturally occurring in camelids. We previously identified distinct llama nanobodies that recognize three non-overlapping epitopes of the extracellular domain of CD38. Here, we fused these VHH domains to the hinge, CH2, and CH3 domains of human IgG1, yielding highly soluble chimeric llama/human heavy chain antibodies (hcAbs). We analyzed the capacity of these hcAbs to mediate complement-dependent cytotoxicity (CDC) to CD38-expressing human multiple myeloma and Burkitt lymphoma cell lines. Combinations of two hcAbs that recognize distinct, non-overlapping epitopes of CD38 mediated potent CDC, in contrast to the hcAb monotherapy with only weak CDC capacity. Similarly, combining daratumumab with a hcAb that recognizes a non-overlapping epitope resulted in dramatically enhanced CDC. Further, introducing the E345R HexaBody mutation into the CH3 domain strongly enhanced the CDC potency of hcAbs to CD38-expressing cells. Exploiting their high solubility, we genetically fused two distinct nanobodies into heteromeric dimers via a flexible peptide linker and then fused these nanobody dimers to the hinge, CH2 and CH3 domains of human IgG1, yielding highly soluble, biparatopic hcAbs. These biparatopic hcAbs elicited CDC toward CD38-expressing myeloma cells more effectively than daratumumab. Our results underscore the advantage of nanobodies vs. pairs of VH and VL domains for constructing bispecific antibodies. Moreover, the CD38-specific biparatopic heavy chain antibodies described here represent potential new powerful therapeutics for treatment of multiple myeloma.
Collapse
Affiliation(s)
- Kerstin Schütze
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Petry
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Hambach
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Niklas Schuster
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - William Fumey
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Levin Schriewer
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana Röckendorf
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Birte Albrecht
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Peter Bannas
- Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
206
|
Lancman G, Arinsburg S, Jhang J, Cho HJ, Jagannath S, Madduri D, Parekh S, Richter J, Chari A. Blood Transfusion Management for Patients Treated With Anti-CD38 Monoclonal Antibodies. Front Immunol 2018; 9:2616. [PMID: 30498492 PMCID: PMC6249335 DOI: 10.3389/fimmu.2018.02616] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/24/2018] [Indexed: 12/31/2022] Open
Abstract
Daratumumab has proven to be highly efficacious for relapsed and refractory multiple myeloma (MM) and has recently been approved in the frontline setting for MM patients ineligible for transplantation. In the future, expanded indications are possible for daratumumab and other anti-CD38 monoclonal antibodies in development. For several years, it has been recognized that these therapies interfere with blood bank testing by binding to CD38 on red blood cells and causing panagglutination on the Indirect Antiglobulin Test. This can lead to redundant testing and significant delays in patient care. Given the anticipated increase in utilization of anti-CD38 monoclonal antibodies, as well as the transfusion needs of MM patients, it is critical to understand the nature of this interference with blood bank testing and to optimize clinical and laboratory procedures. In this review, we summarize the pathophysiology of this phenomenon, examine the clinical data reported to date, describe currently available methods to resolve this issue, and lastly provide a guide to clinical management of blood transfusions for patients receiving anti-CD38 monoclonal antibodies.
Collapse
Affiliation(s)
- Guido Lancman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Suzanne Arinsburg
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jeffrey Jhang
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Hearn Jay Cho
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sundar Jagannath
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Deepu Madduri
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Samir Parekh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Joshua Richter
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ajai Chari
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
207
|
Laubach JP, van de Donk N, Davies FE, Mikhael J. Practical Considerations for Antibodies in Myeloma. Am Soc Clin Oncol Educ Book 2018; 38:667-674. [PMID: 30231321 DOI: 10.1200/edbk_205443] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The development of the monoclonal antibodies daratumumab and elotuzumab has expanded treatment options for multiple myeloma and led to great improvement in patient outcomes. These agents have favorable safety profiles and synergize effectively with established agents used in the management of myeloma, namely immunomodulatory drugs and proteasome inhibitors. This article reviews the rationale for use of monoclonal antibodies in myeloma, current approved indications for daratumumab and elotuzumab, the manner in which these agents are used in the overall management of myeloma, and specific challenges associated with their use in the clinic. It also highlights other, emerging drug combinations that incorporate daratumumab or elotuzumab and profiles new therapeutic antibodies currently under development.
Collapse
Affiliation(s)
- Jacob P Laubach
- From the Department of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA; Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands; Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR; International Myeloma Foundation, North Hollywood, CA
| | - Niels van de Donk
- From the Department of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA; Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands; Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR; International Myeloma Foundation, North Hollywood, CA
| | - Faith E Davies
- From the Department of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA; Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands; Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR; International Myeloma Foundation, North Hollywood, CA
| | - Joseph Mikhael
- From the Department of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA; Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands; Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR; International Myeloma Foundation, North Hollywood, CA
| |
Collapse
|
208
|
Xu XS, Dimopoulos MA, Sonneveld P, Ho PJ, Belch A, Leiba M, Capra M, Gomez D, Medvedova E, Iida S, Min CK, Schecter J, Jansson R, Zhang L, Sun YN, Clemens PL. Pharmacokinetics and Exposure-Response Analyses of Daratumumab in Combination Therapy Regimens for Patients with Multiple Myeloma. Adv Ther 2018; 35:1859-1872. [PMID: 30374808 PMCID: PMC6223994 DOI: 10.1007/s12325-018-0815-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Daratumumab, a human IgG monoclonal antibody targeting CD38, has demonstrated activity as monotherapy and in combination with standard-of-care regimens in multiple myeloma. Population pharmacokinetic analyses were conducted to determine the pharmacokinetics of intravenous daratumumab in combination therapy versus monotherapy, evaluate the effect of patient- and disease-related covariates on drug disposition, and examine the relationships between daratumumab exposure and efficacy/safety outcomes. METHODS Four clinical studies of daratumumab in combination with lenalidomide/dexamethasone (POLLUX and GEN503); bortezomib/dexamethasone (CASTOR); pomalidomide/dexamethasone, bortezomib/thalidomide/dexamethasone, and bortezomib/melphalan/prednisone (EQUULEUS) were included in the analysis. Using various dosing schedules, the majority of patients (684/694) received daratumumab at a dose of 16 mg/kg. In GEN503, daratumumab was administered at a dose of 2 mg/kg (n = 3), 4 mg/kg (n = 3), 8 mg/kg (n = 4), and 16 mg/kg (n = 34). A total of 650 patients in EQUULEUS (n = 128), POLLUX (n = 282), and CASTOR (n = 240) received daratumumab 16 mg/kg. The exposure-efficacy and exposure-safety relationships examined progression-free survival (PFS) and selected adverse events (infusion-related reactions; thrombocytopenia, anemia, neutropenia, lymphopenia, and infections), respectively. RESULTS Pharmacokinetic profiles of daratumumab were similar between monotherapy and combination therapy. Covariate analysis identified no clinically important effects on daratumumab exposure, and no dose adjustments were recommended on the basis of these factors. Maximal clinical benefit on PFS was achieved for the majority of patients (approximately 75%) at the 16 mg/kg dose. No apparent relationship was observed between daratumumab exposure and selected adverse events. CONCLUSION These data support the recommended 16 mg/kg dose of daratumumab and the respective dosing schedules in the POLLUX and CASTOR pivotal studies. FUNDING Janssen Research & Development.
Collapse
Affiliation(s)
- Xu Steven Xu
- Janssen Research & Development, LLC, Raritan, NJ, USA.
| | | | - Pieter Sonneveld
- Department of Hematology, Erasmus MC, Rotterdam, The Netherlands
| | - P Joy Ho
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | | | - Merav Leiba
- Assuta Ashdod University Hospital, Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheba, Israel
| | - Marcelo Capra
- Instituto do Cancer, Hospital Mãe de Deus, Porto Alegre, Brazil
| | - David Gomez
- Hospital Universitario de la UANL, Monterrey, Nuevo Leon, Mexico
| | - Eva Medvedova
- Oregon Health & Science University, Portland, OR, USA
| | | | | | | | | | - Liping Zhang
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Yu-Nien Sun
- Janssen Research & Development, LLC, Spring House, PA, USA
| | | |
Collapse
|
209
|
Zheleznyak A, Shokeen M, Achilefu S. Nanotherapeutics for multiple myeloma. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 10:e1526. [PMID: 29701006 PMCID: PMC6185771 DOI: 10.1002/wnan.1526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/19/2018] [Accepted: 03/23/2018] [Indexed: 01/11/2023]
Abstract
Multiple myeloma (MM) is an age-related hematological malignancy with an estimated 30,000 new cases and 13,000 deaths per year. A disease of antibody-secreting malignant plasma B-cells that grow primarily in the bone marrow (BM), MM causes debilitating fractures, anemia, renal failure, and hypercalcemia. In addition to the abnormal genetic profile of MM cells, the permissive BM microenvironment (BMM) supports MM pathogenesis. Although advances in treatment options have significantly enhanced survival in MM patients, transient perfusion of small-molecule drugs in the BM does not provide sufficient residence to enhance MM cell-drug interaction, thus allowing some myeloma cells to escape the first line of treatment. As such, there remains a crucial need to develop advanced drug delivery systems that can navigate the complex BMM and effectively reach the myeloma cells. The high vascular density and spongy nature of bone structure suggest that nanoparticles (NPs) can serve as smart drug-delivery systems capable of extravasation and retention in various BM compartments to exert a durable therapeutic effect. In this focus article, we first summarize the pathophysiology of MM, emphasizing how the BM niche presents serious challenges for effective treatment of MM with small-molecule drugs. We then pivot to current efforts to develop NP-based drug carriers and intrinsically therapeutic nanotherapeutics. The article concludes with a brief perspective on the opportunities and challenges in developing and translating nanotherapeutics to improve the treatment outcomes of MM patients. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | - Monica Shokeen
- Departments of Radiology, and Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Samuel Achilefu
- Departments of Radiology, Biomedical Engineering, and Biochemistry & Molecular Biophysics, Washington University, St. Louis, MO, USA
| |
Collapse
|
210
|
Bu X, Kato J, Hong JA, Merino MJ, Schrump DS, Lund FE, Moss J. CD38 knockout suppresses tumorigenesis in mice and clonogenic growth of human lung cancer cells. Carcinogenesis 2018; 39:242-251. [PMID: 29228209 DOI: 10.1093/carcin/bgx137] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 11/30/2017] [Indexed: 12/17/2022] Open
Abstract
The ectodomain of the plasma membrane ectoenzyme CD38 functions as both an NAD glycohydrolase and an ADP-ribosyl cyclase by catalyzing, respectively, the conversion of NAD to nicotinamide and ADP-ribose or cyclic ADP-ribose. CD38 is attracting particular attention in cancer therapy. An anti-CD38 monoclonal antibody (daratumumab) was approved for treatment of patients with multiple myeloma. However, the role of CD38 in non-hematological malignancies has not been explored. Previously, we reported that ADP-ribose-acceptor hydrolase (ARH)-1 deficiency in mice was associated with tumor development. In the present study, we found that in wild-type and ARH1-deficient mice deletion of the CD38 gene reduced tumor formation. Significant reductions in tumor number were observed in lymphomas, adenocarcinomas and hemangio/histolytic sarcomas. Consistent with a role for CD38 in tumorigenesis, CRISPR/Cas9-based knockout of CD38 in A549 human adenocarcinoma cells inhibited anchorage-independent cell growth, cell invasion and xenograft growth in nude mice. CD38 mRNA and protein expression were evaluated in human lung cancer cell lines and in human lung cancer specimens. CD38 overexpression in tumor cells was identified in 11 of 27 patient samples. In addition, some human lung cancer cell lines had dramatically higher CD38 mRNA and protein expression than normal cells. Consistent with these observations, search of the Oncomine database showed that some human lung adenocarcinomas had higher CD38 mRNA levels compared to normal lung tissues. In total, our data are consistent with the conclusion that CD38 plays a role in murine and human lung tumorigenesis and that anti-CD38 treatment may have therapeutic potential in lung cancer.
