1
|
Liu Y, Tao Y, Yeung PSW, Lu M, Liu J, Yu F, Shi RZ, Luo RY. An up-conversion fluorescence lateral-flow immunoassay for rapid detection of Daratumumab in serum protein electrophoresis clinical samples. Clin Chim Acta 2024; 559:119677. [PMID: 38636694 DOI: 10.1016/j.cca.2024.119677] [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: 01/09/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Daratumumab (DARA) is a commonly used monoclonal antibody (mAb) drug for the treatment of multiple myeloma (MM). Its appearance as a visible abnormal band in the γ-region of a serum protein electrophoresis (SPEP) gel may interfere with the SPEP result interpretation. With the advantages of portability and rapid testing capabilities, up-conversion fluorescence lateral-flow immunoassay (LFA) can be an ideal solution to detect DARA interference. METHODS An up-conversion fluorescence LFA strip was designed and constructed to perform semi-quantitative DARA testing in clinical samples. The LFA strip test was evaluated for limit of detection (LOD), dynamic range, and analytical interference. RESULTS To demonstrate the clinical utility of the LFA strip, 43 SPEP-positive patient serum samples were tested for the presence of DARA, and the results exactly matched the DARA usage history in patient medical records. CONCLUSIONS The performance of the up-conversion fluorescence LFA strip meets the purpose of clarifying DARA interference in SPEP results. It may be used as an independent and objective confirmation of the presence of DARA in clinical samples. The LFA strip offers a cost-effective rapid on-site test to check for DARA interference alongside standard SPEP equipment, which significantly improves the interpretation of ambiguous SPEP results involving DARA, and does not intervene the current SPEP workflow in clinical laboratory practice.
Collapse
Affiliation(s)
- Yajing Liu
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Yanru Tao
- Polariton Life Technologies, Suzhou, Jiangsu, China
| | - Priscilla S-W Yeung
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA; Clinical Laboratories, Stanford Health Care, Palo Alto, CA, USA
| | - Mengyan Lu
- Polariton Life Technologies, Suzhou, Jiangsu, China
| | - Jing Liu
- Polariton Life Technologies, Suzhou, Jiangsu, China
| | - Fang Yu
- Polariton Life Technologies, Suzhou, Jiangsu, China
| | - Run-Zhang Shi
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA; Clinical Laboratories, Stanford Health Care, Palo Alto, CA, USA
| | - Ruben Yiqi Luo
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA; Clinical Laboratories, Stanford Health Care, Palo Alto, CA, USA.
| |
Collapse
|
2
|
Liu W, Yu J, Sun K, Song Q, Li Y, He Y, Wang Y, Xu G, Wang C, Chen B. Preclinical characterization of a novel investigational monoclonal antibody CM313 with potent CD38-positive cell killing activity. Front Immunol 2024; 15:1410457. [PMID: 38765013 PMCID: PMC11099226 DOI: 10.3389/fimmu.2024.1410457] [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: 04/01/2024] [Accepted: 04/19/2024] [Indexed: 05/21/2024] Open
Abstract
Introduction CM313 is currently under clinical investigation for treatments of multiple myeloma, systemic lupus erythematosus, and immune thrombocytopenia. We aimed to report the preclinical profile of the novel therapeutic anti-CD38 monoclonal antibody (mAb) CM313, with an emphasis on the difference with other CD38-targeting mAb. Methods The binding of CM313 to CD38 recombinant protein across species was assessed using ELISA. The binding of CM313 to CD38-positive (CD38+) cells was detected using flow cytometry assays. CM313-induced complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and apoptosis on different CD38+ cells were assessed by LDH release assays or flow cytometry assays. The effect of CM313 on CD38 enzymatic activity was measured using fluorescence spectroscopy. CM313 immunotoxicity in human blood was assessed using flow cytometry assays, ELISA, and LDH release assays. Anti-tumor activity of CM313 was assessed in multiple mouse xenograft models. Safety profile of CM313 were evaluated in cynomolgus monkeys and human CD38 transgenic (B-hCD38) mice. Results There exist unique sequences at complementarity-determining regions (CDR) of CM313, which facilitates its affinity to CD38 is consistently higher across a spectrum of CD38+ cell lines than daratumumab. In vitro studies showed that CM313 induces comparable killing activity than daratumumab, including ADCC, CDC, ADCP, apoptosis induced by Fc-mediated cross-linking, and effectively inhibited the enzymatic activity of CD38. However, CM313 showed more potent CDC than isatuximab. In vivo, CM313 dose-dependently inhibited xenograft tumor growth, both as a monotherapy and in combination with dexamethasone or lenalidomide. Furthermore, CM313 was well tolerated with no drug-related clinical signs or off-target risks, as evidenced by 4-week repeat-dose toxicology studies in cynomolgus monkeys and B-hCD38 mice, with the later study showing no observed adverse effect level (NOAEL) of 300mg/kg once weekly. Discussion CM313 is a novel investigational humanized mAb with a distinct CDR sequence, showing comparable killing effects with daratumumab and stronger CDC activity than isatuximab, which supports its clinical development.
