1
|
Collier-Bain HD, Emery A, Brown FF, Causer AJ, Oliver R, Eddy R, Leach S, Graby J, Augustine D, Moore S, Crowe J, Murray J, Turner JE, Campbell JP. Characterising how a single bout of exercise in people with myeloma affects clonal plasma cell and immune effector cell frequency in blood, and daratumumab efficacy in vitro. Brain Behav Immun Health 2024; 42:100865. [PMID: 39411424 PMCID: PMC11472639 DOI: 10.1016/j.bbih.2024.100865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Multiple myeloma is a haematological cancer characterised by the accumulation of clonal plasma cells in the bone marrow and is commonly treated with daratumumab, an anti-CD38 monoclonal antibody immunotherapy. Daratumumab often fails to induce stringent complete responses, due in part to resistance to antibody-dependent cellular cytotoxicity (ADCC) exerted by natural killer (NK)-cells and monocytes. Exercise bouts undertaken by healthy people induce lymphocytosis in blood, including to NK-cells and B-cells, but the effects of exercise are unknown in myeloma patients. In addition, whether exercise mobilises plasma cells has not been adequately investigated, and as such the potential impact of exercise on daratumumab treatment is unclear. In this exploratory pilot study, n = 16 smouldering multiple myeloma participants enrolled and n = 9 completed the study which comprised a bout of cycling 15% above anaerobic threshold for ∼30-min, with blood samples collected pre-, immediately post-, and 30-min post-exercise. Peripheral blood mononuclear cells were isolated from blood samples and incubated with the RPMI-8226 plasmacytoma cell line, with or without the presence of daratumumab to determine specific lysis using a calcein-release assay. Daratumumab-mediated cell lysis increased from 18.8% to 23.2% pre- to post-exercise, respectively (p < 0.001), owing to an increased frequency of CD3-CD56+CD16+ NK-cells (+348%), HLA-DR+CD14dimCD16+ monocytes (+125%), and HLA-DR+CD14+CD32+ monocytes (+41%) in blood (p < 0.01). However, overall, total plasma cells (CD38+CD138+) nor clonal plasma cells (CD38brightCD138+CD45-/dimCD19- with light-chain restriction) increased in blood (p > 0.05). Notably, we observed a 305% increase in NK-cells expressing CD38, the daratumumab target antigen, which might render NK-cells more susceptible to daratumumab-mediated fratricide - whereby NK-cells initiate ADCC against daratumumab-bound NK-cells. In conclusion, exercise modestly improved the efficacy of daratumumab-mediated ADCC in vitro. However, plasma cells were largely unchanged, and NK-cells expressing CD38 - the daratumumab target antigen - increased in blood. Future research should consider the optimal timings of exercise during daratumumab treatment in myeloma to avert exacerbation of daratumumab-mediated NK-cell lysis.
Collapse
Affiliation(s)
| | | | - Frankie F. Brown
- Department for Health, University of Bath, United Kingdom
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Adam J. Causer
- Department for Health, University of Bath, United Kingdom
| | - Rebecca Oliver
- Department for Health, University of Bath, United Kingdom
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, United Kingdom
| | - Rachel Eddy
- Department for Health, University of Bath, United Kingdom
| | - Shoji Leach
- Department for Health, University of Bath, United Kingdom
| | - John Graby
- Department for Health, University of Bath, United Kingdom
- Department of Cardiology, Royal United Hospitals Bath NHS Foundation Trust, United Kingdom
| | - Daniel Augustine
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, United Kingdom
| | - Sally Moore
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, United Kingdom
| | - Josephine Crowe
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, United Kingdom
| | - James Murray
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, United Kingdom
| | - James E. Turner
- Department for Health, University of Bath, United Kingdom
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - John P. Campbell
- Department for Health, University of Bath, United Kingdom
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| |
Collapse
|
2
|
Bisht K, Merino A, Igarashi R, Gauthier L, Chiron M, Desjonqueres A, Smith E, Briercheck E, Romee R, Alici E, Vivier E, O'Dwyer M, van de Velde H. Natural killer cell biology and therapy in multiple myeloma: challenges and opportunities. Exp Hematol Oncol 2024; 13:114. [PMID: 39538354 PMCID: PMC11562869 DOI: 10.1186/s40164-024-00578-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Despite therapeutic advancements, multiple myeloma (MM) remains incurable. NK cells have emerged as a promising option for the treatment of MM. NK cells are heterogenous and typically classified based on the relative expression of their surface markers (e.g., CD56 and CD16a). These cells elicit an antitumor response in the presence of low mutational burden and without neoantigen presentation via germline-encoded activating and inhibitory receptors that identify the markers of transformation present on the MM cells. Higher NK cell activity is associated with improved survival and prognosis, whereas lower activity is associated with advanced clinical stage and disease progression in MM. Moreover, not all NK cell phenotypes contribute equally toward the anti-MM effect; higher proportions of certain NK cell phenotypes result in better outcomes. In MM, the proportion, phenotype, and function of NK cells are drastically varied between different disease stages; this is further influenced by the bone marrow microenvironment, proportion of activating and inhibitory receptors on NK cells, expression of homing receptors, and bone marrow hypoxia. Antimyeloma therapies, such as autologous stem cell transplant, immunomodulation, proteasome inhibition, and checkpoint inhibition, further modulate the NK cell landscape in the patients. Thus, NK cells can naturally work in tandem with anti-MM therapies and be strategically modulated for improved anti-MM effect. This review article describes immunotypic and phenotypic differences in NK cells along with the functional changes in homeostatic and malignant states and provides expert insights on strategies to harness the potential of NK cells for improving outcomes in MM.
Collapse
Affiliation(s)
- Kamlesh Bisht
- Research and Development, Sanofi, Cambridge, MA, 02141, USA.
| | - Aimee Merino
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis-Saint Paul, MN, USA
| | - Rob Igarashi
- Research and Development, Sanofi, Cambridge, MA, 02141, USA
| | - Laurent Gauthier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | | | - Eric Smith
- Division of Hematologic Malignancies and Transplantation, Dana Farber Cancer Institute, Boston, MA, USA
| | - Edward Briercheck
- Division of Hematologic Malignancies and Transplantation, Dana Farber Cancer Institute, Boston, MA, USA
| | - Rizwan Romee
- Division of Hematologic Malignancies and Transplantation, Dana Farber Cancer Institute, Boston, MA, USA
| | - Evren Alici
- Department of Medicine, Karolinska Institutet (KI), Huddinge, Sweden
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, CNRS, INSERM, Marseille, France
- Marseille-Immunopôle, APHM, Hôpital de la Timone, Marseille, France
| | - Michael O'Dwyer
- Department of Haematology, University of Galway, Galway, Ireland
| | | |
Collapse
|
3
|
Lee H, Ko N, Namgoong S, Ham S, Koo J. Recent advances in and applications of ex vivo drug sensitivity analysis for blood cancers. Blood Res 2024; 59:37. [PMID: 39503808 PMCID: PMC11541977 DOI: 10.1007/s44313-024-00032-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/06/2024] [Indexed: 11/09/2024] Open
Abstract
Blood cancers, including leukemia, multiple myeloma, and lymphoma, pose significant challenges owing to their heterogeneous nature and the limitations of traditional treatments. Precision medicine has emerged as a transformative approach that offers tailored therapeutic strategies based on individual patient profiles. Ex vivo drug sensitivity analysis is central to this advancement, which enables testing of patient-derived cancer cells against a panel of therapeutic agents to predict clinical responses. This review provides a comprehensive overview of the latest advancements in ex vivo drug sensitivity analyses and their application in blood cancers. We discuss the development of more comprehensive drug response metrics and the evaluation of drug combinations to identify synergistic interactions. Additionally, we present evaluation of the advanced therapeutics such as antibody-drug conjugates using ex vivo assays. This review describes the critical role of ex vivo drug sensitivity analyses in advancing precision medicine by examining technological innovations and clinical applications. Ultimately, these innovations are paving the way for more effective and individualized treatments, improving patient outcomes, and establishing new standards for the management of blood cancers.
Collapse
Affiliation(s)
- Haeryung Lee
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Nahee Ko
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Sujin Namgoong
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Seunghyok Ham
- ImpriMedKorea, Inc., Seoul, 03920, Republic of Korea
| | - Jamin Koo
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
- ImpriMedKorea, Inc., Seoul, 03920, Republic of Korea.
| |
Collapse
|
4
|
Nakayama H, Aisa Y, Ito C, Sakurai A, Nakazato T. The Real-World Outcomes of Relapsed/Refractory Multiple Myeloma Treated with Elotuzumab, Pomalidomide, and Dexamethasone. Hematol Rep 2024; 16:593-602. [PMID: 39449301 PMCID: PMC11503276 DOI: 10.3390/hematolrep16040058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction: A combination of elotuzumab, pomalidomide, and dexamethasone (EPd) was approved for the treatment of relapsed/refractory multiple myeloma (RRMM) following the ELOQUENT-3 phase II clinical trial. However, the clinical experience with this therapy is still limited. In this retrospective study, we analyzed the efficacy and safety of EPd in a real-world cohort of RRMM patients. Patients and Methods: The medical records of 22 patients who received EPd for RRMM at Yokohama Municipal Citizen's Hospital (Japan) between January 2020 and July 2021 were reviewed. Results: The median age of our cohort was 73.5 years. The overall response rate was 55%. With a median follow-up of 20.2 months, the median progression-free survival (PFS) was 9.1 months (95% confidence interval [CI], 2.5-23.0 months). The median PFS was shorter in patients with a poor performance status (PS) than in those with favorable PS (2.5 vs. 10.8 months; p < 0.01). Patients with prior daratumumab had significantly shorter PFS than those without prior daratumumab (2.1 vs. 23.0 months; p < 0.01). Additionally, patients with prior pomalidomide had significantly shorter PFS (1.7 vs. 10.3 months; p < 0.01). In the multivariate analysis, poor PS (hazard ratio [HR] = 4.1, 95% CI: 1.1-15.6; p = 0.04) and prior exposure to daratumumab (HR = 3.8, 95% CI: 1.1-13.8; p = 0.04) remained significantly associated with shorter PFS. Conclusions: The results of our study suggest that EPd is an active and well-tolerated regimen in RRMM, even in real-world patients. Furthermore, EPd may be useful, especially in daratumumab-naïve patients.
Collapse
Affiliation(s)
| | | | | | | | - Tomonori Nakazato
- Department of Hematology, Yokohama Municipal Citizen’s Hospital, 1-1 Mituzawa-Nishicho, Kanagawa-ku, Yokohama-shi 221-0855, Kanagawa, Japan; (H.N.); (Y.A.); (C.I.); (A.S.)
| |
Collapse
|
5
|
Yang G, Nikkhoi SK, Owji H, Li G, Massumi M, Cervelli J, Vandavasi VG, Hatefi A. A Novel Tetravalent Bispecific Immune Cell Engager Activates Natural Killer Cells to Kill Cancer Cells without Mediating Fratricide. Antibodies (Basel) 2024; 13:75. [PMID: 39311380 PMCID: PMC11417942 DOI: 10.3390/antib13030075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024] Open
Abstract
We previously reported the structure, affinity, and anticancer activity of a bivalent bispecific natural killer cell engager (BiKE) composed of one anti-CD16a VHH and one anti-HER2 VHH fused via a linker. In this study, we explored the engineering of a tetravalent BiKE by fusing two anti-CD16a and two anti-HER2 VHHs in tandem, using bivalent BiKE as a template. The tetravalent BiKE was genetically engineered, and its tertiary structure was predicted using in silico modeling. The antigen binding and affinity of the tetravalent BiKE were assessed using ELISA, flow cytometry, and biolayer interferometry. The ability of the BiKEs to kill cancer cells was evaluated through classical and residual antibody-dependent cellular cytotoxicity (ADCC) assays. Additionally, we investigated the potential for NK cell fratricide via CD16a-CD16a crosslinking. Our results revealed that the tetravalent BiKE exhibited at least 100-fold higher affinity toward its target antigens compared to its bivalent counterpart. The residual ADCC assay indicated that the tetravalent BiKE was more effective in killing cancer cells than the bivalent BiKE, attributable to its lower Koff value, which prolonged its binding to NK cell surfaces. Fratricide assays demonstrated that neither the bivalent nor the tetravalent BiKE mediated fratricide. Notably, our findings showed that daratumumab-induced NK fratricide was restricted to CD38-CD38 crosslinking and was not related to ADCC via CD16a-CD38 crosslinking. This study is the first in the literature to show the successful engineering of a tetravalent immune cell engager composed of tandem VHH units, which achieves high affinity and anticancer activity without mediating fratricide.