Collapse
Affiliation(s)
- Xiangning Bu
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jiro Kato
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Julie A Hong
- Thoracic Epigenetics Section, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Maria J Merino
- Translational Surgical Pathology, National Cancer Institute, Bethesda, MD, USA
| | - David S Schrump
- Thoracic Epigenetics Section, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Frances E Lund
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | |
Collapse
|
211
|
Ma X, Wong SW, Zhou P, Chaulagain CP, Doshi P, Klein AK, Sprague K, Kugelmass A, Toskic D, Warner M, Miller KB, Lee L, Varga C, Comenzo RL. Daratumumab binds to mobilized CD34+ cells of myeloma patients in vitro without cytotoxicity or impaired progenitor cell growth. Exp Hematol Oncol 2018; 7:27. [PMID: 30356940 PMCID: PMC6192105 DOI: 10.1186/s40164-018-0119-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/06/2018] [Indexed: 02/08/2023] Open
Abstract
Background The monoclonal antibody daratumumab, approved for treating myeloma, targets CD38, a protein on myeloma and also on CD34+ hematopoietic progenitor cells. Because mobilized CD34+ cells are critical for stem cell transplant, we investigated the in vitro activity of daratumumab on mobilized CD34+ cells from myeloma patients with no prior exposure to daratumumab. Methods We determined the number of CD38 molecules per CD34+ cell, and whether daratumumab bound to CD34+ cells, whether C1q bound to daratumumab-coated CD34+ cells and whether daratumumab-related complement-dependent cytotoxicity (CDC) occurred. We also examined CD34+ cell progenitor cell colony capacity in assays with pre-plating incubation of CD34+ cells with daratumumab alone or with daratumumab and the CD59 inhibitory antibody BRIC229, and also assessed CD34+ cell responses to increasing doses of daratumumab in caspase 3/7 activity assays. Results Although 75% of mobilized CD34+ cells co-express CD38, CD38 was minimally present on CD34+ cells compared to Daudi and KG-1 controls, C1q did not bind to daratumumab-coated CD34+ cells, and CDC did not occur. CD34+ cells incubated in complement-rich human serum with daratumumab alone or with daratumumab and BRIC229, and then plated in progenitor cell assays, produced similar numbers of colonies as controls. In progenitor cell assays with cryopreserved or fresh unselected or CD34-selected cells, daratumumab did not affect progenitor cell capacity, and in caspase 3/7 activity assays CD34+ cells were not affected by increasing doses of daratumumab. Conclusion In vitro, daratumumab is not toxic to mobilized CD34+ progenitor cells from myeloma patients.
Collapse
Affiliation(s)
- Xun Ma
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Sandy W Wong
- 2Division of Hematology and Blood and Marrow Transplantation, Department of Medicine, University of California, San Francisco, CA USA
| | - Ping Zhou
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Chakra P Chaulagain
- Taussig Cancer Institute of Cleveland Clinic, Maroone Cancer Center, Weston, FL USA
| | - Parul Doshi
- 4Janssen Research & Development, Spring House, PA USA
| | - Andreas K Klein
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Kellie Sprague
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Adin Kugelmass
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Denis Toskic
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Melissa Warner
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Kenneth B Miller
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Lisa Lee
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Cindy Varga
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| | - Raymond L Comenzo
- 1The John C Davis Myeloma and Amyloid Program in the Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Box 826, Boston, MA 02111 USA
| |
Collapse
|
212
|
van de Donk NW, Usmani SZ. CD38 Antibodies in Multiple Myeloma: Mechanisms of Action and Modes of Resistance. Front Immunol 2018; 9:2134. [PMID: 30294326 PMCID: PMC6158369 DOI: 10.3389/fimmu.2018.02134] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 08/29/2018] [Indexed: 11/30/2022] Open
Abstract
MM cells express high levels of CD38, while CD38 is expressed at relatively low levels on normal lymphoid and myeloid cells, and in some non-hematopoietic tissues. This expression profile, together with the role of CD38 in adhesion and as ectoenzyme, resulted in the development of CD38 antibodies for the treatment of multiple myeloma (MM). At this moment several CD38 antibodies are at different phases of clinical testing, with daratumumab already approved for various indications both as monotherapy and in combination with standards of care in MM. CD38 antibodies have Fc-dependent immune effector mechanisms, such as complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP). Inhibition of ectoenzymatic function and direct apoptosis induction may also contribute to the efficacy of the antibodies to kill MM cells. The CD38 antibodies also improve host-anti-tumor immunity by the elimination of regulatory T cells, regulatory B cells, and myeloid-derived suppressor cells. Mechanisms of primary and/or acquired resistance include tumor-related factors, such as reduced cell surface expression levels of the target antigen and high levels of complement inhibitors (CD55 and CD59). Differences in frequency or activity of effector cells may also contribute to differences in outcome. Furthermore, the microenvironment protects MM cells to CD38 antibody-induced ADCC by upregulation of anti-apoptotic molecules, such as survivin. Improved understanding of modes of action and mechanisms of resistance has resulted in rationally designed CD38-based combination therapies, which will contribute to further improvement in outcome of MM patients.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/antagonists & inhibitors
- ADP-ribosyl Cyclase 1/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antibody-Dependent Cell Cytotoxicity/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Apoptosis/drug effects
- Apoptosis/immunology
- B-Lymphocytes, Regulatory/drug effects
- B-Lymphocytes, Regulatory/immunology
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immunoglobulin Fc Fragments/immunology
- Immunoglobulin Fc Fragments/metabolism
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/immunology
- Multiple Myeloma/drug therapy
- Multiple Myeloma/immunology
- Multiple Myeloma/pathology
- Myeloid-Derived Suppressor Cells/drug effects
- Myeloid-Derived Suppressor Cells/immunology
- Phagocytosis/drug effects
- Phagocytosis/immunology
- Randomized Controlled Trials as Topic
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Treatment Outcome
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
| | - Saad Z. Usmani
- Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC, United States
| |
Collapse
|
213
|
Spencer A, Lentzsch S, Weisel K, Avet-Loiseau H, Mark TM, Spicka I, Masszi T, Lauri B, Levin MD, Bosi A, Hungria V, Cavo M, Lee JJ, Nooka AK, Quach H, Lee C, Barreto W, Corradini P, Min CK, Scott EC, Chanan-Khan AA, Horvath N, Capra M, Beksac M, Ovilla R, Jo JC, Shin HJ, Sonneveld P, Soong D, Casneuf T, Chiu C, Amin H, Qi M, Thiyagarajah P, Sasser AK, Schecter JM, Mateos MV. Daratumumab plus bortezomib and dexamethasone versus bortezomib and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of CASTOR. Haematologica 2018; 103:2079-2087. [PMID: 30237264 PMCID: PMC6269293 DOI: 10.3324/haematol.2018.194118] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/17/2018] [Indexed: 11/11/2022] Open
Abstract
Daratumumab, a CD38 human monoclonal antibody, demonstrated significant clinical activity in combination with bortezomib and dexamethasone versus bortezomib and dexamethasone alone in the primary analysis of CASTOR, a phase 3 study in relapsed and/or refractory multiple myeloma. A post hoc analysis based on treatment history and longer follow up is presented. After 19.4 (range: 0–27.7) months of median follow up, daratumumab plus bortezomib and dexamethasone prolonged progression-free survival (median: 16.7 versus 7.1 months; hazard ratio, 0.31; 95% confidence interval, 0.24-0.39; P<0.0001) and improved the overall response rate (83.8% versus 63.2%; P<0.0001) compared with bortezomib and dexamethasone alone. The progression-free survival benefit of daratumumab plus bortezomib and dexamethasone was most apparent in patients with 1 prior line of therapy (median: not reached versus 7.9 months; hazard ratio, 0.19; 95% confidence interval, 0.12-0.29; P<0.0001). Daratumumab plus bortezomib and dexamethasone was also superior to bortezomib and dexamethasone alone in subgroups based on prior treatment exposure (bortezomib, thalidomide, or lenalidomide), lenalidomide-refractory status, time since last therapy (≤12, >12, ≤6, or >6 months), or cytogenetic risk. Minimal residual disease–negative rates were >2.5-fold higher with daratumumab across subgroups. The safety profile of daratumumab plus bortezomib and dexamethasone remained consistent with longer follow up. Daratumumab plus bortezomib and dexamethasone demonstrated significant clinical activity across clinically relevant subgroups and provided the greatest benefit to patients treated at first relapse. Trial registration: clinicaltrials.gov identifier: 02136134.
Collapse
Affiliation(s)
- Andrew Spencer
- Malignant Haematology and Stem Cell Transplantation Service, Alfred Health-Monash University, Melbourne, Australia
| | - Suzanne Lentzsch
- Division of Hematology/Oncology, Columbia University, New York, NY, USA
| | - Katja Weisel
- Universitaetsklinikum Tuebingen der Eberhard-Karls-Universitaet, Abteilung fuer Innere Medizin II, Tübingen, Germany
| | | | - Tomer M Mark
- Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Ivan Spicka
- Clinical Department of Haematology, 1 Medical Department, Charles University in Prague, Czech Republic
| | - Tamas Masszi
- Department of Haematology and Stem Cell Transplantation, St László Hospital, 3 Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Birgitta Lauri
- Department of Hematology, Sunderbyn Hospital, Luleå, Sweden
| | - Mark-David Levin
- Albert Schweitzer Hospital Department of Internal Medicine, Dordrecht, the Netherlands
| | - Alberto Bosi
- Department of Hematology, Careggi Hospital and University of Florence, Italy
| | - Vania Hungria
- Irmandade Da Santa Casa De Misericordia De São Paulo, Brazil
| | - Michele Cavo
- "Seràgnoli" Institute of Hematology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Je-Jung Lee
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Jeollanamdo, South Korea
| | - Ajay K Nooka
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Hang Quach
- St. Vincent's Hospital, University of Melbourne, Australia
| | - Cindy Lee
- Royal Adelaide Hospital, North Terrace, Australia
| | | | - Paolo Corradini
- Fondazione IRCCS Instituto Nazionale dei Tumori, University of Milan, Italy
| | | | - Emma C Scott
- Oregon Health & Science University, Portland, OR, USA
| | | | | | - Marcelo Capra
- Instituto do Cancer-Hospital Mae de Deus, Porto Alegre, Brazil
| | | | - Roberto Ovilla
- Hospital Angeles Lomas, Naucalpan de Juárez y alrededores, México
| | | | - Ho-Jin Shin
- Division of Hematology-Oncology, Department of Internal Medicine, School of Medicine, Medical Research Institute, Pusan National University Hospital, Busan, South Korea
| | | | - David Soong
- Janssen Research & Development, LLC, Spring House, PA, USA
| | | | | | - Himal Amin
- Janssen Research & Development, LLC, Raritan, NJ, USA
| | - Ming Qi
- Janssen Research & Development, LLC, Spring House, PA, USA
| | | | | | | | | |
Collapse
|
214
|
Amici SA, Young NA, Narvaez-Miranda J, Jablonski KA, Arcos J, Rosas L, Papenfuss TL, Torrelles JB, Jarjour WN, Guerau-de-Arellano M. CD38 Is Robustly Induced in Human Macrophages and Monocytes in Inflammatory Conditions. Front Immunol 2018; 9:1593. [PMID: 30042766 PMCID: PMC6048227 DOI: 10.3389/fimmu.2018.01593] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 06/27/2018] [Indexed: 11/17/2022] Open
Abstract
Macrophages and their monocyte precursors mediate innate immune responses and can promote a spectrum of phenotypes from pro-inflammatory to pro-resolving. Currently, there are few markers that allow for robust dissection of macrophage phenotype. We recently identified CD38 as a marker of inflammatory macrophages in murine in vitro and in vivo models. However, it is unknown whether CD38 plays a similar marker and/or functional role in human macrophages and inflammatory diseases. Here, we establish that CD38 transcript and protein are robustly induced in human macrophages exposed to LPS (±IFN-γ) inflammatory stimuli, but not with the alternative stimulus, IL-4. Pharmacologic and/or genetic CD38 loss-of-function significantly reduced the secretion of inflammatory cytokines IL-6 and IL-12p40 and glycolytic activity in human primary macrophages. Finally, monocyte analyses in systemic lupus erythematosus patients revealed that, while all monocytes express CD38, high CD38 expression in the non-classical monocyte subpopulation is associated with disease. These data are consistent with an inflammatory marker role for CD38 in human macrophages and monocytes.