Collapse
Affiliation(s)
- Wei Liu
- Research and Development Department, Keymed Biosciences (Chengdu) Limited, Chengdu, China
| | - Juntao Yu
- Research and Development Department, Keymed Biosciences (Chengdu) Limited, Chengdu, China
| | - Kaiwen Sun
- Department of Ecology and Evolutionary Biology, Tulane University, New Orleans, MS, United States
| | - Qin Song
- Research and Development Department, Keymed Biosciences (Chengdu) Limited, Chengdu, China
| | - Yuling Li
- Research and Development Department, Keymed Biosciences (Chengdu) Limited, Chengdu, China
| | - Yanyun He
- Research and Development Department, Keymed Biosciences (Chengdu) Limited, Chengdu, China
| | - Yanrong Wang
- Research and Development Department, Keymed Biosciences (Chengdu) Limited, Chengdu, China
| | - Gang Xu
- Research and Development Department, Keymed Biosciences (Chengdu) Limited, Chengdu, China
| | - Changyu Wang
- Research and Development Department, Keymed Biosciences (Chengdu) Limited, Chengdu, China
| | - Bo Chen
- Research and Development Department, Keymed Biosciences (Chengdu) Limited, Chengdu, China
| |
Collapse
|
3
|
Haran A, Vaxman I, Gatt ME, Lebel E. Immune Therapies in AL Amyloidosis-A Glimpse to the Future. Cancers (Basel) 2024; 16:1605. [PMID: 38672686 PMCID: PMC11048972 DOI: 10.3390/cancers16081605] [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: 03/20/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Light-chain (AL) amyloidosis is a rare plasma cell disorder characterized by the deposition of misfolded immunoglobulin light chains in target organs, leading to multi-organ dysfunction. Treatment approaches have historically mirrored but lagged behind those of multiple myeloma (MM). Recent advancements in MM immunotherapy are gradually being evaluated and adopted in AL amyloidosis. This review explores the current state of immunotherapeutic strategies in AL amyloidosis, including monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T-cell therapy. We discuss the unique challenges and prospects of these therapies in AL amyloidosis, including the exposure of frail AL amyloidosis patients to immune-mediated toxicities such as cytokine release syndrome (CRS) and immune effector-cell-associated neurotoxicity syndrome (ICANS), as well as their efficacy in promoting rapid and deep hematologic responses. Furthermore, we highlight the need for international initiatives and compassionate programs to provide access to these promising therapies and address critical unmet needs in AL amyloidosis management. Finally, we discuss future directions, including optimizing treatment sequencing and mitigating toxicities, to improve outcomes for AL amyloidosis patients.
Collapse
Affiliation(s)
- Arnon Haran
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; (A.H.); (M.E.G.)
| | - Iuliana Vaxman
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva 49100, Israel;
| | - Moshe E. Gatt
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; (A.H.); (M.E.G.)
| | - Eyal Lebel
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; (A.H.); (M.E.G.)