Collapse
Affiliation(s)
- Ge Yang
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Hajar Owji
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA
| | - Geng Li
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA
| | - Mohammad Massumi
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA
| | - Jessica Cervelli
- Environmental and Occupational Health Science Institute, Flow Cytometry Core Facility, Rutgers University, Piscataway, NJ 08854, USA
| | - Venu Gopal Vandavasi
- Department of Chemistry, Biophysics Core Facility, Princeton University, Princeton, NJ 08544, USA
| | - Arash Hatefi
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA
- Cancer Pharmacology Program, Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
6
|
Mayer KA, Schrezenmeier E, Diebold M, Halloran PF, Schatzl M, Schranz S, Haindl S, Kasbohm S, Kainz A, Eskandary F, Doberer K, Patel UD, Dudani JS, Regele H, Kozakowski N, Kläger J, Boxhammer R, Amann K, Puchhammer-Stöckl E, Vietzen H, Beck J, Schütz E, Akifova A, Firbas C, Gilbert HN, Osmanodja B, Halleck F, Jilma B, Budde K, Böhmig GA. A Randomized Phase 2 Trial of Felzartamab in Antibody-Mediated Rejection. N Engl J Med 2024; 391:122-132. [PMID: 38804514 DOI: 10.1056/nejmoa2400763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND Antibody-mediated rejection is a leading cause of kidney-transplant failure. The targeting of CD38 to inhibit graft injury caused by alloantibodies and natural killer (NK) cells may be a therapeutic option. METHODS In this phase 2, double-blind, randomized, placebo-controlled trial, we assigned patients with antibody-mediated rejection that had occurred at least 180 days after transplantation to receive nine infusions of the CD38 monoclonal antibody felzartamab (at a dose of 16 mg per kilogram of body weight) or placebo for 6 months, followed by a 6-month observation period. The primary outcome was the safety and side-effect profile of felzartamab. Key secondary outcomes were renal-biopsy results at 24 and 52 weeks, donor-specific antibody levels, peripheral NK-cell counts, and donor-derived cell-free DNA levels. RESULTS A total of 22 patients underwent randomization (11 to receive felzartamab and 11 to receive placebo). The median time from transplantation until trial inclusion was 9 years. Mild or moderate infusion reactions occurred in 8 patients in the felzartamab group. Serious adverse events occurred in 1 patient in the felzartamab group and in 4 patients in the placebo group; graft loss occurred in 1 patient in the placebo group. At week 24, resolution of morphologic antibody-mediated rejection was more frequent with felzartamab (in 9 of 11 patients [82%]) than with placebo (in 2 of 10 patients [20%]), for a difference of 62 percentage points (95% confidence interval [CI], 19 to 100) and a risk ratio of 0.23 (95% confidence interval [CI], 0.06 to 0.83). The median microvascular inflammation score was lower in the felzartamab group than in the placebo group (0 vs. 2.5), for a mean difference of -1.95 (95% CI, -2.97 to -0.92). Also lower was a molecular score reflecting the probability of antibody-mediated rejection (0.17 vs. 0.77) and the level of donor-derived cell-free DNA (0.31% vs. 0.82%). At week 52, the recurrence of antibody-mediated rejection was reported in 3 of 9 patients who had a response to felzartamab, with an increase in molecular activity and biomarker levels toward baseline levels. CONCLUSIONS Felzartamab had acceptable safety and side-effect profiles in patients with antibody-mediated rejection. (Funded by MorphoSys and Human Immunology Biosciences; ClinicalTrials.gov number, NCT05021484; and EUDRACT number, 2021-000545-40.).
Collapse
Affiliation(s)
- Katharina A Mayer
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Eva Schrezenmeier
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Matthias Diebold
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Philip F Halloran
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Martina Schatzl
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Sabine Schranz
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Susanne Haindl
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Silke Kasbohm
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Alexander Kainz
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Farsad Eskandary
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Konstantin Doberer
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Uptal D Patel
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Jaideep S Dudani
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Heinz Regele
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Nicolas Kozakowski
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Johannes Kläger
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Rainer Boxhammer
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Kerstin Amann
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Elisabeth Puchhammer-Stöckl
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Hannes Vietzen
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Julia Beck
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Ekkehard Schütz
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Aylin Akifova
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Christa Firbas
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Houston N Gilbert
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Bilgin Osmanodja
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Fabian Halleck
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Bernd Jilma
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Klemens Budde
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| | - Georg A Böhmig
- From the Departments of Medicine III (K.A.M., M.D., M.S., S.H., A.K., F.E., K.D., G.A.B.), Clinical Pathology (H.R., N.K., J.K.), and Clinical Pharmacology (S.S., C.F., B.J.) and the Center of Virology (E.P.-S., H.V.), Medical University of Vienna, Vienna; the Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland (M.D.); the Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin (E.S., S.K., A.A., B.O., F.H., K.B.), MorphoSys, Planegg (R.B.), the Department of Pathology, University of Erlangen-Nürnberg, Erlangen (K.A.), and Chronix Biomedical, Göttingen (E.S., J.B.) - all in Germany; the Alberta Transplant Applied Genomics Centre, Faculty of Medicine and Dentistry, Heritage Medical Research Centre, University of Alberta, Edmonton, AB, Canada (P.F.H.); and Human Immunology Biosciences, South San Francisco, CA (U.D.P., J.S.D., H.N.G.)
| |
Collapse
|
7
|
Saltarella I, Link A, Lamanuzzi A, Reichen C, Robinson J, Altamura C, Melaccio A, Solimando AG, Ria R, Mariggiò MA, Vacca A, Frassanito MA, Desaphy JF. Improvement of daratumumab- or elotuzumab-mediated NK cell activity by the bi-specific 4-1BB agonist, DARPin α-FAPx4-1BB: A preclinical study in multiple myeloma. Biomed Pharmacother 2024; 176:116877. [PMID: 38850654 DOI: 10.1016/j.biopha.2024.116877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Multiple myeloma (MM) progression is closely dependent on cells in the bone marrow (BM) microenvironment, including fibroblasts (FBs) and immune cells. In their BM niche, MM cells adhere to FBs sustaining immune evasion, drug resistance and the undetectable endurance of tumor cells known as minimal residual disease (MRD). Here, we describe the novel bi-specific designed ankyrin repeat protein (DARPin) α-FAPx4-1BB (MP0310) with FAP-dependent 4-1BB agonistic activity. The α-FAPx4-1BB DARPin simultaneously binds to FAP and 4-1BB overexpressed by activated FBs and immune cells, respectively. Although flow cytometry analysis showed that T and NK cells from MM patients were not activated and did not express 4-1BB, stimulation with daratumumab or elotuzumab, monoclonal antibodies (mAbs) currently used for the treatment of MM, significantly upregulated 4-1BB both in vitro and in MM patients following mAb-based therapy. The mAb-induced 4-1BB overexpression allowed the engagement of α-FAPx4-1BB that acted as a bridge between FAP+FBs and 4-1BB+NK cells. Therefore, α-FAPx4-1BB enhanced both the adhesion of daratumumab-treated NK cells on FBs as well as their activation by improving release of CD107a and perforin, hence MM cell killing via antibody-mediated cell cytotoxicity (ADCC). Interestingly, α-FAPx4-1BB significantly potentiated daratumumab-mediated ADCC in the presence of FBs, suggesting that it may overcome the BM FBs' immunosuppressive effect. Overall, we speculate that treatment with α-FAPx4-1BB may represent a valuable strategy to improve mAb-induced NK cell activity fostering MRD negativity in MM patients through the eradication of latent MRD cells.
Collapse
MESH Headings
- Killer Cells, Natural/immunology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/metabolism
- Multiple Myeloma/drug therapy
- Multiple Myeloma/immunology
- Multiple Myeloma/pathology
- Humans
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal/pharmacology
- Cell Line, Tumor
- Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists
- Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Membrane Proteins/metabolism
- Membrane Proteins/agonists
- Endopeptidases
Collapse
Affiliation(s)
- Ilaria Saltarella
- Section of Pharmacology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | | | - Aurelia Lamanuzzi
- Section of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | | | | | - Concetta Altamura
- Section of Pharmacology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | | | - Antonio Giovanni Solimando
- Section of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | - Roberto Ria
- Section of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | - Maria Addolorata Mariggiò
- Section of Clinical Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | - Angelo Vacca
- Section of Internal Medicine and Clinical Oncology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy.
| | - Maria Antonia Frassanito
- Section of Clinical Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | - Jean-François Desaphy
- Section of Pharmacology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
8
|
Grab AL, Kim PS, John L, Bisht K, Wang H, Baumann A, Van de Velde H, Sarkar I, Shome D, Reichert P, Manta C, Gryzik S, Reijmers RM, Weinhold N, Raab MS. Pre-Clinical Assessment of SAR442257, a CD38/CD3xCD28 Trispecific T Cell Engager in Treatment of Relapsed/Refractory Multiple Myeloma. Cells 2024; 13:879. [PMID: 38786100 PMCID: PMC11120574 DOI: 10.3390/cells13100879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Current treatment strategies for multiple myeloma (MM) are highly effective, but most patients develop relapsed/refractory disease (RRMM). The anti-CD38/CD3xCD28 trispecific antibody SAR442257 targets CD38 and CD28 on MM cells and co-stimulates CD3 and CD28 on T cells (TCs). We evaluated different key aspects such as MM cells and T cells avidity interaction, tumor killing, and biomarkers for drug potency in three distinct cohorts of RRMM patients. We found that a significantly higher proportion of RRMM patients (86%) exhibited aberrant co-expression of CD28 compared to newly diagnosed MM (NDMM) patients (19%). Furthermore, SAR442257 mediated significantly higher TC activation, resulting in enhanced MM killing compared to bispecific functional knockout controls for all relapse cohorts (Pearson's r = 0.7). Finally, patients refractory to anti-CD38 therapy had higher levels of TGF-β (up to 20-fold) compared to other cohorts. This can limit the activity of SAR442257. Vactoserib, a TGF-β inhibitor, was able to mitigate this effect and restore sensitivity to SAR442257 in these experiments. In conclusion, SAR442257 has high potential for enhancing TC cytotoxicity by co-targeting CD38 and CD28 on MM and CD3/CD28 on T cells.
Collapse
Affiliation(s)
- Anna Luise Grab
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Peter S. Kim
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Lukas John
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kamlesh Bisht
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Hongfang Wang
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Anja Baumann
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Helgi Van de Velde
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Irene Sarkar
- LUMICKS, 1059 CM Amsterdam, The Netherlands; (I.S.); (D.S.); (R.M.R.)
| | - Debarati Shome
- LUMICKS, 1059 CM Amsterdam, The Netherlands; (I.S.); (D.S.); (R.M.R.)
| | - Philipp Reichert
- GMMG Central Study Lab, Biobank, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Calin Manta
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
| | - Stefanie Gryzik
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
| | | | - Niels Weinhold
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Marc S. Raab
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
9
|
Shehata HM, Dogra P, Gierke S, Holder P, Sanjabi S. Efbalropendekin Alfa enhances human natural killer cell cytotoxicity against tumor cell lines in vitro. Front Immunol 2024; 15:1341804. [PMID: 38515757 PMCID: PMC10954783 DOI: 10.3389/fimmu.2024.1341804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
IL-15 has shown preclinical activity by enhancing the functional maturation of natural killer (NK) cells. Clinical evaluation of the potential anticancer activity of most cytokines, including IL-15, has been limited by low tolerability and rapid in vivo clearance. Efbalropendekin Alfa (XmAb24306) is a soluble IL15/IL15-receptor alpha heterodimer complex fused to a half-life extended Fc domain (IL15/IL15Rα-Fc), engineered with mutations to reduce IL-15 affinity for CD122. Reduced affinity drives lower potency, leading to prolonged pharmacodynamic response in cynomolgus monkeys. We show that in vitro, human NK cells treated with XmAb24306 demonstrate enhanced cytotoxicity against various tumor cell lines. XmAb24306-treated NK cells also exhibit enhanced killing of 3D colorectal cancer spheroids. Daratumumab (dara), a monoclonal antibody (mAb) that targets CD38 results in antibody-dependent cellular cytotoxicity (ADCC) of both multiple myeloma (MM) cells and NK cells. Addition of XmAb24306 increases dara-mediated NK cell ADCC against various MM cell lines in vitro. Because NK cells express CD38, XmAb24306 increases dara-mediated NK cell fratricide, but overall does not negatively impact the ADCC activity against a MM cell line likely due to increased NK cell activity of the surviving cells. These data show that XmAb24306 increases direct and ADCC-mediated human NK cell cytotoxicity in vitro.