Collapse
Affiliation(s)
- Stephanie A Amici
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Nicholas A Young
- Division of Rheumatology and Immunology, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Janiret Narvaez-Miranda
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Kyle A Jablonski
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Jesus Arcos
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Lucia Rosas
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Tracey L Papenfuss
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Wael N Jarjour
- Division of Rheumatology and Immunology, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Mireia Guerau-de-Arellano
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States.,Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
215
|
Mele S, Devereux S, Pepper AG, Infante E, Ridley AJ. Calcium-RasGRP2-Rap1 signaling mediates CD38-induced migration of chronic lymphocytic leukemia cells. Blood Adv 2018; 2:1551-1561. [PMID: 29970392 PMCID: PMC6039665 DOI: 10.1182/bloodadvances.2017014506] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/24/2018] [Indexed: 01/08/2023] Open
Abstract
CD38 is a transmembrane exoenzyme that is associated with poor prognosis in chronic lymphocytic leukemia (CLL). High CD38 levels in CLL cells are linked to increased cell migration, but the molecular basis is unknown. CD38 produces nicotinic acid adenine dinucleotide phosphate and adenosine 5'-diphosphate-ribose, both of which can act to increase intracellular Ca2+ levels. Here we show that CD38 expression increases basal intracellular Ca2+ levels and stimulates CLL cell migration both with and without chemokine stimulation. We find that CD38 acts via intracellular Ca2+ to increase the activity of the Ras family GTPase Rap1, which is in turn regulated by the Ca2+-sensitive Rap1 guanine-nucleotide exchange factor RasGRP2. Both Rap1 and RasGRP2 are required for CLL cell migration, and RasGRP2 is polarized in primary CLL cells with high CD38 levels. These results indicate that CD38 promotes RasGRP2/Rap1-mediated CLL cell adhesion and migration by increasing intracellular Ca2+ levels.
Collapse
Affiliation(s)
- Silvia Mele
- Randall Centre for Cell and Molecular Biophysics, and
- School of Cancer Sciences, King's College London, London, United Kingdom
| | - Stephen Devereux
- School of Cancer Sciences, King's College London, London, United Kingdom
| | - Andrea G Pepper
- School of Cancer Sciences, King's College London, London, United Kingdom
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom; and
| | | | - Anne J Ridley
- Randall Centre for Cell and Molecular Biophysics, and
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
216
|
Tang F, Malek E, Math S, Schmotzer CL, Beck RC. Interference of Therapeutic Monoclonal Antibodies With Routine Serum Protein Electrophoresis and Immunofixation in Patients With Myeloma: Frequency and Duration of Detection of Daratumumab and Elotuzumab. Am J Clin Pathol 2018; 150:121-129. [PMID: 29901687 DOI: 10.1093/ajcp/aqy037] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES We investigated the frequency and pattern of detection of therapeutic monoclonal antibodies (t-mAbs) daratumumab and elotuzumab by routine serum protein electrophoresis (SPE) and immunofixation (IF) in treated patients with myeloma. METHODS Detection of t-mAb was assessed in 22 patients by retrospective review of SPE/IF ordered prior to, during, and after 26 individual courses of therapy. RESULTS t-mAb was distinguishable from M-protein in 16 of 26 courses, with daratumumab detected in nine of nine and elotuzumab in six of seven patients. t-mAb was detected on first follow-up SPE/IF in 12 patients, with earliest detection 7 days after therapy initiation and latest detection 70 days after therapy. t-mAb persisted throughout induction therapy in most patients, with loss of detection during maintenance daratumumab. CONCLUSIONS When distinguishable from M-protein, t-mAbs are detectable in 93% of treated patients as soon as 7 days after the initial dose and are consistently observed throughout induction therapy, warranting increased monitoring and careful interpretation of SPE/IF.
Collapse
Affiliation(s)
- Felicia Tang
- School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Ehsan Malek
- School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Hematology/Oncology, University Hospitals of Cleveland Medical Center, Cleveland, OH
| | - Susan Math
- Department of Pathology, University Hospitals of Cleveland Medical Center, Cleveland, OH
| | - Christine L Schmotzer
- School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Pathology, University Hospitals of Cleveland Medical Center, Cleveland, OH
| | - Rose C Beck
- School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Pathology, University Hospitals of Cleveland Medical Center, Cleveland, OH
| |
Collapse
|
217
|
Drgona L, Gudiol C, Lanini S, Salzberger B, Ippolito G, Mikulska M. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Agents targeting lymphoid or myeloid cells surface antigens [II]: CD22, CD30, CD33, CD38, CD40, SLAMF-7 and CCR4). Clin Microbiol Infect 2018; 24 Suppl 2:S83-S94. [DOI: 10.1016/j.cmi.2018.03.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/05/2018] [Accepted: 02/11/2018] [Indexed: 01/12/2023]
|
218
|
Roepcke S, Plock N, Yuan J, Fedyk ER, Lahu G, Zhao L, Smithson G. Pharmacokinetics and pharmacodynamics of the cytolytic anti-CD38 human monoclonal antibody TAK-079 in monkey - model assisted preparation for the first in human trial. Pharmacol Res Perspect 2018; 6:e00402. [PMID: 29864242 PMCID: PMC5980133 DOI: 10.1002/prp2.402] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/10/2018] [Indexed: 02/06/2023] Open
Abstract
We are studying the fully human, IgG1λ cytolytic monoclonal antibody TAK-079, which binds CD38. CD38 is expressed on plasma and natural killer (NK) cells constitutively and upregulated on subsets of B and T lymphocytes upon activation. TAK-079 cross-reacts with CD38 expressed by cynomolgus monkeys and depletes subsets of NK, B, and T cells. Therefore, safety and function of TAK-079 was evaluated in this species, prior to clinical development, using bioanalytical, and flow cytometry assays. We pooled the data from eight studies in healthy monkeys (dose range 0.03-100 mg/kg) and developed mathematical models that describe the pharmacokinetics and the exposure-effect relationship for NK cells, B cells, and T cells. NK cell depletion was identified as the most sensitive pharmacodynamic effect of TAK-079. It was adequately described with a turnover model (C50 = 27.5 μg/mL on depletion rate) and complete depletion was achieved with an IV dose of 0.3 mg/kg. Intermediate effects on T-cell counts were described with a direct response model (C50 = 11.9 μg/mL) and on B-cell counts with a 4-transit-compartment model (C50 = 19.8 μg/mL on depletion rate). Our analyses substantiate the observation that NK, B and T cells are cleared by TAK-079 at different rates and required different time spans to replete the blood compartment. The models were used to simulate pharmacokinetic and cell depletion profiles in humans after applying a straightforward scaling approach for monoclonal antibodies in preparation for the first-in-human clinical trial.
Collapse
Affiliation(s)
- Stefan Roepcke
- Takeda Pharmaceuticals International AG, Zurich, Switzerland
| | - Nele Plock
- Takeda Pharmaceuticals International AG, Zurich, Switzerland
| | - Josh Yuan
- Takeda Pharmaceuticals International, Cambridge, Massachusetts
| | - Eric R Fedyk
- Takeda Pharmaceuticals International, Cambridge, Massachusetts
| | - Gezim Lahu
- Takeda Pharmaceuticals International AG, Zurich, Switzerland
| | - Lin Zhao
- Takeda Pharmaceuticals International, Cambridge, Massachusetts
| | | |
Collapse
|
219
|
Suzuki K, Dimopoulos MA, Takezako N, Okamoto S, Shinagawa A, Matsumoto M, Kosugi H, Yoon SS, Huang SY, Qin X, Qi M, Iida S. Daratumumab, lenalidomide, and dexamethasone in East Asian patients with relapsed or refractory multiple myeloma: subgroup analyses of the phase 3 POLLUX study. Blood Cancer J 2018; 8:41. [PMID: 29712896 PMCID: PMC5928154 DOI: 10.1038/s41408-018-0071-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/08/2018] [Accepted: 02/08/2018] [Indexed: 12/14/2022] Open
Abstract
In the phase 3 POLLUX study, daratumumab plus lenalidomide and dexamethasone (DRd) significantly reduced the risk of progression/death and induced deeper responses vs. lenalidomide and dexamethasone alone (Rd) in patients with relapsed/refractory multiple myeloma (RRMM). We report a subgroup analysis of East Asian (Japanese, Korean, and Taiwanese) patients from POLLUX based on a longer follow-up of 24.7 months. Median progression-free survival was not reached (NR) for DRd vs. 13.8 months for Rd (hazard ratio [HR], 0.42; 95% confidence interval [CI], 0.23–0.76), and overall response rates were higher for DRd vs. Rd (90.2 vs. 72.1%). DRd extended the median duration of response vs. Rd (NR vs. 20.2 months), and minimal residual disease–negative rates at the 10–5 sensitivity threshold were 21.2 vs. 9.1% for DRd vs. Rd. No new safety signals were observed. Similar efficacy and safety were observed in the smaller subgroup of Japanese patients treated with DRd vs. Rd. These results demonstrate favorable efficacy and safety of DRd vs. Rd in East Asian patients and also in the Japanese-only patient subgroup that are consistent with findings in the overall patient population of POLLUX.