| |
Collapse
|
4
|
de Moraes FCA, Sano VKT, Lôbo ADOM, Kelly FA, Morbach V, Pasqualotto E, Burbano RMR. Efficacy and Safety of Anti-CD38 Monoclonal Antibodies in Patients with Relapsed or Refractory Multiple Myeloma: A Meta-Analysis of Randomized Clinical Trials. J Pers Med 2024; 14:360. [PMID: 38672988 PMCID: PMC11051236 DOI: 10.3390/jpm14040360] [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: 02/16/2024] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
The benefit of associating anti-CD38 monoclonal antibodies to proteasome inhibitor (PI)/immunomodulatory agent (IA) and dexamethasone in the treatment of patients with relapsed or refractory multiple myeloma (MM) remains unclear. PubMed, Embase, and Cochrane Library databases were searched for randomized controlled trials that investigated the addition of anti-CD38 monoclonal antibodies to a therapy composed of PI/IA and dexamethasone versus PI/IA and dexamethasone alone for treating relapsed or refractory MM. Hazard ratios (HRs) or risk ratios (RRs) were computed for binary endpoints, with 95% confidence intervals (CIs). Six studies comprising 2191 patients were included. Anti-CD38 monoclonal antibody significantly improved progression-free survival (HR 0.52; 95% CI 0.43-0.61; p < 0.001) and overall survival (HR 0.72; 95% CI 0.63-0.83; p < 0.001). There was a significant increase in hematological adverse events, such as neutropenia (RR 1.41; 95% CI 1.26-1.58; p < 0.01) and thrombocytopenia (RR 1.14; 95% CI 1.02-1.27; p = 0.02), in the group treated with anti-CD38 monoclonal antibody. Also, there was a significant increase in non-hematological adverse events, such as dyspnea (RR 1.72; 95% CI 1.38-2.13; p < 0.01) and pneumonia (RR 1.34; 95% CI 1.13-1.59; p < 0.01), in the group treated with anti-CD38 monoclonal antibody. In conclusion, the incorporation of an anti-CD38 monoclonal antibody demonstrated a promising prospect for reshaping the established MM treatment paradigms.
Collapse
Affiliation(s)
| | | | | | | | - Victória Morbach
- Department of Medicine, Feevale University, Novo Hamburgo 93510-235, Brazil;
| | - Eric Pasqualotto
- Department of Medicine, Federal University of Santa Catarina, Florianopolis 88040-900, Brazil;
| | | |
Collapse
|
5
|
Cui L, Ning J, Yang R, Wang H. Therapeutic Effect and Adverse Event Rate of Different Treatment Methods in Patients with Multiple Myeloma and Renal Insufficiency. Int Arch Allergy Immunol 2024; 185:659-667. [PMID: 38467118 DOI: 10.1159/000536170] [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: 11/23/2023] [Accepted: 01/04/2024] [Indexed: 03/13/2024] Open
Abstract
INTRODUCTION This study involves the collation and analysis of clinical characteristics and laboratory findings in patients with multiple myeloma (MM) combined with renal insufficiency. The objective was to assess the impact of various treatment methods on patient outcomes and the incidence of adverse events in individuals with MM and renal insufficiency. METHODS We analyzed the correlation between clinical characteristics, gene loci, fluorescence in situ hybridization, treatment methods, and prognosis in patients with MM and renal insufficiency. The differences in hematological and therapeutic efficacy indexes between two groups subjected to different treatments were evaluated. The assessment of treatment effectiveness was based on the total effective rate, calculated as the sum of stringent CR rate, complete remission rate, very good partial remission rate, and partial remission rate. RESULTS (1) The renal insufficiency group exhibited higher percentages of bone marrow abnormal plasma cells, lactate dehydrogenase (LDH), blood calcium, white blood cell count, percentage of neutrophils, and blood β2-microglobulin (β2-MG) levels compared to the normal renal function group. Conversely, hemoglobin levels and lymphocyte percentage were lower in the renal insufficiency group. Binary logistic regression analysis identified hemoglobin, blood calcium values, blood β2-MG, and LDH as independent risk factors for the development of renal insufficiency in patients with MM (p < 0.05). (2) Based on the Durie-Salmon staging criteria, the proportion of Stage III patients was the highest (up to 81.8%), indicating that patients with MM usually suffer from insidious disease, often with high tumor load and late-disease stage at the time of consultation. International Staging System (ISS) and Revised ISS staging also revealed a higher proportion of Stage III patients in the renal insufficiency group (p < 0.05), indicating a worse long-term prognosis in patients with MM and renal insufficiency. (3) Before treatment, there was no significant difference between the two groups in the analysis of various indices. Complications such as sepsis, herpes zoster, peripheral neuropathy, thrombosis, secondary pulmonary infection, and cardiac complications were significantly lower in the BCD group (Bortezomib + Cyclophosphamide + Dexamethasone) compared to the BD group (Bortezomib + Dexamethasone) (χ2 = 6.333, p < 0.05), suggesting fewer complications with the BCD regimen. (4) The clinical treatment effects analysis indicated that the BCD group demonstrated a more significant impact than the BD group in the treatment of MM. CONCLUSION The application of the BCD regimen in the treatment of MM has shown significant efficiency, effectively alleviating clinical symptoms with fewer adverse reactions and high safety.