Collapse
Affiliation(s)
- Hesham M. Shehata
- Department of Translational Medicine Oncology, Genentech Inc., South San Francisco, CA, United States
| | - Pranay Dogra
- Department of Translational Medicine Oncology, Genentech Inc., South San Francisco, CA, United States
| | - Sarah Gierke
- Department of Pathology, Genentech Inc., South San Francisco, CA, United States
| | - Patrick Holder
- Department of Protein Chemistry, Genentech Inc., South San Francisco, CA, United States
| | - Shomyseh Sanjabi
- Department of Translational Medicine Oncology, Genentech Inc., South San Francisco, CA, United States
| |
Collapse
|
10
|
Zhang L, Peng X, Ma T, Liu J, Yi Z, Bai J, Li Y, Li L, Zhang L. Natural killer cells affect the natural course, drug resistance, and prognosis of multiple myeloma. Front Cell Dev Biol 2024; 12:1359084. [PMID: 38410372 PMCID: PMC10895066 DOI: 10.3389/fcell.2024.1359084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
Multiple myeloma (MM), a stage-developed plasma cell malignancy, evolves from monoclonal gammopathy of undetermined significance (MGUS) or smoldering MM (SMM). Emerging therapies including immunomodulatory drugs, proteasome inhibitors, monoclonal antibodies, chimeric antigen-T/natural killer (NK) cells, bispecific T-cell engagers, selective inhibitors of nuclear export, and small-molecule targeted therapy have considerably improved patient survival. However, MM remains incurable owing to inevitable drug resistance and post-relapse rapid progression. NK cells with germline-encoded receptors are involved in the natural evolution of MGUS/SMM to active MM. NK cells actively recognize aberrant plasma cells undergoing malignant transformation but are yet to proliferate during the elimination phase, a process that has not been revealed in the immune editing theory. They are potential effector cells that have been neglected in the therapeutic process. Herein, we characterized changes in NK cells regarding disease evolution and elucidated its role in the early clinical monitoring of MM. Additionally, we systematically explored dynamic changes in NK cells from treated patients who are in remission or relapse to explore future combination therapy strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Li Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Xiaohuan Peng
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Tao Ma
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jia Liu
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Zhigang Yi
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Jun Bai
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yanhong Li
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Liansheng Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| |
Collapse
|
11
|
Nishida H, Suzuki R, Nakajima K, Hayashi M, Morimoto C, Yamada T. HDAC Inhibition Induces CD26 Expression on Multiple Myeloma Cells via the c-Myc/Sp1-mediated Promoter Activation. CANCER RESEARCH COMMUNICATIONS 2024; 4:349-364. [PMID: 38284882 PMCID: PMC10854391 DOI: 10.1158/2767-9764.crc-23-0215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/13/2023] [Accepted: 01/24/2024] [Indexed: 01/30/2024]
Abstract
CD26 is ubiquitously and intensely expressed in osteoclasts in patients with multiple myeloma, whereas its expression in plasma cells of patients with multiple myeloma is heterogeneous because of its cellular diversity, immune escape, and disease progression. Decreased expression levels of CD26 in myeloma cells constitute one of the mechanisms underlying resistance to humanized anti-CD26 mAb therapy in multiple myeloma. In the current study, we show that histone deacetylase inhibition (HDACi) with broad or class-specific inhibitors involves the induction of CD26 expression on CD26neg myeloma cells both transcriptionally and translationally. Furthermore, dipeptidyl peptidase Ⅳ (DPPⅣ) enzymatic activity was concomitantly enhanced in myeloma cells. Combined treatment with HDACi plus CD26mAb synergistically facilitated lysis of CD26neg myeloma cells not only by antibody-dependent cellular cytotoxicity but also by the direct effects of mAb. Of note, its combination readily augmented lysis of CD26neg cell populations, refractory to CD26mAb or HDACi alone. Chromatin immunoprecipitation assay revealed that HDACi increased acetylation of histone 3 lysine 27 at the CD26 promoter of myeloma cells. Moreover, in the absence of HDACi, c-Myc was attached to the CD26 promoter via Sp1 on the proximal G-C box of myeloma cells, whereas, in the presence of HDACi, c-Myc was detached from Sp1 with increased acetylation of c-Myc on the promoter, leading to activation of the CD26 promoter and initiation of transcription in myeloma cells. Collectively, these results confirm that HDACi plays crucial roles not only through its anti-myeloma activity but by sensitizing CD26neg myeloma cells to CD26mAb via c-Myc/Sp1-mediated CD26 induction, thereby augmenting its cytotoxicity. SIGNIFICANCE There is a desire to induce and sustain CD26 expression on multiple myeloma cells to elicit superior anti-myeloma response by humanized anti-CD26 mAb therapy. HDACi upregulates the expression levels of CD26 on myeloma cells via the increased acetylation of c-MycK323 on the CD26 promoter, leading to initiation of CD26 transcription, thereby synergistically augments the efficacy of CD26mAb against CD26neg myeloma cells.
Collapse
Grants
- 20K07682,16K07180 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and technology of Japan (C)
- 19H03519 Grant-in Aid for Scientific Research from the Ministry of Education, Culture, Sports and technology of Japan (B)
- 19K22542 Grant-in-Aid for Exploratory Research form the Ministry of Education, Culture Sports, Science and Technology of Japan
- 19H03519 Grant-in Aid for Scientific Research from the Ministry of Education, Culture, Sports and technology of Japan (B)
- 19K22542 Grant-in-Aid for Exploratory Research form the Ministry of Education, Culture Sports, Science and Technology of Japan
Collapse
Affiliation(s)
- Hiroko Nishida
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Division of Hematology, Department of Internal of Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Reiko Suzuki
- Department of Collaborative Research Resources, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kiyora Nakajima
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Mutsumi Hayashi
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Chikao Morimoto
- Department of Pathology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Taketo Yamada
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Juntendo University, Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
12
|
Liao B, Tumanut C, Li L, Corper A, Challa D, Chang A, Begum H, Farokhi E, Woods C, Fan X. Identification of novel anti-CD16a antibody clones for the development of effective natural killer cell engagers. MAbs 2024; 16:2381261. [PMID: 39048914 PMCID: PMC11271076 DOI: 10.1080/19420862.2024.2381261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/03/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024] Open
Abstract
Natural killer (NK) cells are key players in human innate immunity. Cell engager antibody formats that recruit and activate NK cells more effectively have emerged as a promising immunotherapy approach to target cancer cells through more effective antibody-dependent cell-mediated cytotoxicity (ADCC). Monoclonal antibody drugs with ADCC activity have shown clinical benefit and improved outcomes for patients with certain types of cancer. CD16a, a Fc gamma III receptor, is the major component that is responsible for the ADCC activity of NK cells. Screening AvantGen's yeast displayed human antibody libraries led to the isolation of 2 antibody clones, #1A2 and #2-2A2, that selectively recognize both isoforms (F and V) of CD16a on primary NK cells with high affinity, yet minimally (#1A2) or do not (#2-2A2) cross-react with both allelotypes of CD16b (NA1 and NA2) expressed by neutrophils. Epitope mapping studies revealed that they bind to an epitope dependent on residue Y158 of CD16a, since mutation of Y158 to the corresponding CD16b residue H158 completely abolishes binding to CD16a. When formatted as bispecific antibodies targeting CD16a and a tumor-associated antigen (TAA, e.g. CD19), they exhibit specific binding to NK cells and induce potent NK cell activation upon encountering tumor cells, resulting in effective tumor cell killing. Notably, these bispecific antibody engagers stimulate NK cell cytokine release during co-culture with target cells, resulting in target cell cytotoxicity. These anti-CD16a antibody clones are promising candidates for combination with any TAA of interest, offering the potential for novel NK cell engager-based cancer therapeutics that are minimally affected by the high concentrations of human IgG in the circulation.
Collapse
Affiliation(s)
| | | | - Lin Li
- AvantGen, Inc, San Diego, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Edri A, Ben-Haim N, Hailu A, Brycman N, Berhani-Zipori O, Rifman J, Cohen S, Yackoubov D, Rosenberg M, Simantov R, Teru H, Kurata K, Anderson KC, Hendel A, Pato A, Geffen Y. Nicotinamide-Expanded Allogeneic Natural Killer Cells with CD38 Deletion, Expressing an Enhanced CD38 Chimeric Antigen Receptor, Target Multiple Myeloma Cells. Int J Mol Sci 2023; 24:17231. [PMID: 38139060 PMCID: PMC10743602 DOI: 10.3390/ijms242417231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Natural killer (NK) cells are a vital component of cancer immune surveillance. They provide a rapid and potent immune response, including direct cytotoxicity and mobilization of the immune system, without the need for antigen processing and presentation. NK cells may also be better tolerated than T cell therapy approaches and are susceptible to various gene manipulations. Therefore, NK cells have become the focus of extensive translational research. Gamida Cell's nicotinamide (NAM) platform for cultured NK cells provides an opportunity to enhance the therapeutic potential of NK cells. CD38 is an ectoenzyme ubiquitously expressed on the surface of various hematologic cells, including multiple myeloma (MM). It has been selected as a lead target for numerous monoclonal therapeutic antibodies against MM. Monoclonal antibodies target CD38, resulting in the lysis of MM plasma cells through various antibody-mediated mechanisms such as antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity, and antibody-dependent cellular phagocytosis, significantly improving the outcomes of patients with relapsed or refractory MM. However, this therapeutic strategy has inherent limitations, such as the anti-CD38-induced depletion of CD38-expressing NK cells, thus hindering ADCC. We have developed genetically engineered NK cells tailored to treat MM, in which CD38 was knocked-out using CRISPR-Cas9 technology and an enhanced chimeric antigen receptor (CAR) targeting CD38 was introduced using mRNA electroporation. This combined genetic approach allows for an improved cytotoxic activity directed against CD38-expressing MM cells without self-inflicted NK-cell-mediated fratricide. Preliminary results show near-complete abolition of fratricide with a 24-fold reduction in self-lysis from 19% in mock-transfected and untreated NK cells to 0.8% of self-lysis in CD38 knock-out CAR NK cells. Furthermore, we have observed significant enhancements in CD38-mediated activity in vitro, resulting in increased lysis of MM target cell lines. CD38 knock-out CAR NK cells also demonstrated significantly higher levels of NK activation markers in co-cultures with both untreated and αCD38-treated MM cell lines. These NAM-cultured NK cells with the combined genetic approach of CD38 knockout and addition of CD38 CAR represent a promising immunotherapeutic tool to target MM.
Collapse
Affiliation(s)
- Avishay Edri
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Nimrod Ben-Haim
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (N.B.-H.); (M.R.)
| | - Astar Hailu
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Nurit Brycman
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Orit Berhani-Zipori
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Julia Rifman
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Sherri Cohen
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Dima Yackoubov
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Michael Rosenberg
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (N.B.-H.); (M.R.)
| | | | - Hideshima Teru
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; (H.T.); (K.K.); (K.C.A.)
| | - Keiji Kurata
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; (H.T.); (K.K.); (K.C.A.)
| | - Kenneth Carl Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; (H.T.); (K.K.); (K.C.A.)
| | - Ayal Hendel
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (N.B.-H.); (M.R.)
| | - Aviad Pato
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Yona Geffen
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| |
Collapse
|
14
|
Giang KA, Boxaspen T, Diao Y, Nilvebrant J, Kosugi-Kanaya M, Kanaya M, Krokeide SZ, Lehmann F, Svensson Gelius S, Malmberg KJ, Nygren PÅ. Affibody-based hBCMA x CD16 dual engagers for NK cell-mediated killing of multiple myeloma cells. N Biotechnol 2023; 77:139-148. [PMID: 37673373 DOI: 10.1016/j.nbt.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/16/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
We describe the development and characterization of the (to date) smallest Natural Killer (NK) cell re-directing human B Cell Maturation Antigen (hBCMA) x CD16 dual engagers for potential treatment of multiple myeloma, based on combinations of small 58 amino acid, non-immunoglobulin, affibody affinity proteins. Affibody molecules to human CD16a were selected from a combinatorial library by phage display resulting in the identification of three unique binders with affinities (KD) for CD16a in the range of 100 nM-3 µM. The affibody exhibiting the highest affinity demonstrated insensitivity towards the CD16a allotype (158F/V) and did not interfere with IgG (Fc) binding to CD16a. For the construction of hBCMA x CD16 dual engagers, different CD16a binding arms, including bi-paratopic affibody combinations, were genetically fused to a high-affinity hBCMA-specific affibody. Such 15-23 kDa dual engager constructs showed simultaneous hBCMA and CD16a binding ability and could efficiently activate resting primary NK cells and trigger specific lysis of a panel of hBCMA-positive multiple myeloma cell lines. Hence, we report a novel class of uniquely small NK cell engagers with specific binding properties and potent functional profiles.
Collapse
Affiliation(s)
- Kim Anh Giang
- Department of Protein Science, Div. Protein Engineering, AlbaNova University Center, KTH Royal Institute of Technology, S-114 21 Stockholm, Sweden
| | - Thorstein Boxaspen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | - Yumei Diao
- Oncopeptides AB, S-171 48 Stockholm, Sweden
| | - Johan Nilvebrant
- Department of Protein Science, Div. Protein Engineering, AlbaNova University Center, KTH Royal Institute of Technology, S-114 21 Stockholm, Sweden
| | - Mizuha Kosugi-Kanaya
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | - Minoru Kanaya
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | - Silje Zandstra Krokeide
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | | | | | - Karl-Johan Malmberg
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway.
| | - Per-Åke Nygren
- Department of Protein Science, Div. Protein Engineering, AlbaNova University Center, KTH Royal Institute of Technology, S-114 21 Stockholm, Sweden; Science For Life Laboratory, S-171 65 Solna, Sweden.
| |
Collapse
|
15
|
Süsal CC, Kraft L, Ender A, Süsal C, Schwenger A, Amann K, Böhmig GA, Schwenger V. Blood group-specific apheresis in combination with daratumumab as a rescue therapy of acute antibody-mediated rejection in a case of ABO- and human leukocyte antigen-incompatible kidney transplantation. SAGE Open Med Case Rep 2023; 11:2050313X231211050. [PMID: 38022864 PMCID: PMC10631334 DOI: 10.1177/2050313x231211050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
We report a case of antibody-mediated rejection treated with the human CD38 monoclonal antibody daratumumab in a 58-year-old female patient with end-stage kidney disease due to autosomal dominant polycystic kidney disease who received an ABO- and human leukocyte antigen antibody-incompatible living donor kidney transplant. The patient experienced an episode of severe antibody-mediated rejection within the first week of transplantation. Blood-group-antibody selective immunoadsorption in combination with administration of four doses of daratumumab (each 1800 mg s.c.) led to a persistent decrease of ABO- and more interestingly donor-specific human leukocyte antigen antibody reactivity and resulted in clinical and histopathological remission with full recovery of graft function, which has remained stable until post-transplant day 212. This case illustrates the potential of targeting CD38 in antibody-mediated rejection.