Collapse
Affiliation(s)
- Kenshi Suzuki
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | | | - Naoki Takezako
- Department of Hematology, National Hospital Organization Disaster Medical Center of Japan, Tachikawa, Japan
| | | | | | - Morio Matsumoto
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Shibukawa, Japan
| | - Hiroshi Kosugi
- Department of Hematology, Ogaki Municipal Hospital, Ogaki, Japan
| | | | | | - Xiang Qin
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Ming Qi
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Shinsuke Iida
- Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| |
Collapse
|
220
|
Bolzoni M, Toscani D, Costa F, Vicario E, Aversa F, Giuliani N. The link between bone microenvironment and immune cells in multiple myeloma: Emerging role of CD38. Immunol Lett 2018; 205:65-70. [PMID: 29702149 DOI: 10.1016/j.imlet.2018.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 04/23/2018] [Indexed: 12/30/2022]
Abstract
The relationship between bone and immune cells is well established both in physiological and pathological conditions. Multiple myeloma (MM) is a plasma cell malignancy characterized by an increase of number and activity of osteoclasts (OCLs) and a decrease of osteoblasts (OBs). These events are responsible for bone lesions of MM patients. OCLs support MM cells survival in vitro and in vivo. Recently, the possible role of OCLs as immunosuppressive cells in the MM BM microenvironment has been underlined. OCLs protect MM cells against T cell-mediated cytotoxicity through the expression of several molecules including programmed death-ligand (PD-L) 1, galectin (Gal) 9, CD200, and indoleamine-2,3-dioxygenase (IDO). Among the molecules that could be involved in the link between immune-microenvironment and osteoclastogenesis the role of CD38 has been hypothesized. CD38 is a well-known adhesion molecule and an ectoenzyme highly expressed by MM cells. Moreover, CD38 is expressed by OCLs and at the surface level on OCL precursors. Targeting CD38 with monoclonal antibodies showed inhibition of both osteoclastogenesis and OCL-mediated suppression of T cell function. This review elucidates this evidence indicating that osteoclastogenesis affect MM immune-microenvironment being a potential target to improve anti-MM immunity and to ameliorate bone disease.
Collapse
Affiliation(s)
- Marina Bolzoni
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Denise Toscani
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Federica Costa
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Emanuela Vicario
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy; Biopathology and Medical Biotechnologies, Biology and Genetic Section, University of Palermo, 90133 Palermo, Italy
| | - Franco Aversa
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy; Hematology and BMT Center, "Azienda Ospedaliero-Universitaria di Parma", 43126 Parma, Italy
| | - Nicola Giuliani
- Department Medicine and Surgery, University of Parma, 43126 Parma, Italy; Hematology and BMT Center, "Azienda Ospedaliero-Universitaria di Parma", 43126 Parma, Italy.
| |
Collapse
|
221
|
Kriegsmann K, Dittrich T, Neuber B, Awwad MHS, Hegenbart U, Goldschmidt H, Hillengass J, Hose D, Seckinger A, Müller-Tidow C, Ho AD, Schönland S, Hundemer M. Quantification of number of CD38 sites on bone marrow plasma cells in patients with light chain amyloidosis and smoldering multiple myeloma. CYTOMETRY PART B-CLINICAL CYTOMETRY 2018; 94:611-620. [PMID: 29577600 DOI: 10.1002/cyto.b.21636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 02/06/2018] [Accepted: 03/21/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Recent approaches in multiple myeloma (MM) treatment have targeted CD38. As antigen expression levels on plasma cells (PCs) were demonstrated to affect response to monoclonal antibody (mAb) treatment, a precise characterization of PC phenotype is warranted. METHODS Anti-CD38 mAb (isatuximab) was tested for antibody-dependent cellular cytotoxicity (ADCC) in MM cell lines. Quantification of the number of sites (NOS) of CD38 on bone marrow PCs and other immune cells obtained from light chain (AL) amyloidosis (n = 46) and smoldering multiple myeloma (SMM) patients (n = 19) was performed with two different quantitative flow cytometry (QFCM) applications. RESULTS ADCC activity of isatuximab was observed in cell lines with >100 × 103 CD38-NOS only. The average PC CD38-NOS was 153 ± 53 × 103 in AL amyloidosis and 138.7 ± 53 × 103 in SMM patients. Eight (17%) AL amyloidosis and 4 (21%) SMM patients showed a PC CD38-NOS level <100 × 103 . In four AL amyloidosis and two SMM patients <10% of PCs had a CD38-NOS ≥100 × 103 . The CD38-NOS identified on bone marrow lymphocytes, monocytes, and granulocytes was two log units below the CD38-NOS on PCs (P < 0.001). No significant differences in CD38-NOS expression levels on any of the analyzed PC subpopulations in AL amyloidosis and SMM patients were identified. CONCLUSION Levels of CD38 expression affect the isatuximab-mediated ADCC in vitro. As PCs of patients with AL amyloidosis and SMM do not homogenously express high CD38 our data provide a rationale for assessment of CD38-NOS in patients with PC disorders prior to anti-CD38 treatment. © 2018 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| | - Tobias Dittrich
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| | - Mohamed H S Awwad
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| | - Ute Hegenbart
- Amyloidosis Center, Heidelberg University, Heidelberg, Germany
| | - Hartmut Goldschmidt
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany.,National Center for Tumor Diseases, University Hospital, Heidelberg, Germany
| | - Jens Hillengass
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| | - Dirk Hose
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany.,National Center for Tumor Diseases, University Hospital, Heidelberg, Germany
| | - Anja Seckinger
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany.,National Center for Tumor Diseases, University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| | - Anthony D Ho
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| | | | - Michael Hundemer
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
222
|
Wallington-Beddoe CT, Sobieraj-Teague M, Kuss BJ, Pitson SM. Resistance to proteasome inhibitors and other targeted therapies in myeloma. Br J Haematol 2018; 182:11-28. [PMID: 29676460 DOI: 10.1111/bjh.15210] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The number of novel therapies for the treatment of myeloma is rapidly increasing, as are the clinical trials evaluating them in combination with other novel and established therapies. Proteasome inhibitors, immunomodulatory agents and monoclonal antibodies are the most well known and studied classes of novel agents targeting myeloma, with histone deacetylase inhibitors, nuclear export inhibitors and several other approaches also being actively investigated. However, in parallel with the development and clinical use of these novel myeloma therapies is the emergence of novel mechanisms of resistance, many of which remain elusive, particularly for more recently developed agents. Whilst resistance mechanisms have been best studied for proteasome inhibitors, particularly bortezomib, class effects do not universally apply to all class members, and within-class differences in efficacy, toxicity and resistance mechanisms have been observed. Although immunomodulatory agents share the common cellular target cereblon and thus resistance patterns relate to cereblon expression, the unique cell surface antigens to which monoclonal antibodies are directed means these agents frequently exhibit unique within-class differences in clinical efficacy and resistance patterns. This review describes the major classes of novel therapies for myeloma, highlights the major clinical trials within each class and discusses known resistance mechanisms.
Collapse
Affiliation(s)
- Craig T Wallington-Beddoe
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia.,College of Medicine and Public Health, Flinders University, Bedford Park, South Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| | - Magdalena Sobieraj-Teague
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia.,SA Pathology, Adelaide, Australia
| | - Bryone J Kuss
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia.,SA Pathology, Adelaide, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| |
Collapse
|
223
|
Pick M, Vainstein V, Goldschmidt N, Lavie D, Libster D, Gural A, Grisariu S, Avni B, Ben Yehuda D, Gatt ME. Daratumumab resistance is frequent in advanced-stage multiple myeloma patients irrespective of CD38 expression and is related to dismal prognosis. Eur J Haematol 2018; 100:494-501. [DOI: 10.1111/ejh.13046] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Marjorie Pick
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - Vladimir Vainstein
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - Neta Goldschmidt
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - David Lavie
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - Diana Libster
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - Alexander Gural
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - Sigal Grisariu
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - Batia Avni
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - Dina Ben Yehuda
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| | - Moshe E. Gatt
- Department of Hematology; Hadassah Hebrew University Medical Center; Jerusalem Israel
| |
Collapse
|
224
|
Bride KL, Vincent TL, Im SY, Aplenc R, Barrett DM, Carroll WL, Carson R, Dai Y, Devidas M, Dunsmore KP, Fuller T, Glisovic-Aplenc T, Horton TM, Hunger SP, Loh ML, Maude SL, Raetz EA, Winter SS, Grupp SA, Hermiston ML, Wood BL, Teachey DT. Preclinical efficacy of daratumumab in T-cell acute lymphoblastic leukemia. Blood 2018; 131:995-999. [PMID: 29305553 PMCID: PMC5833263 DOI: 10.1182/blood-2017-07-794214] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/11/2017] [Indexed: 12/27/2022] Open
Abstract
As a consequence of acquired or intrinsic disease resistance, the prognosis for patients with relapsed or refractory T-cell acute lymphoblastic leukemia (T-ALL) is dismal. Novel, less toxic drugs are clearly needed. One of the most promising emerging therapeutic strategies for cancer treatment is targeted immunotherapy. Immune therapies have improved outcomes for patients with other hematologic malignancies including B-cell ALL; however no immune therapy has been successfully developed for T-ALL. We hypothesize targeting CD38 will be effective against T-ALL. We demonstrate that blasts from patients with T-ALL have robust surface CD38 surface expression and that this expression remains stable after exposure to multiagent chemotherapy. CD38 is expressed at very low levels on normal lymphoid and myeloid cells and on a few tissues of nonhematopoietic origin, suggesting that CD38 may be an ideal target. Daratumumab is a human immunoglobulin G1κ monoclonal antibody that binds CD38, and has been demonstrated to be safe and effective in patients with refractory multiple myeloma. We tested daratumumab in a large panel of T-ALL patient-derived xenografts (PDX) and found striking efficacy in 14 of 15 different PDX. These data suggest that daratumumab is a promising novel therapy for pediatric T-ALL patients.
Collapse
Affiliation(s)
- Karen L Bride
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tiffaney L Vincent
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Soo-Yeon Im
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Richard Aplenc
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David M Barrett
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - William L Carroll
- Laura and Isaac Perlmutter Cancer Center at NYU Langone, New York University, New York, NY
| | | | - Yunfeng Dai
- Department of Biostatistics, University of Florida, Gainesville, FL
| | | | | | - Tori Fuller
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tina Glisovic-Aplenc
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Terzah M Horton
- Baylor College of Medicine Dan L. Duncan Comprehensive Cancer Center, Houston, TX
| | - Stephen P Hunger
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Mignon L Loh
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Shannon L Maude
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Elizabeth A Raetz
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Stuart S Winter
- Children's Minnesota Cancer and Blood Disorders, Minneapolis, MN; and
| | - Stephan A Grupp
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michelle L Hermiston
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA
| | | | - David T Teachey
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
225
|
Frerichs KA, Nagy NA, Lindenbergh PL, Bosman P, Marin Soto J, Broekmans M, Groen RWJ, Themeli M, Nieuwenhuis L, Stege C, Nijhof IS, Mutis T, Zweegman S, Lokhorst HM, van de Donk NWCJ. CD38-targeting antibodies in multiple myeloma: mechanisms of action and clinical experience. Expert Rev Clin Immunol 2018; 14:197-206. [PMID: 29465271 DOI: 10.1080/1744666x.2018.1443809] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is generally an incurable hematological malignancy with heterogeneous overall survival rates ranging from a few months to more than 10 years. Survival is especially poor for patients who developed disease that is refractory to immunomodulatory drugs and proteasome inhibitors. Areas covered: This review will discuss the importance of CD38-targeting antibodies for the treatment of MM patients to improve their outcome. Expert commentary: Intense immuno-oncological laboratory research has resulted in the development of functionally active monoclonal antibodies against cell surface markers present on MM cells. In this respect, CD38-targeting antibodies such as daratumumab, MOR202, and isatuximab, have high single agent activity in heavily pretreated MM patients by virtue of their pleiotropic mechanisms of action including Fc-dependent effector mechanisms and immunomodulatory activities. Importantly, CD38-targeting antibodies are well tolerated, with infusion reactions as most frequent adverse event. Altogether, this makes them attractive combination partners with other anti-MM agents. Daratumumab is already approved as monotherapy and in combination with lenalidomide-dexamethasone as well as bortezomib-dexamethasone in pretreated MM patients. Furthermore, results from studies evaluating CD38-targeting antibodies in newly diagnosed MM patients are also promising, indicating that CD38-targeting antibodies will be broadly used in MM, resulting in further improvements in survival.