Collapse
Affiliation(s)
- Lijuan Cui
- Department of Hematology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jing Ning
- Department of Hematology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Rui Yang
- Department of Hematology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hainan Wang
- Department of Hematology, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
6
|
Del Dosso A, Tadevosyan E, Berenson JR. Preclinical and clinical evaluation of the Janus Kinase inhibitor ruxolitinib in multiple myeloma. Oncotarget 2024; 15:65-75. [PMID: 38319731 PMCID: PMC10852065 DOI: 10.18632/oncotarget.28547] [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: 11/27/2023] [Accepted: 12/18/2023] [Indexed: 02/08/2024] Open
Abstract
Multiple myeloma (MM) is the most common primary malignancy of the bone marrow. No established curative treatment is currently available for patients diagnosed with MM. In recent years, new and more effective drugs have become available for the treatment of this B-cell malignancy. These new drugs have often been evaluated together and in combination with older agents. However, even these novel combinations eventually become ineffective; and, thus, novel therapeutic approaches are necessary to help overcome resistance to these treatments. Recently, the Janus Kinase (JAK) family of tyrosine kinases, specifically JAK1 and JAK2, has been shown to have a role in the pathogenesis of MM. Preclinical studies have demonstrated a role for JAK signaling in direct and indirect growth of MM and downregulation of anti-tumor immune responses in these patients. Also, inhibition of JAK proteins enhances the anti-MM effects of other drugs used to treat MM. These findings have been confirmed in clinical studies which have further demonstrated the safety and efficacy of JAK inhibition as a means to overcome resistance to currently available anti-MM therapies. Additional studies will provide further support for this promising new therapeutic approach for treating patients with MM.
Collapse
Affiliation(s)
- Ashley Del Dosso
- ONCOtherapeutics, West Hollywood, CA 90069, USA
- These authors contributed equally to this work
| | - Elizabeth Tadevosyan
- Berenson Cancer Center, West Hollywood, CA 90069, USA
- These authors contributed equally to this work
| | - James R. Berenson
- ONCOtherapeutics, West Hollywood, CA 90069, USA
- Berenson Cancer Center, West Hollywood, CA 90069, USA
- Institute for Myeloma and Bone Cancer Research, West Hollywood, CA 90069, USA
| |
Collapse
|
7
|
Yu Z, Li H, Lu Q, Zhang Z, Tong A, Niu T. Fc receptor-like 5 (FCRL5)-directed CAR-T cells exhibit antitumor activity against multiple myeloma. Signal Transduct Target Ther 2024; 9:16. [PMID: 38212320 PMCID: PMC10784595 DOI: 10.1038/s41392-023-01702-2] [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: 05/06/2023] [Revised: 10/11/2023] [Accepted: 11/08/2023] [Indexed: 01/13/2024] Open
Abstract
Multiple myeloma (MM) remains a challenging hematologic malignancy despite advancements in chimeric antigen receptor T-cell (CAR-T) therapy. Current targets of CAR-T cells used in MM immunotherapy have limitations, with a subset of patients experiencing antigen loss resulting in relapse. Therefore, novel targets for enhancing CAR-T cell therapy in MM remain needed. Fc receptor-like 5 (FCRL5) is a protein marker with considerably upregulated expression in MM and has emerged as a promising target for CAR-T cell therapeutic interventions, offering an alternative treatment for MM. To further explore this option, we designed FCRL5-directed CAR-T cells and assessed their cytotoxicity in vitro using a co-culture system and in vivo using MM cell-derived xenograft models, specifically focusing on MM with gain of chromosome 1q21. Given the challenges in CAR-T therapies arising from limited T cell persistence, our approach incorporates interleukin-15 (IL-15), which enhances the functionality of central memory T (TCM) cells, into the design of FCRL5-directed CAR-T cells, to improve cytotoxicity and reduce T-cell dysfunction, thereby promoting greater CAR-T cell survival and efficacy. Both in vitro and xenograft models displayed that FCRL5 CAR-T cells incorporating IL-15 exhibited potent antitumor efficacy, effectively inhibiting the proliferation of MM cells and leading to remarkable tumor suppression. Our results highlight the capacity of FCRL5-specific CAR-T cells with the integration of IL-15 to improve the therapeutic potency, suggesting a potential novel immunotherapeutic strategy for MM treatment.