Collapse
Affiliation(s)
- Can C Süsal
- Department of Nephrology, Klinikum Stuttgart—Katharinenhospital, Stuttgart, Germany
| | - Leonie Kraft
- Department of Nephrology, Klinikum Stuttgart—Katharinenhospital, Stuttgart, Germany
| | - Andrea Ender
- Central Institute for Transfusion Medicine and Blood Donation, Katharinenhospital Stuttgart, Stuttgart, Germany
| | - Caner Süsal
- Transplant Immunology Research Center of Excellence, Koç University Hospital, Istanbul, Turkey
| | - Amelie Schwenger
- Experimental Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Kerstin Amann
- Department of Nephropathology, Department of Pathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Vedat Schwenger
- Department of Nephrology, Klinikum Stuttgart—Katharinenhospital, Stuttgart, Germany
| |
Collapse
|
16
|
Finotti G, Pietronigro E, Balanzin C, Lonardi S, Constantin G, Chao MP, Tecchio C, Vermi W, Cassatella MA. slan+ Monocytes Kill Cancer Cells Coated in Therapeutic Antibody by Trogoptosis. Cancer Immunol Res 2023; 11:1538-1552. [PMID: 37695535 DOI: 10.1158/2326-6066.cir-23-0239] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/04/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Abstract
Monocytes positive for 6-Sulfo LacNAc (slan) are a major subset of nonclassical CD14dimCD16+ monocytes in humans. We have shown that slan+ cells infiltrate lymphomas and elicit an antibody-dependent cellular cytotoxicity (ADCC) of neoplastic B cells mediated by the anti-CD20 therapeutic rituximab. Herein, by performing blocking experiments and flow cytometry analyses, as well as confocal microscopy and live-cell imaging assays, we extended the findings to other humanized antibodies and deciphered the underlying effector mechanism(s). Specifically, we show that, after coculture with target cells coated with anti-CD20 or anti-CD38, slan+ monocytes mediate trogocytosis, a cell-cell contact dependent, antibody-mediated process that triggers an active, mechanic disruption of target cell membranes. Trogocytosis by slan+ monocytes leads to a necrotic type of target cell death known as trogoptosis, which, once initiated, was partially sustained by endogenous TNFα. We also found that slan+ monocytes, unlike natural killer (NK) cells, mediate a direct ADCC with all types of anti-CD47 analyzed, and this was independent of their IgG isotype. The latter findings unveil a potentially relevant contribution by slan+ monocytes in mediating the therapeutic efficacy of anti-CD47 in clinical practice, which could be particularly important when NK cells are exhausted or deficient in number. Overall, our observations shed new light on the cytotoxic mechanisms exerted by slan+ monocytes in antibody-dependent tumor cell targeting and advance our knowledge on how to expand our therapeutic arsenal for cancer therapy.
Collapse
Affiliation(s)
- Giulia Finotti
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Enrica Pietronigro
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Camillo Balanzin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Silvia Lonardi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Mark P Chao
- Division of Hematology, Stanford University, Stanford, California
| | - Cristina Tecchio
- Section of Hematology and Bone Marrow Transplant Unit, Department of Medicine, University of Verona, Verona, Italy
| | - William Vermi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marco A Cassatella
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
17
|
Liu Z, Zhao X, Shen H, Liu X, Xu X, Fu R. Cellular immunity in the era of modern multiple myeloma therapy. Int J Cancer 2023; 153:1436-1447. [PMID: 37306091 DOI: 10.1002/ijc.34609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 06/13/2023]
Abstract
Multiple myeloma (MM) is a relapsing clonal plasma cell malignancy and incurable thus far. With the increasing understanding of myeloma, highlighting the critical importance of the immune system in the pathogenesis of MM is essential. The immune changes in MM patients after treatment are associated with prognosis. In this review, we summarize currently available MM therapies and discuss how they affect cellular immunity. We find that the modern anti-MM treatments enhance antitumour immune responses. A deeper understanding of the therapeutic activity of individual drugs offers more effective treatment approaches that enhance the beneficial immunomodulatory effects. Furthermore, we show that the immune changes after treatment in MM patients can provide useful prognostic marker. Analysing cellular immune responses offers new perspectives for evaluating clinical data and making comprehensive predictions for applying novel therapies in MM patients.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Xianghong Zhao
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Hongli Shen
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Xiaohan Liu
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Xintong Xu
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| |
Collapse
|
18
|
Collier-Bain HD, Brown FF, Causer AJ, Emery A, Oliver R, Moore S, Murray J, Turner JE, Campbell JP. Harnessing the immunomodulatory effects of exercise to enhance the efficacy of monoclonal antibody therapies against B-cell haematological cancers: a narrative review. Front Oncol 2023; 13:1244090. [PMID: 37681023 PMCID: PMC10482436 DOI: 10.3389/fonc.2023.1244090] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are standard care for many B-cell haematological cancers. The modes of action for these mAbs include: induction of cancer cell lysis by activating Fcγ-receptors on innate immune cells; opsonising target cells for antibody-dependent cellular cytotoxicity or phagocytosis, and/or triggering the classical complement pathway; the simultaneous binding of cancer cells with T-cells to create an immune synapse and activate perforin-mediated T-cell cytotoxicity against cancer cells; blockade of immune checkpoints to facilitate T-cell cytotoxicity against immunogenic cancer cell clones; and direct delivery of cytotoxic agents via internalisation of mAbs by target cells. While treatment regimens comprising mAb therapy can lead to durable anti-cancer responses, disease relapse is common due to failure of mAb therapy to eradicate minimal residual disease. Factors that limit mAb efficacy include: suboptimal effector cell frequencies, overt immune exhaustion and/or immune anergy, and survival of diffusely spread tumour cells in different stromal niches. In this review, we discuss how immunomodulatory changes arising from exposure to structured bouts of acute exercise might improve mAb treatment efficacy by augmenting (i) antibody-dependent cellular cytotoxicity, (ii) antibody-dependent cellular phagocytosis, (iii) complement-dependent cytotoxicity, (iv) T-cell cytotoxicity, and (v) direct delivery of cytotoxic agents.
Collapse
Affiliation(s)
| | - Frankie F. Brown
- Department for Health, University of Bath, Bath, United Kingdom
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Adam J. Causer
- Department for Health, University of Bath, Bath, United Kingdom
| | - Annabelle Emery
- Department for Health, University of Bath, Bath, United Kingdom
| | - Rebecca Oliver
- Department for Health, University of Bath, Bath, United Kingdom
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - Sally Moore
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - James Murray
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - James E. Turner
- Department for Health, University of Bath, Bath, United Kingdom
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
19
|
Zhang M, Lam KP, Xu S. Natural Killer Cell Engagers (NKCEs): a new frontier in cancer immunotherapy. Front Immunol 2023; 14:1207276. [PMID: 37638058 PMCID: PMC10450036 DOI: 10.3389/fimmu.2023.1207276] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/03/2023] [Indexed: 08/29/2023] Open
Abstract
Natural Killer (NK) cells are a type of innate lymphoid cells that play a crucial role in immunity by killing virally infected or tumor cells and secreting cytokines and chemokines. NK cell-mediated immunotherapy has emerged as a promising approach for cancer treatment due to its safety and effectiveness. NK cell engagers (NKCEs), such as BiKE (bispecific killer cell engager) or TriKE (trispecific killer cell engager), are a novel class of antibody-based therapeutics that exhibit several advantages over other cancer immunotherapies harnessing NK cells. By bridging NK and tumor cells, NKCEs activate NK cells and lead to tumor cell lysis. A growing number of NKCEs are currently undergoing development, with some already in clinical trials. However, there is a need for more comprehensive studies to determine how the molecular design of NKCEs affects their functionality and manufacturability, which are crucial for their development as off-the-shelf drugs for cancer treatment. In this review, we summarize current knowledge on NKCE development and discuss critical factors required for the production of effective NKCEs.
Collapse
Affiliation(s)
- Minchuan Zhang
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Kong-Peng Lam
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Singapore
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
20
|
Lamothe RC, Storlie MD, Espinosa DA, Rudlaff R, Browne P, Liu J, Rivas A, Devoto A, Oki J, Khoubyari A, Goltsman DSA, Lin JL, Butterfield CN, Brown CT, Thomas BC, Cost GJ. Novel CRISPR-Associated Gene-Editing Systems Discovered in Metagenomic Samples Enable Efficient and Specific Genome Engineering. CRISPR J 2023. [PMID: 37219969 DOI: 10.1089/crispr.2022.0089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023] Open
Abstract
Development of medicines using gene editing has been hampered by enzymological and immunological impediments. We described previously the discovery and characterization of improved, novel gene-editing systems from metagenomic data. In this study, we substantially advance this work with three such gene-editing systems, demonstrating their utility for cell therapy development. All three systems are capable of reproducible, high-frequency gene editing in primary immune cells. In human T cells, disruption of the T cell receptor (TCR) alpha-chain was induced in >95% of cells, both paralogs of the TCR beta-chain in >90% of cells, and >90% knockout of β2-microglobulin, TIGIT, FAS, and PDCD1. Simultaneous double knockout of TRAC and TRBC was obtained at a frequency equal to that of the single edits. Gene editing with our systems had minimal effect on T cell viability. Furthermore, we integrate a chimeric antigen receptor (CAR) construct into TRAC (up to ∼60% of T cells), and demonstrate CAR expression and cytotoxicity. We next applied our novel gene-editing tools to natural killer (NK) cells, B cells, hematopoietic stem cells, and induced pluripotent stem cells, generating similarly efficient cell-engineering outcomes including the creation of active CAR-NK cells. Interrogation of our gene-editing systems' specificity reveals a profile comparable with or better than Cas9. Finally, our nucleases lack preexisting humoral and T cell-based immunity, consistent with their sourcing from nonhuman pathogens. In all, we show these new gene-editing systems have the activity, specificity, and translatability necessary for use in cell therapy development.
Collapse
Affiliation(s)
| | | | | | | | | | - Jason Liu
- Metagenomi, Inc., Emeryville, California, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Verkleij CPM, Frerichs KA, Broekmans MEC, Duetz C, O'Neill CA, Bruins WSC, Homan-Weert PM, Minnema MC, Levin MD, Broijl A, Bos GMJ, Kersten MJ, Klein SK, Shikhagaie MM, Casneuf T, Abraham Y, Smets T, Vanhoof G, Cortes-Selva D, van Steenbergen L, Ramos E, Verona RI, Krevvata M, Sonneveld P, Zweegman S, Mutis T, van de Donk NWCJ. NK Cell Phenotype Is Associated With Response and Resistance to Daratumumab in Relapsed/Refractory Multiple Myeloma. Hemasphere 2023; 7:e881. [PMID: 37153876 PMCID: PMC10155898 DOI: 10.1097/hs9.0000000000000881] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/30/2023] [Indexed: 05/10/2023] Open
Abstract
The CD38-targeting antibody daratumumab has marked activity in multiple myeloma (MM). Natural killer (NK) cells play an important role during daratumumab therapy by mediating antibody-dependent cellular cytotoxicity via their FcγRIII receptor (CD16), but they are also rapidly decreased following initiation of daratumumab treatment. We characterized the NK cell phenotype at baseline and during daratumumab monotherapy by flow cytometry and cytometry by time of flight to assess its impact on response and development of resistance (DARA-ATRA study; NCT02751255). At baseline, nonresponding patients had a significantly lower proportion of CD16+ and granzyme B+ NK cells, and higher frequency of TIM-3+ and HLA-DR+ NK cells, consistent with a more activated/exhausted phenotype. These NK cell characteristics were also predictive of inferior progression-free survival and overall survival. Upon initiation of daratumumab treatment, NK cells were rapidly depleted. Persisting NK cells exhibited an activated and exhausted phenotype with reduced expression of CD16 and granzyme B, and increased expression of TIM-3 and HLA-DR. We observed that addition of healthy donor-derived purified NK cells to BM samples from patients with either primary or acquired daratumumab-resistance improved daratumumab-mediated MM cell killing. In conclusion, NK cell dysfunction plays a role in primary and acquired daratumumab resistance. This study supports the clinical evaluation of daratumumab combined with adoptive transfer of NK cells.
Collapse
Affiliation(s)
- Christie P M Verkleij
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Kristine A Frerichs
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Marloes E C Broekmans
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Carolien Duetz
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Chloe A O'Neill
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Wassilis S C Bruins
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Paola M Homan-Weert
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Monique C Minnema
- University Medical Center Utrecht, Department of Hematology, Utrecht University, The Netherlands
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Annemiek Broijl
- Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gerard M J Bos
- Department of Hematology, Maastricht University Medical Center, The Netherlands
| | - Marie José Kersten
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam UMC Location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | - Saskia K Klein
- Department of Internal Medicine, Meander Medical Center, Amersfoort, The Netherlands
- Department of Hematology, University Medical Center Groningen, The Netherlands
| | - Medya M Shikhagaie
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | | | - Yann Abraham
- Janssen Research and Development, Beerse, Belgium
| | - Tina Smets
- Janssen Research and Development, Beerse, Belgium
| | | | | | | | | | | | | | - Pieter Sonneveld
- Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sonja Zweegman
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Tuna Mutis
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Niels W C J van de Donk
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Xing L, Liu Y, Liu J. Targeting BCMA in Multiple Myeloma: Advances in Antibody-Drug Conjugate Therapy. Cancers (Basel) 2023; 15:cancers15082240. [PMID: 37190168 DOI: 10.3390/cancers15082240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Multiple myeloma (MM) is an incurable cancer of the plasma cells. In the last twenty years, treatment strategies have evolved toward targeting MM cells-from the shotgun chemotherapy approach to the slightly more targeted approach of disrupting important MM molecular pathways to the immunotherapy approach that specifically targets MM cells based on protein expression. Antibody-drug conjugates (ADCs) are introduced as immunotherapeutic drugs which utilize an antibody to deliver cytotoxic agents to cancer cells distinctively. Recent investigations of ADCs for MM treatment focus on targeting B cell maturation antigen (BCMA), which regulates B cell proliferation, survival, maturation, and differentiation into plasma cells (PCs). Given its selective expression in malignant PCs, BCMA is one of the most promising targets in MM immunotherapy. Compared to other BCMA-targeting immunotherapies, ADCs have several benefits, such as lower price, shorter production period, fewer infusions, less dependence on the patient's immune system, and they are less likely to over-activate the immune system. In clinical trials, anti-BCMA ADCs have shown safety and remarkable response rates in patients with relapsed and refractory MM. Here, we review the properties and clinical applications of anti-BCMA ADC therapies and discuss the potential mechanisms of resistance and ways to overcome them.