Collapse
Affiliation(s)
- Kristine A Frerichs
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Noemi Anna Nagy
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Pieter L Lindenbergh
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Patty Bosman
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Jhon Marin Soto
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Marloes Broekmans
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Richard W J Groen
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Maria Themeli
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Louise Nieuwenhuis
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Claudia Stege
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Inger S Nijhof
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Tuna Mutis
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Sonja Zweegman
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Henk M Lokhorst
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | | |
Collapse
|
226
|
van de Donk NWCJ, Casneuf T, Di Cara A, Parren PW, Zweegman S, van Kessel B, Lokhorst HM, Usmani SZ, Lonial S, Richardson PG, Chiu C, Mutis T, Nijhof IS, Sasser AK. Impact of Fc gamma receptor polymorphisms on efficacy and safety of daratumumab in relapsed/refractory multiple myeloma. Br J Haematol 2018; 184:475-479. [DOI: 10.1111/bjh.15122] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
| | | | | | - Paul W. Parren
- Genmab; Utrecht The Netherlands
- Department of Cancer and inflammation Research; Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
- Department of Immunohaematology and Blood Transfusion; Leiden University Medical Centre; Leiden the Netherlands
| | - Sonja Zweegman
- Department of Haematology; VU University Medical Centre; Amsterdam The Netherlands
| | - Berris van Kessel
- Department of Haematology; VU University Medical Centre; Amsterdam The Netherlands
| | - Henk M. Lokhorst
- Department of Haematology; VU University Medical Centre; Amsterdam The Netherlands
| | - Saad Z. Usmani
- Levine Cancer Institute/Carolinas Healthcare System; Charlotte NC USA
| | - Sagar Lonial
- Winship Cancer Institute; Emory University School of Medicine; Atlanta GA USA
| | - Paul G. Richardson
- Jerome Lipper Myeloma Center, Department of Medical Oncology; Dana-Farber Cancer Institute; Harvard Medical School; Boston MA USA
| | | | - Tuna Mutis
- Department of Haematology; VU University Medical Centre; Amsterdam The Netherlands
| | - Inger S. Nijhof
- Department of Haematology; VU University Medical Centre; Amsterdam The Netherlands
| | | |
Collapse
|
227
|
Cuesta-Mateos C, Alcaraz-Serna A, Somovilla-Crespo B, Muñoz-Calleja C. Monoclonal Antibody Therapies for Hematological Malignancies: Not Just Lineage-Specific Targets. Front Immunol 2018; 8:1936. [PMID: 29387053 PMCID: PMC5776327 DOI: 10.3389/fimmu.2017.01936] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022] Open
Abstract
Today, monoclonal antibodies (mAbs) are a widespread and necessary tool for biomedical science. In the hematological cancer field, since rituximab became the first mAb approved by the Food and Drug Administration for the treatment of B-cell malignancies, a number of effective mAbs targeting lineage-specific antigens (LSAs) have been successfully developed. Non-LSAs (NLSAs) are molecules that are not restricted to specific leukocyte subsets or tissues but play relevant pathogenic roles in blood cancers including the development, proliferation, survival, and refractoriness to therapy of tumor cells. In consequence, efforts to target NLSAs have resulted in a plethora of mAbs-marketed or in development-to achieve different goals like neutralizing oncogenic pathways, blocking tumor-related chemotactic pathways, mobilizing malignant cells from tumor microenvironment to peripheral blood, modulating immune-checkpoints, or delivering cytotoxic drugs into tumor cells. Here, we extensively review several novel mAbs directed against NLSAs undergoing clinical evaluation for treating hematological malignancies. The review focuses on the structure of these antibodies, proposed mechanisms of action, efficacy and safety profile in clinical studies, and their potential applications in the treatment of hematological malignancies.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
- IMMED S.L., Immunological and Medicinal Products, Madrid, Spain
| | - Ana Alcaraz-Serna
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
| | - Beatriz Somovilla-Crespo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
| |
Collapse
|
228
|
de Oliveira GC, Kanamori KS, Auxiliadora-Martins M, Chini CCS, Chini EN. Measuring CD38 Hydrolase and Cyclase Activities: 1,N 6-Ethenonicotinamide Adenine Dinucleotide (ε-NAD) and Nicotinamide Guanine Dinucleotide (NGD) Fluorescence-based Methods. Bio Protoc 2018; 8:e2938. [PMID: 30112426 DOI: 10.21769/bioprotoc.2938] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
CD38 is a multifunctional enzyme involved in calcium signaling and Nicotinamide Adenine Dinucleotide (NAD+) metabolism. Through its major activity, the hydrolysis of NAD+, CD38 helps maintain the appropriate levels of this molecule for all NAD+-dependent metabolic processes to occur. Due to current advances and studies relating NAD+ decline and the development of multiple age-related conditions and diseases, CD38 gained importance in both basic science and clinical settings. The discovery and development of strategies to modulate its function and, possibly, treat diseases and improve health span put CD38 under the spotlights. Therefore, a consistent and reliable method to measure its activity and explore its use in medicine is required. We describe here the methods how our group measures both the hydrolase and cyclase activity of CD38, utilizing a fluorescence-based enzymatic assay performed in a plate reader using 1,N6-Ethenonicotinamide Adenine Dinucleotide (ε-NAD) and Nicotinamide Guanine Dinucleotide (NGD) as substrates, respectively.
Collapse
Affiliation(s)
- Guilherme C de Oliveira
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, USA
| | - Karina S Kanamori
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, USA
| | - Maria Auxiliadora-Martins
- Fundação de Apoio ao Ensino, Pesquisa e Assistência do Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FAEPA), São Paulo, Brazil
| | - Claudia C S Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, USA
| | - Eduardo N Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, USA
| |
Collapse
|
229
|
Nijhof IS, van de Donk NWCJ, Zweegman S, Lokhorst HM. Current and New Therapeutic Strategies for Relapsed and Refractory Multiple Myeloma: An Update. Drugs 2018; 78:19-37. [PMID: 29188449 PMCID: PMC5756574 DOI: 10.1007/s40265-017-0841-y] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although survival of multiple myeloma patients has at least doubled during recent years, most patients eventually relapse, and treatment at this stage may be particularly complex. At the time of relapse, the use of alternative drugs to those given upfront is current practice. However, many new options are currently available for the treatment of relapsed multiple myeloma, including recently approved drugs, such as the second- and third-generation proteasome inhibitors carfilzomib and ixazomib, the immunomodulatory agent pomalidomide, the monoclonal antibodies daratumumab and elotuzumab and the histone deacetylase inhibitor panobinostat, but also new targeted agents are under active investigation (e.g. signal transduction modulators, kinesin spindle protein inhibitors, and inhibitors of NF-kB, MAPK, AKT). We here describe a new paradigm for the treatment of relapsed multiple myeloma. The final goal should be finding a balance among efficacy, toxicity, and cost and, at the end of the road, achieving long-lasting control of the disease and eventually even cure in a subset of patients.
Collapse
Affiliation(s)
- Inger S Nijhof
- Department of Hematology, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Niels W C J van de Donk
- Department of Hematology, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Henk M Lokhorst
- Department of Hematology, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| |
Collapse
|
230
|
Bharani V, Sharma P, Bal A, Prakash G. A Plasma Cell Myeloma With Post-Therapy Anaplastic Morphology, Osteomyelosclerosis, and Strong Pan-Cytokeratin (AE1/AE3) Expression: A Potential Diagnostic Pitfall. Int J Surg Pathol 2017; 26:232-235. [DOI: 10.1177/1066896917750472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Vani Bharani
- Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prashant Sharma
- Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amanjit Bal
- Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gaurav Prakash
- Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
231
|
Cho SF, Lin L, Xing L, Yu T, Wen K, Anderson KC, Tai YT. Monoclonal Antibody: A New Treatment Strategy against Multiple Myeloma. Antibodies (Basel) 2017; 6:antib6040018. [PMID: 31548533 PMCID: PMC6698817 DOI: 10.3390/antib6040018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 12/22/2022] Open
Abstract
2015 was a groundbreaking year for the multiple myeloma community partly due to the breakthrough approval of the first two monoclonal antibodies in the treatment for patients with relapsed and refractory disease. Despite early disappointments, monoclonal antibodies targeting CD38 (daratumumab) and signaling lymphocytic activation molecule F7 (SLAMF7) (elotuzumab) have become available for patients with multiple myeloma in the same year. Specifically, phase 3 clinical trials of combination therapies incorporating daratumumab or elotuzumab indicate both efficacy and a very favorable toxicity profile. These therapeutic monoclonal antibodies for multiple myeloma can kill target cells via antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent phagocytosis, as well as by direct blockade of signaling cascades. In addition, their immunomodulatory effects may simultaneously inhibit the immunosuppressive bone marrow microenvironment and restore the key function of immune effector cells. In this review, we focus on monoclonal antibodies that have shown clinical efficacy or promising preclinical anti-multiple myeloma activities that warrant further clinical development. We summarize mechanisms that account for the in vitro and in vivo anti-myeloma effects of these monoclonal antibodies, as well as relevant preclinical and clinical results. Monoclonal antibody-based immunotherapies have already and will continue to transform the treatment landscape in multiple myeloma.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Liang Lin
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Lijie Xing
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324, Jingwu Road, Jinan 250021, China.
| | - Tengteng Yu
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Kenneth Wen
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
232
|
Shu B, Feng Y, Gui Y, Lu Q, Wei W, Xue X, Sun X, He W, Yang J, Dai C. Blockade of CD38 diminishes lipopolysaccharide-induced macrophage classical activation and acute kidney injury involving NF-κB signaling suppression. Cell Signal 2017; 42:249-258. [PMID: 29080804 DOI: 10.1016/j.cellsig.2017.10.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/11/2017] [Accepted: 10/24/2017] [Indexed: 10/18/2022]
Abstract
The CD38, possessing ADP-ribosyl cyclase (ADPR-cyclase) and cyclic ADP-ribose hydrolase (cADPR-hydrolase), is able to regulate a variety of cellular activities. However, the role and mechanisms for CD38 in macrophage activation and sepsis-induced acute kidney injury (AKI) remain to be determined. Here we report that in cultured macrophages, Lipopolysaccharide (LPS) could upregulate CD38 expression in time and dose dependent manner. Knocking down or blockade of CD38 in macrophages could inhibit LPS-induced macrophage M1 polarization accompanied by diminished NF-κB signaling activation. In mouse model with LPS-induced acute kidney injury, blocking CD38 with quercetin could significantly relieve kidney dysfunction, kidney pathological changes as well as inflammatory cell accumulation. Similar to those in the cultured cells, quercetin could inhibit macrophage M1 polarization and NF-κB signaling activation in macrophages from kidneys and spleens in mice after LPS injection. Together, these results demonstrate that CD38 mediates LPS-induced macrophage activation and AKI, which may be treated as a therapeutic target for sepsis-induced AKI in patients.