Collapse
Affiliation(s)
- Zhengyu Yu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hexian Li
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qizhong Lu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongliang Zhang
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Aiping Tong
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ting Niu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
Gil‐Fernández JJ, García Ramírez P, Callejas Charavía M. Isatuximab-carfilzomib-dexamethasone immediately after failing of the quadruplet Daratumumab-bortezomib-lenalidomide-dexamethasone (Dara-VRD): Striking response with no washout in a newly diagnosed multiple myeloma. Clin Case Rep 2024; 12:e8449. [PMID: 38268620 PMCID: PMC10805998 DOI: 10.1002/ccr3.8449] [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: 11/05/2023] [Revised: 12/23/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024] Open
Abstract
Biochemical evolution of serum IgG-Kappa monoclonal component during the first line with VRD (x1), DARA-VRD (x4), and the second line with ISA-KD (x4).
Collapse
|
9
|
Regidor BS, Jew S, Goldwater MS, Beatty BM, Bujarski S, ElSayed A, Danis R, Kim S, Swift R, Schwartz G, Berenson JR. Efficacy of isatuximab in combination with steroids for the treatment of relapsed/refractory multiple myeloma patients exhibiting only biochemical progression-A single center retrospective study. Eur J Haematol 2023; 111:628-635. [PMID: 37485542 DOI: 10.1111/ejh.14057] [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: 03/07/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/25/2023]
Abstract
OBJECTIVES Isatuximab is approved for treatment of relapsed/refractory multiple myeloma (RRMM) with dexamethasone and carfilzomib or pomalidomide. Patients receiving these three-drug regimens have exhibited more Grade ≥ 3 adverse events (AEs) compared to the two-drug class combination of isatuximab and steroids alone. Thus, this single-center retrospective study investigated the efficacy of isatuximab with dexamethasone and methylprednisolone (ISAdm) for RRMM patients showing only biochemical progression (BP) of their disease. METHODS Twenty-four RRMM patients exhibiting only BP were administered isatuximab at 10 mg/kg with dexamethasone once weekly for cycle 1 of a 28-day cycle, followed by every other week for each cycle thereafter. Starting in cycle 2, oral methylprednisolone was added every other day stopping 48 h before and starting 48 h after each dexamethasone infusion. RESULTS Overall response rate and clinical benefit rate were 63% and 79%, respectively. Progression free survival was 12.9 months. There were only 5 AEs of Grade ≥ 3 which included lymphocytopenia (13%), leukopenia (4%), and neutropenia (4%). No Grade ≥ 3 AE related to respiratory infection, anemia, or thrombocytopenia were reported. CONCLUSION This study shows that the two-drug class combination of ISAdm is an effective and well tolerated treatment option for RRMM patients exhibiting only BP.