Collapse
Affiliation(s)
- Lijie Xing
- Department of Hematology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Yuntong Liu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Jiye Liu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
23
|
Karvouni M, Vidal-Manrique M, Susek KH, Hussain A, Gilljam M, Zhang Y, Gray JD, Lund J, Kaufmann G, Ljunggren HG, Ji H, Lundqvist A, Wagner AK, Guo W, Alici E. Challenges in αCD38-chimeric antigen receptor (CAR)-expressing natural killer (NK) cell-based immunotherapy in multiple myeloma: Harnessing the CD38dim phenotype of cytokine-stimulated NK cells as a strategy to prevent fratricide. Cytotherapy 2023:S1465-3249(23)00068-3. [PMID: 37055320 DOI: 10.1016/j.jcyt.2023.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/27/2023] [Accepted: 03/13/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND AIMS Adoptive cell therapy with chimeric antigen receptor (CAR)-expressing natural killer (NK) cells is an emerging approach that holds promise in multiple myeloma (MM). However, the generation of CAR-NK cells targeting CD38 is met with obstacles due to the expression of CD38 on NK cells. Knock-out of CD38 is currently explored as a strategy, although the consequences of the lack of CD38 expression with regards to engraftment and activity in the bone marrow microenvironment are not fully elucidated. Here, we present an alternative approach by harnessing the CD38dim phenotype occurring during long-term cytokine stimulation of primary NK cells. METHODS Primary NK cells were expanded from peripheral blood mononuclear cells by long-term IL-2 stimulation. During expansion, the CD38 expression was monitored in order to identify a time point when introduction of a novel affinity-optimized αCD38-CAR confered optimal viability, i.e. prevented fratricide. CD38dim NK cells were trasduced with retroviral vectors encoding for the CAR trasngene and their functionality was assessed in in vitro activation and cytotoxicity assays. RESULTS We verified the functionality of the αCD38-CAR-NK cells against CD38+ cell lines and primary MM cells. Importantly, we demonstrated that αCD38-CAR-NK cells derived from patients with MM have increased activity against autologous MM samples ex vivo. CONCLUSIONS Overall, our results highlight that incorporation of a functional αCD38-CAR construct into a suitable NK-cell expansion and activation protocol results in a potent and feasible immunotherapeutic strategy for the treatment of patients with MM.
Collapse
Affiliation(s)
- Maria Karvouni
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Marcos Vidal-Manrique
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Katharina H Susek
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Alamdar Hussain
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Mari Gilljam
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - J Dixon Gray
- Sorrento Therapeutics, Inc., San Diego, California, USA
| | - Johan Lund
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Henry Ji
- Sorrento Therapeutics, Inc., San Diego, California, USA
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Arnika K Wagner
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Wenzhong Guo
- Sorrento Therapeutics, Inc., San Diego, California, USA
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Klimentova M, Shelikhova L, Ilushina M, Kozlovskaya S, Blagov S, Popov A, Kashpor S, Fadeeva M, Olshanskaya J, Glushkova S, Pershin D, Balashov D, Maschan A, Maschan M. Targeted Therapy With Venetoclax and Daratumumab as Part of HSCT Preparative Regimen in Children With Chemorefractory Acute Myeloid Leukemia. Transplant Cell Ther 2023; 29:127.e1-127.e9. [PMID: 36436779 DOI: 10.1016/j.jtct.2022.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/16/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
The long-term outcome of allogeneic hematopoietic stem cell transplantation (HSCT) in chemorefractory acute myeloid leukemia (AML) remains suboptimal because of a high relapse rate. Enhancement of conditioning regimens by the incorporation of targeted anti-leukemia agents is a potential approach to improve the efficacy of HSCT. In a pilot trial and extended access cohort, we evaluated the safety and potential value of adding combinations of venetoclax and daratumumab to a preparative regimen among children with chemorefractory acute myeloid leukemia grafted with αβ T-cell-depleted peripheral blood stem cells. All 20 patients had active disease status of AML at the time of transplantation. The preparative regimen included myeloablative conditioning based on either total body irradiation or treosulfan. A haploidentical related donor was used as a graft source for all patients. Engraftment was not compromised, and no excess toxicity was noted. Minimal residual disease-negative complete remission was achieved in 17 patients (85%). The cumulative incidence of grade II to IV acute graft-versus-host disease (GVHD) was 17%, and the cumulative incidence of chronic GVHD was 7%. At 2 years, nonrelapse mortality was 10%, relapse incidence was 46%, event-free survival was 44%, and overall survival was 65%. Our data show the possibility of safely adding targeted agents to conditioning regimens; however, no evidence of a significant improvement in long-term transplantation outcomes in this cohort of patients was observed.
Collapse
Affiliation(s)
- Maria Klimentova
- Department of Hematopoietic Stem Cell Transplantation Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Larisa Shelikhova
- Department of Hematopoietic Stem Cell Transplantation Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Maria Ilushina
- Department of Hematopoietic Stem Cell Transplantation Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Svetlana Kozlovskaya
- Department of Hematopoietic Stem Cell Transplantation Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Sergei Blagov
- Department of Hematopoietic Stem Cell Transplantation Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Alexander Popov
- Immunophenotyping of Hemoblastoses Laboratory Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Svetlana Kashpor
- Cytogenetics and Molecular Genetics Laboratory Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Maria Fadeeva
- Transplantation Immunology and Immunotherapy Laboratory Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Julia Olshanskaya
- Immunophenotyping of Hemoblastoses Laboratory Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Svetlana Glushkova
- Immunophenotyping of Hemoblastoses Laboratory Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Dmitriy Pershin
- Immunophenotyping of Hemoblastoses Laboratory Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Dmitriy Balashov
- Department of Hematopoietic Stem Cell Transplantation Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia
| | - Alexei Maschan
- Pediatric Hematology Dmitriy Rogachev National Medical Center Of Pediatric Hematology, Oncology And Immunology, Moscow, Russia
| | - Michael Maschan
- Department of Hematopoietic Stem Cell Transplantation Dmitriy Rogachev National Medical Center Of Pediatric Hemotology, Oncology And Immunology, Moscow, Russia.
| |
Collapse
|
25
|
Dimopoulos MA, Dytfeld D, Grosicki S, Moreau P, Takezako N, Hori M, Leleu X, LeBlanc R, Suzuki K, Raab MS, Richardson PG, Popa McKiver M, Jou YM, Yao D, Das P, San-Miguel J. Elotuzumab Plus Pomalidomide and Dexamethasone for Relapsed/Refractory Multiple Myeloma: Final Overall Survival Analysis From the Randomized Phase II ELOQUENT-3 Trial. J Clin Oncol 2023; 41:568-578. [PMID: 35960908 PMCID: PMC9870233 DOI: 10.1200/jco.21.02815] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
PURPOSE In the phase II ELOQUENT-3 trial (ClinicalTrials.gov identifier: NCT02654132), elotuzumab combined with pomalidomide/dexamethasone (EPd) significantly improved progression-free survival (PFS) versus pomalidomide/dexamethasone (Pd) in patients with relapsed/refractory multiple myeloma (RRMM) previously treated with lenalidomide and a proteasome inhibitor (PI). Here, we present the final overall survival (OS) results. METHODS Patients with RRMM who had received ≥ 2 prior lines of therapy, with disease refractory to last therapy and either refractory or relapsed and refractory to lenalidomide and a PI were randomly assigned (1:1) to receive EPd or Pd. The primary end point was PFS per investigator assessment. ORR and OS were secondary end points planned to be tested hierarchically. RESULTS A total of 117 patients were randomly assigned to EPd (n = 60) and Pd (n = 57). Among treated patients (EPd 60, Pd 55), there were 37 (61.7%) deaths in the EPd group and 41 (74.5%) in the Pd group, most commonly because of disease progression (EPd 41.7%, Pd 49.1%). Median (95% CI) OS was significantly improved with EPd (29.8 [22.9 to 45.7] months) versus Pd (17.4 [13.8 to 27.7] months), with a hazard ratio of 0.59 (95% CI, 0.37 to 0.93; P = .0217). OS benefit with EPd was observed in most patient subgroups. The safety profile of EPd was consistent with prior reports with no new safety signals detected. CONCLUSION EPd demonstrated a statistically significant improvement in OS versus Pd in patients with RRMM previously treated with lenalidomide and a PI who had disease refractory to last therapy. In this setting, ELOQUENT-3 is the first randomized study of a triplet regimen incorporating a monoclonal antibody and Pd to improve both PFS and OS significantly.
Collapse
Affiliation(s)
- Meletios A. Dimopoulos
- National and Kapodistrian University of Athens School of Medicine, Athens, Greece,Meletios A. Dimopoulos, MD, National and Kapodistrian University of Athens School of Medicine, Alexandra Hospital, 80 Vasilissis Sofias Ave, Athens 11528, Greece; e-mail:
| | - Dominik Dytfeld
- Karol Marcinkowski University of Medical Sciences, Poznań, Poland
| | | | | | - Naoki Takezako
- National Hospital Organization Disaster Medical Center, Tokyo, Japan
| | - Mitsuo Hori
- Ibaraki Prefectural Central Hospital, Kasama, Japan
| | - Xavier Leleu
- Centre Hospitalier Universitaire de Poitiers–La Milétrie, Poitiers, France
| | - Richard LeBlanc
- Hôpital Maisonneuve-Rosemont, University of Montreal, Montreal, Québec, Canada
| | | | - Marc S. Raab
- Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | | | | | - Jesús San-Miguel
- Clínica Universidad de Navarra (CCUN), Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra (IDISNA), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| |
Collapse
|
26
|
Improving NK cell function in multiple myeloma with NKTR-255, a novel polymer-conjugated human IL-15. Blood Adv 2023; 7:9-19. [PMID: 35882498 DOI: 10.1182/bloodadvances.2022007985] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/27/2022] [Accepted: 07/12/2022] [Indexed: 01/18/2023] Open
Abstract
Multiple myeloma (MM) is characterized by an immunosuppressive microenvironment that enables tumor development. One of the mechanisms of immune evasion used by MM cells is the inhibition of natural killer (NK) cell effector functions; thus, the restoration of NK cell antitumor activity represents a key goal to increase tumor cell recognition, avoid tumor escape and potentially enhancing the effect of other drugs. In this study, we evaluated the ability of the investigational medicine NKTR-255, an IL-15 receptor agonist, to engage the IL-15 pathway and stimulate NK cells against MM cells. We observed that incubation with NKTR-255 was able to tilt the balance toward an activated phenotype in NK cells isolated from peripheral blood mononuclear cells of patients with MM, with increased expression of activating receptors on the surface of treated NK cells. This resulted in an enhanced degranulation, cytokine release, and anti-tumor cytotoxicity when the NK cells were exposed to both MM cell lines and primary MM cells. We further evaluated the in vivo effect of NKTR-255 in fully humanized immunocompetent mice subcutaneously engrafted with H929 MM cells. Compared with placebo, weekly injection of the mice with NKTR-255 increased the number of circulating NK cells in peripheral blood and delayed tumor growth. Finally, we observed that combination of NKTR-255 with the anti-CD38 antibody, daratumumab, was effective against MM cells in vitro and in vivo. Taken together, our data suggest a significant impact of NKTR-255 in inducing NK cell function against MM cells with important translational implications.
Collapse
|
27
|
Bertuglia G, Cani L, Larocca A, Gay F, D'Agostino M. Normalization of the Immunological Microenvironment and Sustained Minimal Residual Disease Negativity: Do We Need Both for Long-Term Control of Multiple Myeloma? Int J Mol Sci 2022; 23:15879. [PMID: 36555520 PMCID: PMC9781462 DOI: 10.3390/ijms232415879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Over the past two decades, the treatment landscape for multiple myeloma (MM) has progressed significantly, with the introduction of several new drug classes that have greatly improved patient outcomes. At present, it is well known how the bone marrow (BM) microenvironment (ME) exerts an immunosuppressive action leading to an exhaustion of the immune system cells and promoting the proliferation and sustenance of tumor plasma cells. Therefore, having drugs that can reconstitute a healthy BM ME can improve results in MM patients. Recent findings clearly demonstrated that achieving minimal residual disease (MRD) negativity and sustaining MRD negativity over time play a pivotal prognostic role. However, despite the achievement of MRD negativity, patients may still relapse. The understanding of immunologic changes in the BM ME during treatment, complemented by a deeper knowledge of plasma cell genomics and biology, will be critical to develop future therapies to sustain MRD negativity over time and possibly achieve an operational cure. In this review, we focus on the components of the BM ME and their role in MM, on the prognostic significance of MRD negativity and, finally, on the relative contribution of tumor plasma cell biology and BM ME to long-term disease control.
Collapse
Affiliation(s)
- Giuseppe Bertuglia
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Lorenzo Cani
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Alessandra Larocca
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Francesca Gay
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Mattia D'Agostino
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| |
Collapse
|
28
|
Rahmani S, Yazdanpanah N, Rezaei N. Natural killer cells and acute myeloid leukemia: promises and challenges. Cancer Immunol Immunother 2022; 71:2849-2867. [PMID: 35639116 PMCID: PMC10991240 DOI: 10.1007/s00262-022-03217-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
Acute myeloid leukemia (AML) is considered as one of the most malignant conditions of the bone marrow. Over the past few decades, despite substantial progresses in the management of AML, relapse remission remains a major problem. Natural killer cells (NK cells) are known as a unique component of the innate immune system. Due to swift tumor detection, distinct cytotoxic action, and extensive immune interaction, NK cells have been used in various cancer settings for decades. It has been a growing knowledge of therapeutic magnitudes ranging from adoptive NK cell transfer to chimeric antigen receptor NK cells, aiming to achieve better therapeutic responses in patients with AML. In this article, the potentials of NK cells for treatment of AML are highlighted, and challenges for such therapeutic methods are discussed. In addition, the clinical application of NK cells, mainly in patients with AML, is pictured according to the existing evidence.