Collapse
Affiliation(s)
- Bingyan Shu
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China
| | - Ye Feng
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China
| | - Yuan Gui
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China
| | - Qingmiao Lu
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China
| | - Wei Wei
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China
| | - Xian Xue
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China
| | - Xiaoli Sun
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China
| | - Weichun He
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China
| | - Junwei Yang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China
| | - Chunsun Dai
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, PR China.
| |
Collapse
|
233
|
Chini CCS, Tarragó MG, Chini EN. NAD and the aging process: Role in life, death and everything in between. Mol Cell Endocrinol 2017; 455:62-74. [PMID: 27825999 PMCID: PMC5419884 DOI: 10.1016/j.mce.2016.11.003] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 09/22/2016] [Accepted: 11/03/2016] [Indexed: 12/11/2022]
Abstract
Life as we know it cannot exist without the nucleotide nicotinamide adenine dinucleotide (NAD). From the simplest organism, such as bacteria, to the most complex multicellular organisms, NAD is a key cellular component. NAD is extremely abundant in most living cells and has traditionally been described to be a cofactor in electron transfer during oxidation-reduction reactions. In addition to participating in these reactions, NAD has also been shown to play a key role in cell signaling, regulating several pathways from intracellular calcium transients to the epigenetic status of chromatin. Thus, NAD is a molecule that provides an important link between signaling and metabolism, and serves as a key molecule in cellular metabolic sensoring pathways. Importantly, it has now been clearly demonstrated that cellular NAD levels decline during chronological aging. This decline appears to play a crucial role in the development of metabolic dysfunction and age-related diseases. In this review we will discuss the molecular mechanisms responsible for the decrease in NAD levels during aging. Since other reviews on this subject have been recently published, we will concentrate on presenting a critical appraisal of the current status of the literature and will highlight some controversial topics in the field. In particular, we will discuss the potential role of the NADase CD38 as a driver of age-related NAD decline.
Collapse
Affiliation(s)
- Claudia C S Chini
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Oncology Research, GI Signaling Center, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Mariana G Tarragó
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Oncology Research, GI Signaling Center, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Eduardo N Chini
- Signal Transduction Laboratory, Kogod Aging Center, Department of Anesthesiology, Oncology Research, GI Signaling Center, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
234
|
Nanobodies effectively modulate the enzymatic activity of CD38 and allow specific imaging of CD38 + tumors in mouse models in vivo. Sci Rep 2017; 7:14289. [PMID: 29084989 PMCID: PMC5662768 DOI: 10.1038/s41598-017-14112-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/29/2017] [Indexed: 01/04/2023] Open
Abstract
The cell surface ecto-enzyme CD38 is a promising target antigen for the treatment of hematological malignancies, as illustrated by the recent approval of daratumumab for the treatment of multiple myeloma. Our aim was to evaluate the potential of CD38-specific nanobodies as novel diagnostics for hematological malignancies. We successfully identified 22 CD38-specific nanobody families using phage display technology from immunized llamas. Crossblockade analyses and in-tandem epitope binning revealed that the nanobodies recognize three different non-overlapping epitopes, with four nanobody families binding complementary to daratumumab. Three nanobody families inhibit the enzymatic activity of CD38 in vitro, while two others were found to act as enhancers. In vivo, fluorochrome-conjugated CD38 nanobodies efficiently reach CD38 expressing tumors in a rodent model within 2 hours after intravenous injection, thereby allowing for convenient same day in vivo tumor imaging. These nanobodies represent highly specific tools for modulating the enzymatic activity of CD38 and for diagnostic monitoring CD38-expressing tumors.
Collapse
|
235
|
Casneuf T, Xu XS, Adams HC, Axel AE, Chiu C, Khan I, Ahmadi T, Yan X, Lonial S, Plesner T, Lokhorst HM, van de Donk NWCJ, Clemens PL, Sasser AK. Effects of daratumumab on natural killer cells and impact on clinical outcomes in relapsed or refractory multiple myeloma. Blood Adv 2017; 1:2105-2114. [PMID: 29296857 PMCID: PMC5728278 DOI: 10.1182/bloodadvances.2017006866] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/06/2017] [Indexed: 12/18/2022] Open
Abstract
Daratumumab, a human CD38 imunoglobulin G 1κ monoclonal antibody, has demonstrated clinical activity and a manageable safety profile in monotherapy and combination therapy clinical trials in relapsed and/or refractory multiple myeloma. CD38 is expressed at high levels on myeloma cells and, to a lesser extent, on immune effector cells, including natural killer (NK) cells, which are important for daratumumab-mediated antibody-dependent cellular cytotoxicity (ADCC). Here, the pharmacodynamic effects of daratumumab monotherapy on NK cells, and the effect of NK cell dynamics on daratumumab efficacy and safety, were assessed. Daratumumab, like other CD38 antibodies, reduced NK-cell counts in peripheral blood mononuclear cells (PBMCs) of healthy donors in vitro. Data on NK-cell counts, clinical efficacy, and adverse events were pooled from two single-agent daratumumab studies, GEN501 and SIRIUS. In daratumumab-treated myeloma patients, total and activated NK-cell counts reduced rapidly in peripheral blood after the first dose, remained low over the course of treatment, and recovered after treatment ended. There was a clear maximum effect relationship between daratumumab dose and maximum reduction in NK cells. Similar reductions were observed in bone marrow. PBMCs from daratumumab-treated patients induced lysis by ADCC of CD38+ tumor cells in vitro, suggesting that the remaining NK cells retained cytotoxic functionality. There was no relationship between NK-cell count reduction and the efficacy or safety profile of daratumumab. Furthermore, although NK cell numbers are reduced after daratumumab treatment, they are not completely depleted and may still contribute to ADCC, clinical efficacy, and infection control.
Collapse
Affiliation(s)
| | - Xu Steven Xu
- Janssen Research & Development, LLC, Raritan, NJ
| | - Homer C Adams
- Janssen Research & Development, LLC, Spring House, PA
| | - Amy E Axel
- Janssen Research & Development, LLC, Spring House, PA
| | | | - Imran Khan
- Janssen Research & Development, LLC, Raritan, NJ
| | | | - Xiaoyu Yan
- Janssen Research & Development, LLC, Raritan, NJ
| | - Sagar Lonial
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA
| | - Torben Plesner
- Vejle Hospital and University of Southern Denmark, Vejle, Denmark; and
| | - Henk M Lokhorst
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | - A Kate Sasser
- Janssen Research & Development, LLC, Spring House, PA
| |
Collapse
|
236
|
De Vooght KMK, Oostendorp M, van Solinge WW. New mAb therapies in multiple myeloma: interference with blood transfusion compatibility testing. Curr Opin Hematol 2017; 23:557-562. [PMID: 27389485 DOI: 10.1097/moh.0000000000000276] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Immunotherapeutic strategies are emerging as novel therapeutic approaches in multiple myeloma, with several mAbs being in advanced stages of clinical development. Of these, CD38 targeting antibodies appear very promising. In trials with anti-CD38 mAb daratumumab, all patients demonstrated panreactivity in red blood cell (RBC) panel testing, complicating the selection of compatible RBCs for transfusion. This review provides an overview of the interferences and solutions to safely transfuse these patients. RECENT FINDINGS CD38 is weakly expressed on human erythrocytes. Since the first reports on the interference, different solutions have been reported, including the neutralization of anti-CD38 mAbs in plasma by sCD38 or antiidiotype antibodies, CD38 depletion of RBCs using dithiothreitol or cord blood test cells, and transfusion of extensively typed RBCs. SUMMARY All methods have (dis)advantages, and it depends on the facilities of the immunohematology laboratory what strategy to choose. As the selection of suitable RBC units can be seriously delayed, hospitals should have protocols to communicate this interference with patients, laboratories, and physicians in a timely manner. As CD38 antibodies may also have a role in the treatment of diseases beyond hematological malignancies, chances are high that health professionals will encounter this issue in the nearby future.
Collapse
Affiliation(s)
- Karen M K De Vooght
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | |
Collapse
|
237
|
Mykytiv V, Alwaheed A, Mohd Hashim NA. Double CD38 -/CD138 - negative multiple myeloma. Hematol Oncol Stem Cell Ther 2017; 12:64-66. [PMID: 29079129 DOI: 10.1016/j.hemonc.2017.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/23/2017] [Accepted: 08/07/2017] [Indexed: 10/18/2022] Open
Abstract
The standard diagnosis of multiple myeloma by flow cytometry is based on selection of population of CD38+/CD138+ positives cells. As the result treatment with proteasome inhibitors, CD138 may be underexpressed on atypical plasma cells. Thus, in order to improve this strategy, recently new CD138-independent method, based on CD38 positivity of plasma cells was developed. We present an unusual case of CD138- negative multiple myeloma which had become double CD138-/CD38- negative after treatment with daratumumab by which we would like to illustrate potential pitfalls of both strategies.
Collapse
Affiliation(s)
- Vitaliy Mykytiv
- Haematology Department, Cork University Hospital, Cork, Ireland.
| | - Abrar Alwaheed
- Haematology Department, Cork University Hospital, Cork, Ireland.
| | | |
Collapse
|
238
|
Ghai A, Maji D, Cho N, Chanswangphuwana C, Rettig M, Shen D, DiPersio J, Akers W, Dehdashti F, Achilefu S, Vij R, Shokeen M. Preclinical Development of CD38-Targeted [ 89Zr]Zr-DFO-Daratumumab for Imaging Multiple Myeloma. J Nucl Med 2017; 59:216-222. [PMID: 29025987 DOI: 10.2967/jnumed.117.196063] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/30/2017] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is a plasma B-cell hematologic cancer that causes significant skeletal morbidity. Despite improvements in survival, heterogeneity in response remains a major challenge in MM. Cluster of differentiation 38 (CD38) is a type II transmembrane glycoprotein overexpressed in myeloma cells and is implicated in MM cell signaling. Daratumumab is a U.S. Food and Drug Administration-approved high-affinity monoclonal antibody targeting CD38 that is clinically benefiting refractory MM patients. Here, we evaluated [89Zr]Zr-desferrioxamine (DFO)-daratumumab PET/CT imaging in MM tumor models. Methods: Daratumumab was conjugated to DFO-p-benzyl-isothiocyanate (DFO-Bz-NCS) for radiolabeling with 89Zr. Chelator conjugation was confirmed by electrospray ionization-mass spectrometry, and radiolabeling was monitored by instant thin-layer chromatography. Daratumumab was conjugated to Cyanine5 (Cy5) dye for cell microscopy. In vitro and in vivo evaluation of [89Zr]Zr-DFO-daratumumab was performed using CD38+ human myeloma MM1.S-luciferase (MM1.S) cells. Cellular studies determined the affinity, immunoreactivity, and specificity of [89Zr]Zr-DFO-daratumumab. A 5TGM1-luciferase (5TGM1)/KaLwRij MM mouse model served as control for imaging background noise. [89Zr]Zr-DFO-daratumumab PET/CT small-animal imaging was performed in severe combined immunodeficient mice bearing solid and disseminated MM tumors. Tissue biodistribution (7 d after tracer administration, 1.11 MBq/animal, n = 4-6/group) was performed in wild-type and MM1.S tumor-bearing mice. Results: A specific activity of 55.5 MBq/nmol (0.37 MBq/μg) was reproducibly obtained with [89Zr]Zr-daratumumab-DFO. Flow cytometry confirmed CD38 expression (>99%) on the surface of MM1.S cells. Confocal microscopy with daratumumab-Cy5 demonstrated specific cell binding. Dissociation constant, 3.3 nM (±0.58), and receptor density, 10.1 fmol/mg (±0.64), was obtained with a saturation binding assay. [89Zr]Zr-DFO-daratumumab/PET demonstrated specificity and sensitivity for detecting CD38+ myeloma tumors of variable sizes (8.5-128 mm3) with standardized uptake values ranging from 2.1 to 9.3. Discrete medullar lesions, confirmed by bioluminescence images, were efficiently imaged with [89Zr]Zr-DFO-daratumumab/PET. Biodistribution at 7 d after administration of [89Zr]Zr-DFO-daratumumab showed prominent tumor uptake (27.7 ± 7.6 percentage injected dose per gram). In vivo blocking was achieved with a 200-fold excess of unlabeled daratumumab. Conclusion: [89Zr]Zr-DFO- and Cy5-daratumumab demonstrated superb binding to CD38+ human MM cells and significantly low binding to CD38low cells. Daratumumab bioconjugates are being evaluated for image-guided delivery of therapeutic radionuclides.