Collapse
Affiliation(s)
| | - Scott Jew
- Berenson Cancer Center, West Hollywood, California, USA
- Institute for Myeloma and Bone Cancer Research, West Hollywood, California, USA
| | | | | | - Sean Bujarski
- Berenson Cancer Center, West Hollywood, California, USA
- Institute for Myeloma and Bone Cancer Research, West Hollywood, California, USA
| | - Adam ElSayed
- Berenson Cancer Center, West Hollywood, California, USA
- Institute for Myeloma and Bone Cancer Research, West Hollywood, California, USA
| | - Ryan Danis
- Berenson Cancer Center, West Hollywood, California, USA
| | - Susanna Kim
- Oncotherapeutics, West Hollywood, California, USA
| | - Regina Swift
- Berenson Cancer Center, West Hollywood, California, USA
| | - Gary Schwartz
- Berenson Cancer Center, West Hollywood, California, USA
| | - James R Berenson
- Berenson Cancer Center, West Hollywood, California, USA
- Institute for Myeloma and Bone Cancer Research, West Hollywood, California, USA
- Oncotherapeutics, West Hollywood, California, USA
| |
Collapse
|
10
|
Candelaria PV, Nava M, Daniels-Wells TR, Penichet ML. A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma. Cancers (Basel) 2023; 15:4533. [PMID: 37760502 PMCID: PMC10526502 DOI: 10.3390/cancers15184533] [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: 07/05/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Multiple myeloma (MM) is an incurable malignancy of plasma cells and the second most common hematologic malignancy in the United States. Although antibodies in clinical cancer therapy are generally of the IgG class, antibodies of the IgE class have attractive properties as cancer therapeutics, such as their high affinity for Fc receptors (FcεRs), the low serum levels of endogenous IgE allowing for less competition for FcR occupancy, and the lack of inhibitory FcRs. Importantly, the FcεRs are expressed on immune cells that elicit antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), and/or antigen presentation such as mast cells, eosinophils, macrophages, and dendritic cells. We now report the development of a fully human IgE targeting human CD38 as a potential MM therapy. We targeted CD38 given its high and uniform expression on MM cells. The novel anti-CD38 IgE, expressed in mammalian cells, is properly assembled and secreted, exhibits the correct molecular weight, binds antigen and the high affinity FcεRI, and induces degranulation of FcεRI expressing cells in vitro and also in vivo in transgenic BALB/c mice expressing human FcεRIα. Moreover, the anti-CD38 IgE induces ADCC and ADCP mediated by monocytes/macrophages against human MM cells (MM.1S). Importantly, the anti-CD38 IgE also prolongs survival in a preclinical disseminated xenograft mouse model using SCID-Beige mice and human MM.1S cells when administered with human peripheral blood mononuclear cells (PBMCs) as a source of monocyte effector cells. Our results suggest that anti-CD38 IgE may be effective in humans bearing MM and other malignancies expressing CD38.
Collapse
Affiliation(s)
- Pierre V. Candelaria
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Miguel Nava
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Tracy R. Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Manuel L. Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- The Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
Effer B, Perez I, Ulloa D, Mayer C, Muñoz F, Bustos D, Rojas C, Manterola C, Vergara-Gómez L, Dappolonnio C, Weber H, Leal P. Therapeutic Targets of Monoclonal Antibodies Used in the Treatment of Cancer: Current and Emerging. Biomedicines 2023; 11:2086. [PMID: 37509725 PMCID: PMC10377242 DOI: 10.3390/biomedicines11072086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer is one of the leading global causes of death and disease, and treatment options are constantly evolving. In this sense, the use of monoclonal antibodies (mAbs) in immunotherapy has been considered a fundamental aspect of modern cancer therapy. In order to avoid collateral damage, it is indispensable to identify specific molecular targets or biomarkers of therapy and/or diagnosis (theragnostic) when designing an appropriate immunotherapeutic regimen for any type of cancer. Furthermore, it is important to understand the currently employed mAbs in immunotherapy and their mechanisms of action in combating cancer. To achieve this, a comprehensive understanding of the biology of cancer cell antigens, domains, and functions is necessary, including both those presently utilized and those emerging as potential targets for the design of new mAbs in cancer treatment. This review aims to provide a description of the therapeutic targets utilized in cancer immunotherapy over the past 5 years, as well as emerging targets that hold promise as potential therapeutic options in the application of mAbs for immunotherapy. Additionally, the review explores the mechanisms of actin of the currently employed mAbs in immunotherapy.