Collapse
Affiliation(s)
- Shayan Rahmani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Ho M, Xiao A, Yi D, Zanwar S, Bianchi G. Treating Multiple Myeloma in the Context of the Bone Marrow Microenvironment. Curr Oncol 2022; 29:8975-9005. [PMID: 36421358 PMCID: PMC9689284 DOI: 10.3390/curroncol29110705] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The treatment landscape of multiple myeloma (MM) has evolved considerably with the FDA-approval of at least 15 drugs over the past two decades. Together with the use of autologous stem cell transplantation, these novel therapies have resulted in significant survival benefit for patients with MM. In particular, our improved understanding of the BM and immune microenvironment has led to the development of highly effective immunotherapies that have demonstrated unprecedented response rates even in the multiple refractory disease setting. However, MM remains challenging to treat especially in a high-risk setting. A key mediator of therapeutic resistance in MM is the bone marrow (BM) microenvironment; a deeper understanding is necessary to facilitate the development of therapies that target MM in the context of the BM milieu to elicit deeper and more durable responses with the ultimate goal of long-term control or a cure of MM. In this review, we discuss our current understanding of the role the BM microenvironment plays in MM pathogenesis, with a focus on its immunosuppressive nature. We also review FDA-approved immunotherapies currently in clinical use and highlight promising immunotherapeutic approaches on the horizon.
Collapse
Affiliation(s)
- Matthew Ho
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Alexander Xiao
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Dongni Yi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Saurabh Zanwar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Giada Bianchi
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02120, USA
| |
Collapse
|
30
|
Thangaraj JL, Jung SH, Vo MC, Chu TH, Phan MTT, Lee KH, Ahn SY, Kim M, Song GY, Ahn JS, Yang DH, Kim HJ, Cho D, Lee JJ. Expanded natural killer cells potentiate the antimyeloma activity of daratumumab, lenalidomide, and dexamethasone in a myeloma xenograft model. Cancer Immunol Immunother 2022; 72:1233-1246. [PMID: 36385211 PMCID: PMC10110729 DOI: 10.1007/s00262-022-03322-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022]
Abstract
AbstractThe development of new treatment agents in recent decades has significantly improved the survival of patients with multiple myeloma (MM). Nonetheless, MM remains an incurable disease; therefore, novel combination therapies are required. Natural killer (NK) cells are one of the safest immunotherapeutic options. In this study, we found that the anti-myeloma activity of expanded NK cells (eNKs) was improved by daratumumab, lenalidomide, and dexamethasone (DRd) in an MM xenograft mouse model. NK cells expanded from peripheral blood mononuclear cells collected from MM patients were highly cytotoxic against DRd pretreated tumor cells in vitro. To mimic the clinical protocol, a human MM xenograft model was developed using human RPMI8226-RFP-FLuc cells in NOD/SCID IL-2Rγnull (NSG) mice. MM bearing mice were randomly divided into six groups: no treatment, eNK, Rd, Rd + eNKs, DRd, and DRd + eNKs. DRd significantly enhanced the cytotoxicity of eNKs by upregulating NK cell activation ligands and effector function. DRd in combination with eNKs significantly reduced the serum M-protein level and prolonged mouse survival. In addition, DRd significantly increased the persistence of eNK and homing to MM sites. These results show that the anti-myeloma activity of ex vivo-expanded and activated NK cells is augmented by the immunomodulatory effect of DRd in MM-bearing mice, suggesting the therapeutic potential of this combination for MM patients.
Collapse
|
31
|
Pinto S, Pahl J, Schottelius A, Carter PJ, Koch J. Reimagining antibody-dependent cellular cytotoxicity in cancer: the potential of natural killer cell engagers. Trends Immunol 2022; 43:932-946. [PMID: 36306739 DOI: 10.1016/j.it.2022.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 01/12/2023]
Abstract
Bi-, tri- and multispecific antibodies have enabled the development of targeted cancer immunotherapies redirecting immune effector cells to eliminate malignantly transformed cells. These antibodies allow for simultaneous binding of surface antigens on malignant cells and activating receptors on innate immune cells, such as natural killer (NK) cells, macrophages, and neutrophils. Significant progress with such antibodies has been achieved, particularly in hematological malignancies. Nevertheless, several major challenges remain, including increasing their immunotherapeutic efficacy in a greater proportion of patients, particularly in those harboring solid tumors, and overcoming dose-limiting toxicities and immunogenicity. Here, we discuss novel antibody-engineering developments designed to maximize the potential of NK cells by NK cell engagers mediating antibody-dependent cellular cytotoxicity (ADCC), thereby expanding the armamentarium for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Paul J Carter
- Genentech, Department of Antibody Engineering, San Francisco, CA, USA
| | | |
Collapse
|
32
|
Molecular Determinants Underlying the Anti-Cancer Efficacy of CD38 Monoclonal Antibodies in Hematological Malignancies. Biomolecules 2022; 12:biom12091261. [PMID: 36139103 PMCID: PMC9496523 DOI: 10.3390/biom12091261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
CD38 was first discovered as a T-cell antigen and has since been found ubiquitously expressed in various hematopoietic cells, including plasma cells, NK cells, B cells, and granulocytes. More importantly, CD38 expression levels on malignant hematopoietic cells are significantly higher than counterpart healthy cells, thus presenting itself as a promising therapeutic target. In fact, for many aggressive hematological cancers, including CLL, DLBCL, T-ALL, and NKTL, CD38 expression is significantly associated with poorer prognosis and a hyperproliferative or metastatic phenotype. Studies have shown that, beyond being a biomarker, CD38 functionally mediates dysregulated survival, adhesion, and migration signaling pathways, as well as promotes an immunosuppressive microenvironment conducive for tumors to thrive. Thus, targeting CD38 is a rational approach to overcoming these malignancies. However, clinical trials have surprisingly shown that daratumumab monotherapy has not been very effective in these other blood malignancies. Furthermore, extensive use of daratumumab in MM is giving rise to a subset of patients now refractory to daratumumab treatment. Thus, it is important to consider factors modulating the determinants of response to CD38 targeting across different blood malignancies, encompassing both the transcriptional and post-transcriptional levels so that we can diversify the strategy to enhance daratumumab therapeutic efficacy, which can ultimately improve patient outcomes.
Collapse
|
33
|
Targeting CD38 in Neoplasms and Non-Cancer Diseases. Cancers (Basel) 2022; 14:cancers14174169. [PMID: 36077708 PMCID: PMC9454480 DOI: 10.3390/cancers14174169] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 01/12/2023] Open
Abstract
Simple Summary CD38 remains an interesting target for anticancer therapy. Its relatively high abundance in neoplasms and crucial impact on NAD+/cADPR metabolism and the activity of T cells allows for changing the immune response in autoimmune diseases, neoplasms, and finally the induction of cell death. Antibody-dependent cell cytotoxicity is responsible for cell death induced by targeting the tumor with anti-CD38 antibodies, such as daratumumab. A wide range of laboratory experiments and clinical trials show an especially promising role of anti-CD38 therapy against multiple myeloma, NK cell lymphomas, and CD19- B-cell malignancies. More studies are required to include more diseases in the therapeutic protocols involving the modulation of CD38 activity. Abstract CD38 is a myeloid antigen present both on the cell membrane and in the intracellular compartment of the cell. Its occurrence is often enhanced in cancer cells, thus making it a potential target in anticancer therapy. Daratumumab and isatuximab already received FDA approval, and novel agents such as MOR202, TAK079 and TNB-738 undergo clinical trials. Also, novel therapeutics such as SAR442085 aim to outrank the older antibodies against CD38. Multiple myeloma and immunoglobulin light-chain amyloidosis may be effectively treated with anti-CD38 immunotherapy. Its role in other hematological malignancies is also important concerning both diagnostic process and potential treatment in the future. Aside from the hematological malignancies, CD38 remains a potential target in gastrointestinal, neurological and pulmonary system disorders. Due to the strong interaction of CD38 with TCR and CD16 on T cells, it may also serve as the biomarker in transplant rejection in renal transplant patients. Besides, CD38 finds its role outside oncology in systemic lupus erythematosus and collagen-induced arthritis. CD38 plays an important role in viral infections, including AIDS and COVID-19. Most of the undergoing clinical trials focus on the use of anti-CD38 antibodies in the therapy of multiple myeloma, CD19- B-cell malignancies, and NK cell lymphomas. This review focuses on targeting CD38 in cancer and non-cancerous diseases using antibodies, cell-based therapies and CD38 inhibitors. We also provide a summary of current clinical trials targeting CD38.
Collapse
|
34
|
Valeri A, García-Ortiz A, Castellano E, Córdoba L, Maroto-Martín E, Encinas J, Leivas A, Río P, Martínez-López J. Overcoming tumor resistance mechanisms in CAR-NK cell therapy. Front Immunol 2022; 13:953849. [PMID: 35990652 PMCID: PMC9381932 DOI: 10.3389/fimmu.2022.953849] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the impressive results of autologous CAR-T cell therapy in refractory B lymphoproliferative diseases, CAR-NK immunotherapy emerges as a safer, faster, and cost-effective approach with no signs of severe toxicities as described for CAR-T cells. Permanently scrutinized for its efficacy, recent promising data in CAR-NK clinical trials point out the achievement of deep, high-quality responses, thus confirming its potential clinical use. Although CAR-NK cell therapy is not significantly affected by the loss or downregulation of its CAR tumor target, as in the case of CAR-T cell, a plethora of common additional tumor intrinsic or extrinsic mechanisms that could also disable NK cell function have been described. Therefore, considering lessons learned from CAR-T cell therapy, the emergence of CAR-NK cell therapy resistance can also be envisioned. In this review we highlight the processes that could be involved in its development, focusing on cytokine addiction and potential fratricide during manufacturing, poor tumor trafficking, exhaustion within the tumor microenvironment (TME), and NK cell short in vivo persistence on account of the limited expansion, replicative senescence, and rejection by patient’s immune system after lymphodepletion recovery. Finally, we outline new actively explored alternatives to overcome these resistance mechanisms, with a special emphasis on CRISPR/Cas9 mediated genetic engineering approaches, a promising platform to optimize CAR-NK cell function to eradicate refractory cancers.
Collapse
Affiliation(s)
- Antonio Valeri
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Almudena García-Ortiz
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Eva Castellano
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Laura Córdoba
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Elena Maroto-Martín
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Jessica Encinas
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Alejandra Leivas
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Paula Río
- Division of Hematopoietic Innovative Therapies, Biomedical Innovation Unit, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) and Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Joaquín Martínez-López
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- *Correspondence: Joaquín Martínez-López,
| |
Collapse
|
35
|
Venglar O, Bago JR, Motais B, Hajek R, Jelinek T. Natural Killer Cells in the Malignant Niche of Multiple Myeloma. Front Immunol 2022; 12:816499. [PMID: 35087536 PMCID: PMC8787055 DOI: 10.3389/fimmu.2021.816499] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells represent a subset of CD3- CD7+ CD56+/dim lymphocytes with cytotoxic and suppressor activity against virus-infected cells and cancer cells. The overall potential of NK cells has brought them to the spotlight of targeted immunotherapy in solid and hematological malignancies, including multiple myeloma (MM). Nonetheless, NK cells are subjected to a variety of cancer defense mechanisms, leading to impaired maturation, chemotaxis, target recognition, and killing. This review aims to summarize the available and most current knowledge about cancer-related impairment of NK cell function occurring in MM.
Collapse
Affiliation(s)
- Ondrej Venglar
- Faculty of Science, University of Ostrava, Ostrava, Czechia.,Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| | - Julio Rodriguez Bago
- Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| | - Benjamin Motais
- Faculty of Science, University of Ostrava, Ostrava, Czechia.,Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Roman Hajek
- Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| | - Tomas Jelinek
- Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| |
Collapse
|
36
|
Wu HT, Zhao XY. Regulation of CD38 on Multiple Myeloma and NK Cells by Monoclonal Antibodies. Int J Biol Sci 2022; 18:1974-1988. [PMID: 35342342 PMCID: PMC8935232 DOI: 10.7150/ijbs.68148] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/19/2022] [Indexed: 11/14/2022] Open
Abstract
CD38 is highly expressed on multiple myeloma (MM) cells and plays a role in regulating tumor generation and development. CD38 monoclonal antibodies (mAbs) have been used as an effective therapy for MM treatment by various mechanisms, including complement-dependent cytotoxic effects, antibody-dependent cell-mediated cytotoxicity, antibody-dependent cellular phagocytosis, programmed cell death, enzymatic modulation, and immunomodulation. Although CD38 mAbs inhibit the proliferation and survival of MM cells, there are substantial side effects on antitumoral NK cells. The NK-mediated immune response needs to be further evaluated to minimize the adverse effects of NK cell loss. The killing effect of CD38 mAbs on CD38high NK cells should be minimized and the potential combination of CD38low/- NK cells and CD38 mAbs should be maximized to better benefit from their therapeutic efficacy against MM. CD38 mAb effects against MM can be maximized by combination therapies with immunomodulatory imide drugs (IMiDs), proteasome inhibitors (PIs), anti-programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) antibodies, or cellular therapies for the treatment of MM, especially in patients with relapsed or refractory MM (R/R MM) and drug-resistant MM.