Collapse
Affiliation(s)
- Anchal Ghai
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Dolonchampa Maji
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Nicholas Cho
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | | | - Michael Rettig
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Duanwen Shen
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - John DiPersio
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Walter Akers
- Center for In Vivo Imaging and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee; and
| | - Farrokh Dehdashti
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri.,Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri
| | - Ravi Vij
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Monica Shokeen
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri .,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
239
|
Guo L, Sun X, Hao Z, Huang J, Han X, You Y, Li Y, Shen M, Ozawa T, Kishi H, Muraguchi A, Jin A. Identification of Novel Epitopes with Agonistic Activity for the Development of Tumor Immunotherapy Targeting TRAIL-R1. J Cancer 2017; 8:2542-2553. [PMID: 28900492 PMCID: PMC5595084 DOI: 10.7150/jca.19918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 05/30/2017] [Indexed: 02/07/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor-1/2 (TRAIL-R1/R2), also known as death receptors, are expressed in a wide variety of tumor cells. Although TRAIL can induce cell apoptosis by engaging its cognate TRAIL-R1/R2, some tumor cells are or become resistant to TRAIL treatment. Monoclonal antibodies (mAbs) against TRAIL-R1/R2 have been developed to use as potential antitumor therapeutic agents instead of TRAIL. However, TRAIL-R1/R2-based tumor therapy has not yet been realized. We previously generated a series of fully human monoclonal antibodies against TRAIL-R1 (TR1-mAbs) that induced tumor cell apoptosis. In this study, we identified the antigenic binding sites of these TR1-mAbs and proposed two major epitopes on the extracellular domain of TRAIL-R1. The analysis revealed that the epitopes of some TR1-mAbs partially overlaps with the beginning of TRAIL-binding sites, and other epitopes are located within the TRAIL-binding region. Among these mAbs, TR1-422 and TR1-419 mAbs have two antigenic binding sites that bound to the same binding region, but they have different essential amino acid residues and binding site sizes. Furthermore, we investigated the apoptosis activity of TR1-419 and TR1-422 mAbs in the form of IgG and IgM. In contrast to the IgG-type TR1-419 and TR1-422 mAbs, which enhanced and inhibited TRAIL-induced apoptosis, respectively, both IgM-type TR1-419 and TR1-422 mAb strongly induced cell apoptosis with or without soluble TRAIL (sTRAIL). Moreover, the results showed that IgM-type TR1-419 and TR1-422 mAbs alone can sufficiently activate the extrinsic and intrinsic apoptosis signaling pathways and suppress tumor growth in vivo. Consequently, we identified two antigenic binding sites with agonistic activity, and their specific IgM-type mAbs exhibited strong cytotoxic activity in tumor cells in vitro and in vivo. Thus, these agonistic antigenic binding sites may be useful for the development of effective Ab-based drugs or Ab-based cell immunotherapy for various human solid tumors.
Collapse
Affiliation(s)
- Lu Guo
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150081, China.,Department of Basic Medical Sciences, Heilongjiang Nursing College, Harbin, Heilongjiang 150086, China
| | - Xin Sun
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhichao Hao
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Jingjing Huang
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Xiaojian Han
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yajie You
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yaying Li
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Meiying Shen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150000, China
| | - Tatsuhiko Ozawa
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Hiroyuki Kishi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Atsushi Muraguchi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Aishun Jin
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| |
Collapse
|
240
|
Angioi A, Lepori N, López AC, Sethi S, Fervenza FC, Pani A. Treatment of primary membranous nephropathy: where are we now? J Nephrol 2017; 31:489-502. [PMID: 28875476 DOI: 10.1007/s40620-017-0427-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/26/2017] [Indexed: 02/02/2023]
Abstract
In the last 10 years, basic science and clinical research have made important contributions to the understanding and management of primary membranous nephropathy (MN). The identification of antibodies directed against the M-type phospholipase A2 receptor (PLA2R) and thrombospondin type-1 domain-containing 7A protein have added a new perspective on diagnosis, monitoring the immunological activity, predicting prognosis and guiding therapy in patients with primary MN. Mounting evidence suggests that quantification and follow-up of antiPLA2R Abs levels can help in assessing prognosis and evaluate the response to treatment. The kidney disease improving global outcomes guidelines published in 2012 have not been updated. New data on the use of rituximab suggest it should be considered as a potential initial therapy in the treatment of patients with primary MN.
Collapse
Affiliation(s)
- Andrea Angioi
- Division of Nephrology and Dialysis, Azienda Ospedaliera G. Brotzu, Piazzale Ricchi n 1, 09100, Cagliari, Italy
| | - Nicola Lepori
- Division of Nephrology and Dialysis, Azienda Ospedaliera G. Brotzu, Piazzale Ricchi n 1, 09100, Cagliari, Italy
| | - Ana Coloma López
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Sanjeev Sethi
- Department of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Antonello Pani
- Division of Nephrology and Dialysis, Azienda Ospedaliera G. Brotzu, Piazzale Ricchi n 1, 09100, Cagliari, Italy.
| |
Collapse
|
241
|
Mansour A, Wakkach A, Blin-Wakkach C. Emerging Roles of Osteoclasts in the Modulation of Bone Microenvironment and Immune Suppression in Multiple Myeloma. Front Immunol 2017; 8:954. [PMID: 28848556 PMCID: PMC5554508 DOI: 10.3389/fimmu.2017.00954] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 07/26/2017] [Indexed: 12/26/2022] Open
Abstract
Multiple myeloma (MM) is one of the most common forms of hematologic malignancy resulting from cancerous proliferation of mature malignant plasma cells (MPCs). But despite the real improvement in therapeutics in the past years, it remains largely incurable. MM is the most frequent cancer to involve bone due to the stimulation of osteoclast (OCL) differentiation and activity. OCLs have a unique capacity to resorb bone. However, recent studies reveal that they are not restrained to this sole function. They participate in the control of angiogenesis, medullary niches, and immune responses, including in MM. Therefore, therapeutic approaches targeting OCLs probably affect not only bone resorption but also many other functions, and OCLs should not be considered anymore only as targets to improve the bone phenotype but also to modulate bone microenvironment. In this review, we explore these novel contributions of OCLs to MM which reveal their strong implication in the MM physiopathology. We also underline the therapeutic interest of targeting OCLs not only to overcome bone lesions, but also to improve bone microenvironment and anti-tumoral immune responses.
Collapse
Affiliation(s)
- Anna Mansour
- CNRS, UMR7370, LP2M, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France.,Faculté de Médecine, Université Aix-Marseille, Marseille, France
| | - Abdelilah Wakkach
- CNRS, UMR7370, LP2M, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| | - Claudine Blin-Wakkach
- CNRS, UMR7370, LP2M, Faculté de Médecine, Nice, France.,Université Nice Sophia Antipolis, Nice, France
| |
Collapse
|
242
|
Role of daratumumab in transfusion medicine: a must know entity. Rev Bras Hematol Hemoter 2017; 39:375-378. [PMID: 29150115 PMCID: PMC5693279 DOI: 10.1016/j.bjhh.2017.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 01/09/2023] Open
|
243
|
Katsyuba E, Auwerx J. Modulating NAD + metabolism, from bench to bedside. EMBO J 2017; 36:2670-2683. [PMID: 28784597 DOI: 10.15252/embj.201797135] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 12/11/2022] Open
Abstract
Discovered in the beginning of the 20th century, nicotinamide adenine dinucleotide (NAD+) has evolved from a simple oxidoreductase cofactor to being an essential cosubstrate for a wide range of regulatory proteins that include the sirtuin family of NAD+-dependent protein deacylases, widely recognized regulators of metabolic function and longevity. Altered NAD+ metabolism is associated with aging and many pathological conditions, such as metabolic diseases and disorders of the muscular and neuronal systems. Conversely, increased NAD+ levels have shown to be beneficial in a broad spectrum of diseases. Here, we review the fundamental aspects of NAD+ biochemistry and metabolism and discuss how boosting NAD+ content can help ameliorate mitochondrial homeostasis and as such improve healthspan and lifespan.
Collapse
Affiliation(s)
- Elena Katsyuba
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
244
|
Abstract
PURPOSE OF REVIEW The treatment landscape of multiple myeloma is rapidly changing; however, despite improvement in patients' survival, it still remains a largely incurable disease. One hallmark of myeloma is substantial immune dysfunction leading to an increased infection rate and the inability of immune surveillance to detect neoplastic cells. Here, we critically analyze clinical approaches to harness the immune system to overcome this defect with a focus on antibody based and adoptive cellular therapies. RECENT FINDINGS Clinical trials exploring these immunotherapies to treat myeloma are now well underway and show promising results. In relapsed myeloma, monoclonal antibodies directed against plasma cell antigens and immune checkpoints have already shown substantial efficacy. In parallel, trials of adoptive cellular therapy have exciting promise in myeloma, having induced dramatic responses in a handful of early study participants. Taken together, immunotherapeutic approaches hold enormous potential in the field of multiple myeloma and in the near future can be combined with or even replace the current standard of care.
Collapse
Affiliation(s)
- Mattia D'Agostino
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Mario Boccadoro
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Eric L Smith
- Myeloma Service, Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
245
|
Abstract
In patients with membranous nephropathy, alkylating agents (cyclophosphamide or chlorambucil) alone or in combination with steroids achieve remission of nephrotic syndrome more effectively than conservative treatment or steroids alone, but can cause myelotoxicity, infections, and cancer. Calcineurin inhibitors can improve proteinuria, but are nephrotoxic. Most patients relapse after treatment withdrawal and can become treatment dependent, which increases the risk of nephrotoxicity. The discovery of nephritogenic autoantibodies against podocyte M-type phospholipase A2 receptor (PLA2R) and thrombospondin type-1 domain- containing protein 7A (THSD7A) antigens provides a clear pathophysiological rationale for interventions that specifically target B-cell lineages to prevent antibody production and subepithelial deposition. The anti-CD20 monoclonal antibody rituximab is safe and achieves remission of proteinuria in approximately two-thirds of patients with membranous nephropathy. In those with PLA2R-related disease, remission can be predicted by anti-PLA2R antibody depletion and relapse by antibody re-emergence into the circulation. Thus, integrated evaluation of serology and proteinuria could guide identification of affected patients and treatment with individually tailored protocols. Nonspecific and toxic immunosuppressive regimens will fall out of use. B-cell modulation by rituximab and second-generation anti-CD20 antibodies (or plasma cell-targeted therapy in anti-CD20 resistant forms of disease) will lead to a novel therapeutic paradigm for patients with membranous nephropathy.