Collapse
Affiliation(s)
- Brian Effer
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Isabela Perez
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Daniel Ulloa
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Carolyn Mayer
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Francisca Muñoz
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Diego Bustos
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Claudio Rojas
- Programa de Doctorado en Ciencias Médicas, Universidad de la Frontera, Temuco 4811230, Chile
- Centro de Estudios Morfológicos y Quirúrgicos de La, Universidad de La Frontera, Temuco 4811230, Chile
| | - Carlos Manterola
- Programa de Doctorado en Ciencias Médicas, Universidad de la Frontera, Temuco 4811230, Chile
- Centro de Estudios Morfológicos y Quirúrgicos de La, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis Vergara-Gómez
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Camila Dappolonnio
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Helga Weber
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
| | - Pamela Leal
- Center of Excellence in Translational Medicine (CEMT) and Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
- Department of Agricultural Sciences and Natural Resources, Faculty of Agricultural and Forestry Science, Universidad de La Frontera, Temuco 4810296, Chile
| |
Collapse
|
12
|
Pouleau B, Estoppey C, Suere P, Nallet E, Laurendon A, Monney T, Pais Ferreira D, Drake A, Carretero-Iglesia L, Macoin J, Berret J, Pihlgren M, Doucey MA, Gudi GS, Menon V, Udupa V, Maiti A, Borthakur G, Srivastava A, Blein S, Mbow ML, Matthes T, Kaya Z, Edwards CM, Edwards JR, Menoret E, Kervoëlen C, Pellat-Deceunynck C, Moreau P, Zhukovsky E, Perro M, Chimen M. Preclinical characterization of ISB 1342, a CD38 × CD3 T-cell engager for relapsed/refractory multiple myeloma. Blood 2023; 142:260-273. [PMID: 37192303 PMCID: PMC10644056 DOI: 10.1182/blood.2022019451] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/06/2023] [Accepted: 05/01/2023] [Indexed: 05/18/2023] Open
Abstract
Although treatment of multiple myeloma (MM) with daratumumab significantly extends the patient's lifespan, resistance to therapy is inevitable. ISB 1342 was designed to target MM cells from patients with relapsed/refractory MM (r/r MM) displaying lower sensitivity to daratumumab. ISB 1342 is a bispecific antibody with a high-affinity Fab binding to CD38 on tumor cells on a different epitope than daratumumab and a detuned scFv domain affinity binding to CD3ε on T cells, to mitigate the risk of life-threatening cytokine release syndrome, using the Bispecific Engagement by Antibodies based on the TCR (BEAT) platform. In vitro, ISB 1342 efficiently killed cell lines with different levels of CD38, including those with a lower sensitivity to daratumumab. In a killing assay where multiple modes of action were enabled, ISB 1342 showed higher cytotoxicity toward MM cells compared with daratumumab. This activity was retained when used in sequential or concomitant combinations with daratumumab. The efficacy of ISB 1342 was maintained in daratumumab-treated bone marrow patient samples showing lower sensitivity to daratumumab. ISB 1342 induced complete tumor control in 2 therapeutic mouse models, unlike daratumumab. Finally, in cynomolgus monkeys, ISB 1342 displayed an acceptable toxicology profile. These data suggest that ISB 1342 may be an option in patients with r/r MM refractory to prior anti-CD38 bivalent monoclonal antibody therapies. It is currently being developed in a phase 1 clinical study.
Collapse
Affiliation(s)
- Blandine Pouleau
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | - Carole Estoppey
- Department of Antibody Engineering, Ichnos Sciences SA, Epalinges, Switzerland
| | - Perrine Suere
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | - Emilie Nallet
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | - Amélie Laurendon
- Department of Antibody Engineering, Ichnos Sciences SA, Epalinges, Switzerland
| | - Thierry Monney
- Department of Antibody Engineering, Ichnos Sciences SA, Epalinges, Switzerland
| | | | - Adam Drake
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | | | - Julie Macoin
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | - Jérémy Berret
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | - Maria Pihlgren
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | | | - Girish S. Gudi
- Department of Pharmacokinetics and Translational Sciences, Ichnos Sciences Inc, New York, NY
| | - Vinu Menon
- Department of Pharmacokinetics and Translational Sciences, Ichnos Sciences Inc, New York, NY
| | - Venkatesha Udupa
- Department of Toxicology, Glenmark Pharmaceuticals Limited, Mumbai, India
| | - Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ankita Srivastava
- Department of Antibody Engineering, Ichnos Sciences SA, Epalinges, Switzerland
| | - Stanislas Blein
- Department of Antibody Engineering, Ichnos Sciences SA, Epalinges, Switzerland
| | - M. Lamine Mbow