Collapse
Affiliation(s)
- Hao-Tian Wu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China
- Collaborative Innovation Center of Hematology, China
| |
Collapse
|
37
|
Clara JA, Levy ER, Reger R, Barisic S, Chen L, Cherkasova E, Chakraborty M, Allan DSJ, Childs R. High-affinity CD16 integration into a CRISPR/Cas9-edited CD38 locus augments CD38-directed antitumor activity of primary human natural killer cells. J Immunother Cancer 2022; 10:jitc-2021-003804. [PMID: 35135865 PMCID: PMC8830298 DOI: 10.1136/jitc-2021-003804] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2021] [Indexed: 11/06/2022] Open
Abstract
Background Adoptive transfer of natural killer (NK) cells with augmented antibody-dependent cellular cytotoxicity (ADCC) capabilities and resistance to CD38 targeting has the potential to enhance the clinical anti-myeloma activity of daratumumab (DARA). Therefore, we sought to develop an efficient CRISPR/Cas9-based gene editing platform to disrupt CD38 expression (CD38 knockout (KO)) in ex vivo expanded NK cells and simultaneously arm CD38KO NK cells with a high-affinity CD16 (CD16-158V) receptor. Methods CD38KO human NK cells were generated using Cas9 ribonucleoprotein complexes. The platform was expanded by incorporating messenger RNA (mRNA) transfection of CD38KO NK cells and targeted gene insertion at the CD38 locus to mediate gene knockin (KI). The capacity of these gene-edited NK cells to persist and mediate ADCC in the presence of DARA was tested in vitro and in a MM.1S xenograft mouse model. Results Highly efficient CD38 gene disruption was achieved in ex vivo expanded NK cells without affecting their proliferative or functional capacity. CD38 KO conferred resistance to DARA-induced NK cell fratricide, enabling persistence and augmented ADCC against myeloma cell lines in the presence of DARA in vitro and in a MM.1S xenograft mouse model. CD38KO NK cells could be further modified by transfection with mRNA encoding a CD16-158V receptor, resulting in augmented DARA-mediated ADCC. Finally, we observed that a homology-directed repair template targeted to the CD38 locus facilitated an efficient 2-in-1 CD38 KO coupled with KI of a truncated CD34 reporter and CD16-158V receptor, with CD38KO/CD16KI NK cells demonstrating a further enhancement of DARA-mediated ADCC both in vitro and in vivo. Conclusions Adoptive immunotherapy using ex vivo expanded CD38KO/CD16KI NK cells has the potential to boost the clinical efficacy of DARA. By incorporating complementary genetic engineering strategies into a CD38 KO manufacturing platform, we generated NK cells with substantially augmented CD38-directed antitumor activity, establishing a strong rationale for exploring this immunotherapy strategy in the clinic.
Collapse
Affiliation(s)
- Joseph Andrew Clara
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Emily R Levy
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA.,Biologics Process Research and Development, Merck & Co Inc, Kenilworth, New Jersey, USA
| | - Robert Reger
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stefan Barisic
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Long Chen
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elena Cherkasova
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mala Chakraborty
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - David S J Allan
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Richard Childs
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
38
|
Schmidt D, Ebrahimabadi S, Gomes KRDS, de Moura Aguiar G, Cariati Tirapelle M, Nacasaki Silvestre R, de Azevedo JTC, Tadeu Covas D, Picanço-Castro V. Engineering CAR-NK cells: how to tune innate killer cells for cancer immunotherapy. IMMUNOTHERAPY ADVANCES 2022; 2:ltac003. [PMID: 35919494 PMCID: PMC9327111 DOI: 10.1093/immadv/ltac003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Cell therapy is an innovative approach that permits numerous possibilities in the field of cancer treatment. CAR-T cells have been successfully used in patients with hematologic relapsed/refractory. However, the need for autologous sources for T cells is still a major drawback. CAR-NK cells have emerged as a promising resource using allogeneic cells that could be established as an off-the-shelf treatment. NK cells can be obtained from various sources, such as peripheral blood (PB), bone marrow, umbilical cord blood (CB), and induced pluripotent stem cells (iPSC), as well as cell lines. Genetic engineering of NK cells to express different CAR constructs for hematological cancers and solid tumors has shown promising preclinical results and they are currently being explored in multiple clinical trials. Several strategies have been employed to improve CAR-NK-cell expansion and cytotoxicity efficiency. In this article, we review the latest achievements and progress made in the field of CAR-NK-cell therapy.
Collapse
Affiliation(s)
- Dayane Schmidt
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sima Ebrahimabadi
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Kauan Ribeiro de Sena Gomes
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Graziela de Moura Aguiar
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Mariane Cariati Tirapelle
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Renata Nacasaki Silvestre
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Júlia Teixeira Cottas de Azevedo
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Virginia Picanço-Castro
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
39
|
Reina-Ortiz C, Giraldos D, Azaceta G, Palomera L, Marzo I, Naval J, Villalba M, Anel A. Harnessing the Potential of NK Cell-Based Immunotherapies against Multiple Myeloma. Cells 2022; 11:cells11030392. [PMID: 35159200 PMCID: PMC8834301 DOI: 10.3390/cells11030392] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cell-based therapies have emerged as promising anticancer treatments due to their potency as cytolytic effectors and synergy with concurrent treatments. Multiple myeloma (MM) is an aggressive B-cell malignancy that, despite development of novel therapeutic agents, remains incurable with a high rate of relapse. In MM, the inhospitable tumor microenvironment prevents host NK cells from exerting their cytolytic function. The development of NK cell immunotherapy works to overcome this altered immune landscape and can be classified in two major groups based on the origin of the cell: autologous or allogeneic. In this review, we compare the treatments in each group, such as autologous chimeric antigen receptor (CAR) NKs and allogeneic off-the-shelf NK cell infusions, and their combinatorial effect with existing MM therapies including monoclonal antibodies and proteasome inhibitors. We also discuss their placement in clinical treatment regimens based on the immune profile of each patient. Through this examination, we would like to discover precisely when each NK cell-based treatment will produce the maximum benefit to the MM patient.
Collapse
Affiliation(s)
- Chantal Reina-Ortiz
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
- Correspondence: (C.R.-O.); (A.A.)
| | - David Giraldos
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
| | - Gemma Azaceta
- Hematology Department, Lozano Blesa Hospital, 50009 Zaragoza, Spain; (G.A.); (L.P.)
| | - Luis Palomera
- Hematology Department, Lozano Blesa Hospital, 50009 Zaragoza, Spain; (G.A.); (L.P.)
| | - Isabel Marzo
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
| | - Javier Naval
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
| | - Martín Villalba
- Institut of Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CNRS, University Hospital Center Montpellier, 34000 Montpellier, France;
- Institut Sainte-Catherine, 84918 Avignon, France
| | - Alberto Anel
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
- Correspondence: (C.R.-O.); (A.A.)
| |
Collapse
|
40
|
Kakiuchi-Kiyota S, Ross T, Wallweber HA, Kiefer JR, Schutten MM, Adedeji AO, Cai H, Hendricks R, Cohen S, Myneni S, Liu L, Fullerton A, Corr N, Yu L, de Almeida Nagata D, Zhong S, Leong SR, Li J, Nakamura R, Sumiyoshi T, Li J, Ovacik AM, Zheng B, Dillon M, Spiess C, Wingert S, Rajkovic E, Ellwanger K, Reusch U, Polson AG. A BCMA/CD16A bispecific innate cell engager for the treatment of multiple myeloma. Leukemia 2022; 36:1006-1014. [PMID: 35001074 DOI: 10.1038/s41375-021-01478-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/05/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022]
Abstract
Despite the recent progress, multiple myeloma (MM) is still essentially incurable and there is a need for additional effective treatments with good tolerability. RO7297089 is a novel bispecific BCMA/CD16A-directed innate cell engager (ICE®) designed to induce BCMA+ MM cell lysis through high affinity binding of CD16A and retargeting of NK cell cytotoxicity and macrophage phagocytosis. Unlike conventional antibodies approved in MM, RO7297089 selectively targets CD16A with no binding of other Fcγ receptors, including CD16B on neutrophils, and irrespective of 158V/F polymorphism, and its activity is less affected by competing IgG suggesting activity in the presence of M-protein. Structural analysis revealed this is due to selective interaction with a single residue (Y140) uniquely present in CD16A opposite the Fc binding site. RO7297089 induced tumor cell killing more potently than conventional antibodies (wild-type and Fc-enhanced) and induced lysis of BCMA+ cells at very low effector-to-target ratios. Preclinical toxicology data suggested a favorable safety profile as in vitro cytokine release was minimal and no RO7297089-related mortalities or adverse events were observed in cynomolgus monkeys. These data suggest good tolerability and the potential of RO7297089 to be a novel effective treatment of MM patients.
Collapse
Affiliation(s)
| | | | | | - James R Kiefer
- Genentech Research and Early Development, San Francisco, CA, USA
| | | | | | - Hao Cai
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Robert Hendricks
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Sivan Cohen
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Srividya Myneni
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Luna Liu
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Aaron Fullerton
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Nicholas Corr
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Lanlan Yu
- Genentech Research and Early Development, San Francisco, CA, USA
| | | | - Shelly Zhong
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Steven R Leong
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Ji Li
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Rin Nakamura
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Teiko Sumiyoshi
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Jinze Li
- Genentech Research and Early Development, San Francisco, CA, USA
| | | | - Bing Zheng
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Mike Dillon
- Genentech Research and Early Development, San Francisco, CA, USA
| | - Christoph Spiess
- Genentech Research and Early Development, San Francisco, CA, USA
| | | | | | | | | | - Andrew G Polson
- Genentech Research and Early Development, San Francisco, CA, USA.
| |
Collapse
|
41
|
Clara JA, Childs RW. Harnessing natural killer cells for the treatment of multiple myeloma. Semin Oncol 2022; 49:69-85. [DOI: 10.1053/j.seminoncol.2022.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 01/08/2022] [Indexed: 12/11/2022]
|
42
|
Hill E, Morrison C, Kazandjian D. Daratumumab: A review of current indications and future directions. Semin Oncol 2022; 49:48-59. [DOI: 10.1053/j.seminoncol.2022.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/09/2022] [Indexed: 12/17/2022]
|
43
|
Cho SF, Xing L, Anderson KC, Tai YT. Promising Antigens for the New Frontier of Targeted Immunotherapy in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13236136. [PMID: 34885245 PMCID: PMC8657018 DOI: 10.3390/cancers13236136] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Defining the specificity and biological sequalae induced by receptors differentiated expressed in multiple myeloma cells are critical for the development of effective immunotherapies based on monoclonal antibodies. Ongoing studies continue to discover new antigens with superior tumor selectivity and defined function in regulating the pathophysiology of myeloma cells directly or indirectly in the immunosuppressive bone marrow microenvironment. Meanwhile, it is urgent to identify mechanisms of immune resistance and design more potent immunotherapies, alone and/or with best combination partners to further prolong anti-MM immunity. Abstract The incorporation of novel agents in recent treatments in multiple myeloma (MM) has improved the clinical outcome of patients. Specifically, the approval of monoclonal antibody (MoAb) against CD38 (daratumumab) and SLAMF7 (elotuzumab) in relapsed and refractory MM (RRMM) represents an important milestone in the development of targeted immunotherapy in MM. These MoAb-based agents significantly induce cytotoxicity of MM cells via multiple effector-dependent mechanisms and can further induce immunomodulation to repair a dysfunctional tumor immune microenvironment. Recently, targeting B cell maturation antigen (BCMA), an even MM-specific antigen, has shown high therapeutic activities by chimeric antigen receptor T cells (CAR T), antibody-drug conjugate (ADC), bispecific T-cell engager (BiTE), as well as bispecific antibody (BiAb), with some already approved for heavily pretreated RRMM patients. New antigens, such as orphan G protein-coupled receptor class C group 5 member D (GPRC5D) and FcRH5, were identified and rapidly moved to ongoing clinical studies. We here summarized the pathobiological function of key MM antigens and the status of the corresponding immunotherapies. The potential challenges and emerging treatment strategies are also discussed.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
- 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
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Lijie Xing
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China;
| | - Kenneth C. Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
- Correspondence: ; Tel.: +1-617-632-3875; Fax: +1-617-632-2140
| |
Collapse
|
44
|
Kastritis E, Rousakis P, Kostopoulos IV, Gavriatopoulou M, Theodorakakou F, Fotiou D, Dialoupi I, Migkou M, Roussou M, Kanellias N, Tselegkidi MI, Eleutherakis-Papaiakovou E, Papanikolaou A, Gakiopoulou C, Psimenou E, Spyropoulou-Vlachou M, Gatou A, Terpos E, Tsitsilonis O, Dimopoulos MA. Consolidation with a short course of daratumumab in patients with AL amyloidosis or light chain deposition disease. Amyloid 2021; 28:259-266. [PMID: 34468250 DOI: 10.1080/13506129.2021.1971192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Daratumumab has major and rapid activity in AL amyloidosis with favourable toxicity. We used as a consolidation a short course of daratumumab in 25 patients with AL amyloidosis or light chain deposition disease (LCDD), who had not achieved a haematologic complete response (hemCR) after standard therapy with bortezomib, cyclophosphamide and dexamethasone (VCD). We evaluated minimal residual disease (MRD) and changes in the bone marrow (BM) microenvironment before and after consolidation using next generation flow cytometry (NGF). At the time of consolidation, 21 patients were in very good partial response (VGPR) and four in partial response (PR); all had detectable MRD. One month after consolidation completion, 8 patients (32%) achieved a hemCR, of whom 5 (20%) became also MRD negative. In the BM, we observed significant changes in B-cell precursors, naïve B-cells, T-cells, CD27+ NK & NKT cells, mast cells and erythroblasts. After a median follow-up of 25 months, none of the patients in hemCR has relapsed and all have achieved an organ response; a haematologic relapse occurred in 6/17 patients that did not achieve hemCR. In conclusion, consolidation with a short course of daratumumab can improve depth of response in patients with AL amyloidosis or LCDD and significantly affects BM environment.