Collapse
|
246
|
Golay J. Direct targeting of cancer cells with antibodies: What can we learn from the successes and failure of unconjugated antibodies for lymphoid neoplasias? J Autoimmun 2017; 85:6-19. [PMID: 28666691 DOI: 10.1016/j.jaut.2017.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/11/2017] [Indexed: 12/26/2022]
Abstract
Following approval in 1997 of the anti-CD20 antibody rituximab for the treatment of B-NHL and CLL, many other unconjugated IgG1 MAbs have been tested in pre-clinical and clinical trials for the treatment of lymphoid neoplasms. Relatively few have been approved however and these are directed against a limited number of target antigens (CD20, CD52, CCR4, CD38, CD319). We review here the known biological properties of these antibodies and discuss which factors may have led to their success or may, on the contrary, limit their clinical application. Common factors of the approved MAbs are that the target antigen is expressed at relatively high levels on the neoplastic targets and their mechanism of action is mostly immune-mediated. Indeed most of these MAbs induce ADCC and phagocytosis by macrophages, and many also activate complement, leading to target cell lysis. In contrast direct cell death induction is not a common feature but may enhance efficacy in some cases. Interestingly, a key factor for the success of several MAbs appears to be their capacity to skew immunity towards an anti-tumour mode, by inhibiting/depleting suppressor cells and/or activating immune cells within the microenvironment, independently of FcγRs. We also expose here some of the strategies employed by industry to expand the clinical use of these molecules beyond their original indication. Interestingly, due to the central role of lymphocytes in the control of the immune response, several of the antibodies are now successfully used to treat many different autoimmune diseases and have also been formally approved for some of these new indications. There is little doubt that this trend will continue and that the precise mechanisms of therapeutic MAbs will be further dissected and better understood in the context of both tumour immunology and autoimmunity.
Collapse
Affiliation(s)
- Josée Golay
- Center of Cellular Therapy "G. Lanzani", USC Haematology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Via Garibaldi 11-13, 24128, Bergamo, Italy.
| |
Collapse
|
247
|
Lee CH, Romain G, Yan W, Watanabe M, Charab W, Todorova B, Lee J, Triplett K, Donkor M, Lungu OI, Lux A, Marshall N, Lindorfer MA, Goff ORL, Balbino B, Kang TH, Tanno H, Delidakis G, Alford C, Taylor RP, Nimmerjahn F, Varadarajan N, Bruhns P, Zhang YJ, Georgiou G. IgG Fc domains that bind C1q but not effector Fcγ receptors delineate the importance of complement-mediated effector functions. Nat Immunol 2017; 18:889-898. [PMID: 28604720 DOI: 10.1038/ni.3770] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/16/2017] [Indexed: 12/17/2022]
Abstract
Engineered crystallizable fragment (Fc) regions of antibody domains, which assume a unique and unprecedented asymmetric structure within the homodimeric Fc polypeptide, enable completely selective binding to the complement component C1q and activation of complement via the classical pathway without any concomitant engagement of the Fcγ receptor (FcγR). We used the engineered Fc domains to demonstrate in vitro and in mouse models that for therapeutic antibodies, complement-dependent cell-mediated cytotoxicity (CDCC) and complement-dependent cell-mediated phagocytosis (CDCP) by immunological effector molecules mediated the clearance of target cells with kinetics and efficacy comparable to those of the FcγR-dependent effector functions that are much better studied, while they circumvented certain adverse reactions associated with FcγR engagement. Collectively, our data highlight the importance of CDCC and CDCP in monoclonal-antibody function and provide an experimental approach for delineating the effect of complement-dependent effector-cell engagement in various therapeutic settings.
Collapse
Affiliation(s)
- Chang-Han Lee
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Gabrielle Romain
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Wupeng Yan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Makiko Watanabe
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Wissam Charab
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Biliana Todorova
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France.,INSERM, U760, Paris, France
| | - Jiwon Lee
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Kendra Triplett
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Moses Donkor
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Oana I Lungu
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Anja Lux
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Nicholas Marshall
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Odile Richard-Le Goff
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France.,INSERM, U760, Paris, France
| | - Bianca Balbino
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France.,INSERM, U760, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Tae Hyun Kang
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Hidetaka Tanno
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Corrine Alford
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Navin Varadarajan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Pierre Bruhns
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France.,INSERM, U760, Paris, France
| | - Yan Jessie Zhang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA.,Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA.,Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA.,Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA.,Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA.,Center for Systems and Synthetic Biology University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
248
|
Ehlerding EB, England CG, Jiang D, Graves SA, Kang L, Lacognata S, Barnhart TE, Cai W. CD38 as a PET Imaging Target in Lung Cancer. Mol Pharm 2017; 14:2400-2406. [PMID: 28573863 DOI: 10.1021/acs.molpharmaceut.7b00298] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Daratumumab (Darzalex, Janssen Biotech) is a clinically approved antibody targeting CD38 for the treatment of multiple myeloma. However, CD38 is also expressed by other cancer cell types, including lung cancer, where its expression or absence may offer prognostic value. We therefore developed a PET tracer based upon daratumumab for tracking CD38 expression, utilizing murine models of non-small cell lung cancer to verify its specificity. Daratumumab was prepared for radiolabeling with 89Zr (t1/2 = 78.4 h) through conjugation with desferrioxamine (Df). Western blot, flow cytometry, and saturation binding assays were utilized to characterize CD38 expression and binding of daratumumab to three non-small cell lung cancer cell lines: A549, H460, and H358. Murine xenograft models of the cell lines were also generated for further in vivo studies. Longitudinal PET imaging was performed following injection of 89Zr-Df-daratumumab out to 120 h postinjection, and nonspecific uptake was also evaluated through the injection of a radiolabeled control IgG antibody in A549 mice, 89Zr-Df-IgG. Ex vivo biodistribution and histological analyses were also performed after the terminal imaging time point at 120 h postinjection. Through cellular studies, A549 cells were found to express higher levels of CD38 than the H460 or H358 cell lines. PET imaging and ex vivo biodistribution studies verified in vitro trends, with A549 tumor uptake peaking at 8.1 ± 1.2%ID/g at 120 h postinjection according to PET analysis, and H460 and H358 at lower levels at the same time point (6.7 ± 0.7%ID/g and 5.1 ± 0.4%ID/g, respectively; n = 3 or 4). Injection of a nonspecific radiolabeled IgG into A549 tumor-bearing mice also demonstrated lower tracer uptake of 4.4 ± 1.3%ID/g at 120 h. Immunofluorescent staining of tumor tissues showed higher staining levels present in A549 tissues over H460 and H358. Thus, 89Zr-Df-daratumumab is able to image CD38-expressing tissues in vivo using PET, as verified through the exploration of non-small cell lung cancer models in this study. This agent therefore holds potential to image CD38 in other malignancies and aid in patient stratification and elucidation of the biodistribution of CD38.
Collapse
Affiliation(s)
- Emily B Ehlerding
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Christopher G England
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Dawei Jiang
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Stephen A Graves
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Lei Kang
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Saige Lacognata
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | - Weibo Cai
- Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States.,Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin 53705, United States
| |
Collapse
|
249
|
Shallis RM, Terry CM, Lim SH. The multi-faceted potential of CD38 antibody targeting in multiple myeloma. Cancer Immunol Immunother 2017; 66:697-703. [PMID: 28341874 PMCID: PMC11029060 DOI: 10.1007/s00262-017-1990-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 03/16/2017] [Indexed: 10/19/2022]
Abstract
CD38, an adenine dinucleotide phosphate (ADP) ribose cyclase and a cyclic ADP ribose hydrolase, is widely expressed on the surface of multiple myeloma (MM) cells. It is known to play a pivotal role in the downstream pathways that mediate MM cell growth, signal transduction, and adhesion. The clinical use of CD38 monoclonal antibodies (MoAbs), such as daratumumab, either as monotherapy or in combination with other anti-MM agents, has produced impressive results in patients who have failed standard MM therapy. CD38 MoAbs exhibit several cytotoxic mechanisms on MM cells. In addition to the classical effector mechanisms associated with antibody therapy, CD38 MoAbs induce MM apoptosis and clonal T-cell expansion. Here, we summarize the results of some pivotal clinical studies using a human CD38 MoAb, daratumumab, in patients with MM, discuss the anti-MM effector mechanisms induced by CD38 MoAbs, and review the potential tumor antigens that may be suitable targets for immunotherapy of MM. Finally, we present a paradigm of immunotherapy for MM patients using CD38 MoAbs followed by GM-CSF and an immune checkpoint inhibitor in patients who have undergone high dose chemotherapy and autologous stem cell transplant. CD38 MoAbs have emerged as a novel and ultimately very promising immunotherapeutic agent for MM because of its ability to induce MM cytotoxicity through both arms of the adaptive immune responses.
Collapse
Affiliation(s)
- Rory M Shallis
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Room 140, APC Building, 593 Eddy Street, Providence, RI, 02903, USA
| | - Christopher M Terry
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Room 140, APC Building, 593 Eddy Street, Providence, RI, 02903, USA
| | - Seah H Lim
- Division of Hematology and Oncology, Rhode Island Hospital/Brown University Warren Alpert Medical School, Room 140, APC Building, 593 Eddy Street, Providence, RI, 02903, USA.
| |
Collapse
|
250
|
Abstract
INTRODUCTION Proteasome inhibitors and immunomodulatory drugs have contributed to the dramatic improvement in survival for patients with myeloma over the past decades. However, the disease typically relapses and new classes of drugs are needed. In 2015, two monoclonal antibodies were approved for the treatment of patients with relapsed multiple myeloma, and immunotherapy has rapidly become indispensable in the management of myeloma patients. Areas covered: Here, the authors discuss the published data regarding the mechanism of action, safety and clinical efficacy of the CD38-targeted monoclonal antibody daratumumab for the treatment of patients with multiple myeloma. Expert opinion: Daratumumab is indicated for myeloma patients who have received at least 3 prior therapies, including bortezomib, lenalidomide and pomalidomide. In 2016, daratumumab in combination with lenalidomide and dexamethasone, or bortezomib and dexamethasone was approved for the treatment of patients with multiple myeloma who have received at least one prior therapy. Daratumumab displays an excellent safety profile. Moderate-grade infusion-related reactions occurring mostly during the first infusion are the main treatment-emergent adverse event. In the context of daratumumab therapy, attention should be paid to interference with blood compatibility testing and response assessment. Daratumumab-based combination therapies are currently under evaluation in relapsed and newly diagnosed patients.
Collapse
Affiliation(s)
- Cyrille Touzeau
- a Department of Hematology , University Hospital , Nantes , France.,b CRCNA, INSERM, CNRS , University of Nantes , Nantes , France
| | - Philippe Moreau
- a Department of Hematology , University Hospital , Nantes , France.,b CRCNA, INSERM, CNRS , University of Nantes , Nantes , France
| |
Collapse
|