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | - Thomas Matthes
- Hematology Service, Department of Oncology and Clinical Pathology Service, Department of Diagnostics, University Hospital Geneva, Geneva, Switzerland
| | - Zeynep Kaya
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, United Kingdom
| | - Claire M. Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, United Kingdom
| | - James R. Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, United Kingdom
| | - Emmanuelle Menoret
- Nantes Université, INSERM, Centre national de la recherche scientifique, Université d'Angers, Nantes, France
- Therassay Core Facility, Department of Onco-Hematology, Capacités, Nantes Université, Nantes, France
| | - Charlotte Kervoëlen
- Nantes Université, INSERM, Centre national de la recherche scientifique, Université d'Angers, Nantes, France
- Therassay Core Facility, Department of Onco-Hematology, Capacités, Nantes Université, Nantes, France
| | - Catherine Pellat-Deceunynck
- Nantes Université, INSERM, Centre national de la recherche scientifique, Université d'Angers, Nantes, France
- SIRIC ILIAD, Angers, Nantes, France
| | - Philippe Moreau
- Nantes Université, INSERM, Centre national de la recherche scientifique, Université d'Angers, Nantes, France
- SIRIC ILIAD, Angers, Nantes, France
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU Nantes, Nantes, France
| | - Eugene Zhukovsky
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | - Mario Perro
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| | - Myriam Chimen
- Department of Oncology, Ichnos Sciences SA, Epalinges, Switzerland
| |
Collapse
|
13
|
Vassilopoulos S, Vassilopoulos A, Kalligeros M, Shehadeh F, Mylonakis E. Cumulative Incidence and Relative Risk of Infection in Patients With Multiple Myeloma Treated With Anti-CD38 Monoclonal Antibody-Based Regimens: A Systematic Review and Meta-analysis. Open Forum Infect Dis 2022; 9:ofac574. [PMID: 36438616 PMCID: PMC9685179 DOI: 10.1093/ofid/ofac574] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Patients with multiple myeloma are at higher risk for infections due to disease pathogenesis and administered therapies. The purpose of this study was to estimate the risk for any grade and severe infections associated with the use of anti-CD38 monoclonal antibodies in patients with multiple myeloma. METHODS We searched PubMed and EMBASE for randomized controlled trials (RCTs) that included patients with multiple myeloma who received CD38-targeting monoclonal antibody regimens and reported outcomes of infection and performed a random-effects meta-analysis to estimate the relative risk for infections. RESULTS After screening 673 citations, we retrieved 17 studies providing data on 11 RCTs. Overall, the included reports evaluated 5316 patients (2797 in the intervention arm and 2519 in the control arm). The relative risk (RR) for both any grade or severe infections was 1.27 (95% CI, 1.17-1.37 and 1.14-1.41, respectively). The cumulative incidence of any grade infections for patients who received anti-CD38 agents was 77% (95% CI, 68%-86%), while for severe infections it was 28% (95% CI, 23%-34%). Patients treated with anti-CD38 agents had a 39% higher risk for any grade pneumonia (RR, 1.39; 95% CI, 1.12-1.72) and a 38% higher risk for severe pneumonia (RR, 1.38; 95% CI, 1.09-1.75). For upper respiratory tract infections, the relative risk was 1.51 and 1.71 for any grade and severe infections, respectively. Regarding varicella-zoster virus (VZV) reactivation, we found no evidence of increased risk (RR, 3.86; 95% CI, 0.66-22.50). CONCLUSIONS Patients with multiple myeloma treated with regimens that included an anti-CD38 monoclonal antibody were at higher risk for any grade or severe infections without an associated higher mortality rate during the follow-up period of the retrieved studies. No evidence of increased risk for VZV reactivation was noted, but there was a significant association between CD38-targeting treatment and pneumonia risk. Increased surveillance for infections, development of effective prophylactic strategies, and studies with long follow-up are needed for patients with multiple myeloma treated with anti-CD38-based regimens.
Collapse
Affiliation(s)
- Stephanos Vassilopoulos
- Infectious Diseases Division, Rhode Island Hospital, Providence, Rhode Island, USA
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Athanasios Vassilopoulos
- Infectious Diseases Division, Rhode Island Hospital, Providence, Rhode Island, USA
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Markos Kalligeros
- Infectious Diseases Division, Rhode Island Hospital, Providence, Rhode Island, USA
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Fadi Shehadeh
- Infectious Diseases Division, Rhode Island Hospital, Providence, Rhode Island, USA
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
- School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Rhode Island Hospital, Providence, Rhode Island, USA
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|