Collapse
Affiliation(s)
- Efstathios Kastritis
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Pantelis Rousakis
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis V Kostopoulos
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Foteini Theodorakakou
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Fotiou
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Dialoupi
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Magdalini Migkou
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Roussou
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kanellias
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Irini Tselegkidi
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Charikleia Gakiopoulou
- 1st Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece
| | - Erasmia Psimenou
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Evangelos Terpos
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Ourania Tsitsilonis
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios A Dimopoulos
- Plasma Cell Dyscrasia Unit, Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
45
|
Brauneck F, Seubert E, Wellbrock J, Schulze zur Wiesch J, Duan Y, Magnus T, Bokemeyer C, Koch-Nolte F, Menzel S, Fiedler W. Combined Blockade of TIGIT and CD39 or A2AR Enhances NK-92 Cell-Mediated Cytotoxicity in AML. Int J Mol Sci 2021; 22:ijms222312919. [PMID: 34884723 PMCID: PMC8657570 DOI: 10.3390/ijms222312919] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 01/17/2023] Open
Abstract
This study aimed to characterize different natural killer (NK) cell phenotypes on bone marrow and peripheral blood cells from acute myeloid leukemia (AML) patients and healthy donors (HDs). Our data show that CD56dimCD16− and CD56brightCD16− NK cells represent the predominant NK cell subpopulations in AML, while the CD56dimCD16+ NK cells are significantly reduced compared to HDs. Moreover, TIGIT+ and PVRIG+ cells cluster on the CD56dimCD16+ subset whereas CD39+ and CD38+ cells do so on CD56brightCD16− NK cells in AML. Furthermore, functional effects of (co-)blockade of TIGIT and CD39 or A2AR on NK cell functionality were analyzed. These experiments revealed that the single blockade of the TIGIT receptor results in an increased NK-92 cell-mediated killing of AML cells in vitro. Combined targeting of CD39 or A2AR significantly augments the anti-TIGIT-mediated lysis of AML cells. Our data indicate that distinct NK cell subsets in AML exhibit different immunosuppressive patterns (via the TIGIT/PVRIG receptors and the purinergic pathway). In summary, we conclude that TIGIT, CD39, and A2AR constitute relevant inhibitory checkpoints of NK cells in AML patients. A combinatorial blockade synergistically strengthens NK-92 cell-mediated cytotoxicity. As inhibitors of TIGIT, CD39, and A2AR are clinically available, studies on their combined use could be conducted in the near future.
Collapse
Affiliation(s)
- Franziska Brauneck
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.B.); (E.S.); (J.W.); (C.B.)
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
| | - Elisa Seubert
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.B.); (E.S.); (J.W.); (C.B.)
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.B.); (E.S.); (J.W.); (C.B.)
| | - Julian Schulze zur Wiesch
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
| | - Yinghui Duan
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (Y.D.); (T.M.)
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (Y.D.); (T.M.)
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.B.); (E.S.); (J.W.); (C.B.)
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
| | - Stephan Menzel
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.B.); (E.S.); (J.W.); (C.B.)
- Correspondence:
| |
Collapse
|
46
|
Janakiram M, Arora N, Bachanova V, Miller JS. Novel Cell and Immune Engagers in Optimizing Tumor- Specific Immunity Post-Autologous Transplantation in Multiple Myeloma. Transplant Cell Ther 2021; 28:61-69. [PMID: 34634499 DOI: 10.1016/j.jtct.2021.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/07/2021] [Accepted: 10/03/2021] [Indexed: 11/18/2022]
Abstract
Autologous stem cell transplantation (ASCT) is an important component of treatment of multiple myeloma (MM). The post-ASCT setting offers a unique opportunity to increase myeloma specific immunity through enhancement of T and NK cell responses. The vast array of therapeutics being developed for MM, including cell-based therapies, dendritic vaccines, bispecific antibodies, and IL-15 agonists, provide the opportunity to increase tumor-specific immunity. Maintenance therapies, including immunomodulatory drugs, proteasome inhibitors, and daratumumab, exhibit a significant anti-myeloma response by modulating the immune system. Lenalidomide promotes an antitumoral immune microenvironment, whereas daratumumab can potentially cause NK cell fratricide. Thus, understanding the effects of commonly used maintenance drugs on the immune system is important. In this review, we look at current and emerging therapeutics and their integration post-ASCT in the context of immune reconstitution to improve clinical responses in patients with MM. © 2021 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc.
Collapse
Affiliation(s)
- Murali Janakiram
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.
| | - Nivedita Arora
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Veronika Bachanova
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey S Miller
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
47
|
Nakamura A, Suzuki S, Kanasugi J, Ejiri M, Hanamura I, Ueda R, Seto M, Takami A. Synergistic Effects of Venetoclax and Daratumumab on Antibody-Dependent Cell-Mediated Natural Killer Cytotoxicity in Multiple Myeloma. Int J Mol Sci 2021; 22:ijms221910761. [PMID: 34639102 PMCID: PMC8509545 DOI: 10.3390/ijms221910761] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 02/06/2023] Open
Abstract
The prognosis of multiple myeloma (MM) has drastically improved owing to the development of new drugs, such as proteasome inhibitors and immunomodulatory drugs. Nevertheless, MM is an extremely challenging disease, and many patients are still refractory to the existing therapies, thus requiring new treatment alternatives. Venetoclax is a selective, orally bioavailable inhibitor of BCL-2 that shows efficacy in MM not only as a single agent but also in combination therapy, especially for MM patients with translocation t(11;14). However, many patients are refractory to this drug. Here, we treated the MM cell lines KMS12PE and KMS27 with a combination treatment of venetoclax targeting BCL-2 and daratumumab targeting CD38 to evaluate the synergistic cytotoxicity of these drugs in vitro. MM cell lines were co-cultured with natural killer (NK) cells at an effector:target ratio of 0.3:1 in the presence of serial concentrations of daratumumab and venetoclax, and the resulting apoptotic MM cells were detected by flow cytometry using annexin V. These results indicated that the antibody-dependent cell-mediated NK cytotoxicity was enhanced in KMS12PE and KMS27 cells harboring t(11;14) with a high BCL-2 expression, suggesting that the combination treatment of venetoclax and daratumumab should be especially effective in patients with these characteristics.
Collapse
Affiliation(s)
- Ayano Nakamura
- Department of Internal Medicine, Division of Hematology, Aichi Medical University School of Medicine, Nagakute 480-1195, Japan; (A.N.); (J.K.); (I.H.); (M.S.); (A.T.)
| | - Susumu Suzuki
- Research Creation Support Center, Aichi Medical University, Nagakute 480-1195, Japan
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute 480-1195, Japan;
- Correspondence: ; Tel.: +81-561-62-3311 (ext. 11426)
| | - Jo Kanasugi
- Department of Internal Medicine, Division of Hematology, Aichi Medical University School of Medicine, Nagakute 480-1195, Japan; (A.N.); (J.K.); (I.H.); (M.S.); (A.T.)
| | - Masayuki Ejiri
- Department of Pharmacy, University Hospital, Aichi Medical University, Nagakute 480-1195, Japan;
| | - Ichiro Hanamura
- Department of Internal Medicine, Division of Hematology, Aichi Medical University School of Medicine, Nagakute 480-1195, Japan; (A.N.); (J.K.); (I.H.); (M.S.); (A.T.)
| | - Ryuzo Ueda
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute 480-1195, Japan;
| | - Masao Seto
- Department of Internal Medicine, Division of Hematology, Aichi Medical University School of Medicine, Nagakute 480-1195, Japan; (A.N.); (J.K.); (I.H.); (M.S.); (A.T.)
| | - Akiyoshi Takami
- Department of Internal Medicine, Division of Hematology, Aichi Medical University School of Medicine, Nagakute 480-1195, Japan; (A.N.); (J.K.); (I.H.); (M.S.); (A.T.)
| |
Collapse
|
48
|
Suzuki K, Nishiwaki K, Yano S. Treatment Strategy for Multiple Myeloma to Improve Immunological Environment and Maintain MRD Negativity. Cancers (Basel) 2021; 13:4867. [PMID: 34638353 PMCID: PMC8508145 DOI: 10.3390/cancers13194867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 02/06/2023] Open
Abstract
Improving the immunological environment and eradicating minimal residual disease (MRD) are the two main treatment goals for long-term survival in patients with multiple myeloma (MM). Immunomodulatory drugs (IMiDs), monoclonal antibody drugs (MoAbs), and autologous grafts for autologous stem cell transplantation (ASCT) can improve the immunological microenvironment. ASCT, MoAbs, and proteasome inhibitors (PIs) may be important for the achievement of MRD negativity. An improved immunological environment may be useful for maintaining MRD negativity, although the specific treatment for persistent MRD negativity is unknown. However, whether the ongoing treatment should be continued or changed if the MRD status remains positive is controversial. In this case, genetic, immunophenotypic, and clinical analysis of residual myeloma cells may be necessary to select the effective treatment for the residual myeloma cells. The purpose of this review is to discuss the MM treatment strategy to "cure MM" based on currently available therapies, including IMiDs, PIs, MoAbs, and ASCT, and expected immunotherapies, such as chimeric antigen receptor T cell (CAR-T) therapy, via improvement of the immunological environment and maintenance of MRD negativity.
Collapse
Affiliation(s)
- Kazuhito Suzuki
- Department of Internal Medicine, Division of Clinical Oncology and Hematology, The Jikei University Kashiwa Hospital, Tokyo 277-8567, Japan;
- Department of Internal Medicine, Division of Clinical Oncology and Hematology, The Jikei University School of Medicine, Tokyo 105-8461, Japan;
| | - Kaichi Nishiwaki
- Department of Internal Medicine, Division of Clinical Oncology and Hematology, The Jikei University Kashiwa Hospital, Tokyo 277-8567, Japan;
- Department of Internal Medicine, Division of Clinical Oncology and Hematology, The Jikei University School of Medicine, Tokyo 105-8461, Japan;
| | - Shingo Yano
- Department of Internal Medicine, Division of Clinical Oncology and Hematology, The Jikei University School of Medicine, Tokyo 105-8461, Japan;
| |
Collapse
|
49
|
Szudy-Szczyrek A, Ahern S, Kozioł M, Majowicz D, Szczyrek M, Krawczyk J, Hus M. Therapeutic Potential of Innate Lymphoid Cells for Multiple Myeloma Therapy. Cancers (Basel) 2021; 13:4806. [PMID: 34638291 PMCID: PMC8507621 DOI: 10.3390/cancers13194806] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 01/08/2023] Open
Abstract
Innate lymphoid cells (ILCs) are a recently identified family of lymphocyte-like cells lacking a specific antigen receptor. They are part of the innate immune system. They play a key role in tissue homeostasis and also control inflammatory and neoplastic processes. In response to environmental stimuli, ILCs change their phenotype and functions, and influence the activity of other cells in the microenvironment. ILC dysfunction can lead to a wide variety of diseases, including cancer. ILC can be divided into three subgroups: ILC Group 1, comprising NK cells and ILC1; Group 2, including ILC2 alone; and Group 3, containing Lymphoid Tissue inducers (LTi) and ILC3 cells. While Group 1 ILCs mainly exert antitumour activity, Group 2 and Group 3 ILCs are protumorigenic in nature. A growing body of preclinical and clinical data support the role of ILCs in the pathogenesis of multiple myeloma (MM). Therefore, targeting ILCs may be of clinical benefit. In this manuscript, we review the available data on the role of ILCs in MM immunology and therapy.
Collapse
Affiliation(s)
- Aneta Szudy-Szczyrek
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland; (M.K.); (D.M.)
| | - Sean Ahern
- Department of Haematology, University Hospital Galway, H91 TK33 Galway, Ireland; (S.A.); (J.K.)
- National University of Ireland, H91 TK33 Galway, Ireland
| | - Magdalena Kozioł
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland; (M.K.); (D.M.)
| | - Daria Majowicz
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland; (M.K.); (D.M.)
| | - Michał Szczyrek
- Chair and Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Janusz Krawczyk
- Department of Haematology, University Hospital Galway, H91 TK33 Galway, Ireland; (S.A.); (J.K.)
- National University of Ireland, H91 TK33 Galway, Ireland
| | - Marek Hus
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland; (M.K.); (D.M.)
| |
Collapse
|
50
|
Baum N, Eggers M, Koenigsdorf J, Menzel S, Hambach J, Staehler T, Fliegert R, Kulow F, Adam G, Haag F, Bannas P, Koch-Nolte F. Mouse CD38-Specific Heavy Chain Antibodies Inhibit CD38 GDPR-Cyclase Activity and Mediate Cytotoxicity Against Tumor Cells. Front Immunol 2021; 12:703574. [PMID: 34539634 PMCID: PMC8446682 DOI: 10.3389/fimmu.2021.703574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022] Open
Abstract
CD38 is the major NAD+-hydrolyzing ecto-enzyme in most mammals. As a type II transmembrane protein, CD38 is also a promising target for the immunotherapy of multiple myeloma (MM). Nanobodies are single immunoglobulin variable domains from heavy chain antibodies that naturally occur in camelids. Using phage display technology, we isolated 13 mouse CD38-specific nanobodies from immunized llamas and produced these as recombinant chimeric mouse IgG2a heavy chain antibodies (hcAbs). Sequence analysis assigned these hcAbs to five distinct families that bind to three non-overlapping epitopes of CD38. Members of families 4 and 5 inhibit the GDPR-cyclase activity of CD38. Members of families 2, 4 and 5 effectively induce complement-dependent cytotoxicity against CD38-expressing tumor cell lines, while all families effectively induce antibody dependent cellular cytotoxicity. Our hcAbs present unique tools to assess cytotoxicity mechanisms of CD38-specific hcAbs in vivo against tumor cells and potential off-target effects on normal cells expressing CD38 in syngeneic mouse tumor models, i.e. in a fully immunocompetent background.
Collapse
Affiliation(s)
- Natalie Baum
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marie Eggers
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Koenigsdorf
- 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.,Mildred Scheel Cancer Career Center HaTriCS4, 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
| | - Tobias Staehler
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Fliegert
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederike Kulow
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Adam
- Department of Radiology, 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
|