1
|
Loyau J, Monney T, Montefiori M, Bokhovchuk F, Streuli J, Blackburn M, Goepfert A, Caro LN, Chakraborti S, De Angelis S, Grandclément C, Blein S, Mbow ML, Srivastava A, Perro M, Sammicheli S, Zhukovsky EA, Dyson M, Dreyfus C. Biparatopic binding of ISB 1442 to CD38 in trans enables increased cell antibody density and increased avidity. MAbs 2025; 17:2457471. [PMID: 39882744 PMCID: PMC11784651 DOI: 10.1080/19420862.2025.2457471] [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: 10/17/2024] [Revised: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 01/31/2025] Open
Abstract
ISB 1442 is a bispecific biparatopic antibody in clinical development to treat hematological malignancies. It consists of two adjacent anti-CD38 arms targeting non-overlapping epitopes that preferentially drive binding to tumor cells and a low-affinity anti-CD47 arm to enable avidity-induced blocking of proximal CD47 receptors. We previously reported the pharmacology of ISB 1442, designed to reestablish synthetic immunity in CD38+ hematological malignancies. Here, we describe the discovery, optimization and characterization of the ISB 1442 antigen binding fragment (Fab) arms, their assembly to 2 + 1 format, and present the high-resolution co-crystal structures of the two anti-CD38 Fabs, in complex with CD38. This, with biophysical and functional assays, elucidated the underlying mechanism of action of ISB 1442. In solution phase, ISB 1442 forms a 2:2 complex with CD38 as determined by size-exclusion chromatography with multi-angle light scattering and electron microscopy. The predicted antibody-antigen stoichiometries at different CD38 surface densities were experimentally validated by surface plasmon resonance and cell binding assays. The specific design and structural features of ISB 1442 enable: 1) enhanced trans binding to adjacent CD38 molecules to increase Fc density at the cancer cell surface; 2) prevention of avid cis binding to monomeric CD38 to minimize blockade by soluble shed CD38; and 3) greater binding avidity, with a slower off-rate at high CD38 density, for increased specificity. The superior CD38 targeting of ISB 1442, at both high and low receptor densities, by its biparatopic design, will enhance proximal CD47 blockade and thus counteract a major tumor escape mechanism in multiple myeloma patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mario Perro
- Ichnos Glenmark Innovation, New York, NY, USA
| | | | | | | | | |
Collapse
|
2
|
Liu X, Feng Y, Song Z, Liu J, Luo Z, Yu G, Wang J. Novel and effective tandem CD38 and CD19 targeting CAR-T cells inhibit hematological tumor immune escape. Cell Immunol 2025; 411-412:104950. [PMID: 40239552 DOI: 10.1016/j.cellimm.2025.104950] [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: 01/09/2025] [Revised: 03/28/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025]
Abstract
Targeting CD19 with chimeric antigen receptor (CAR)-T cells is clinically effective, but tumor immune escape and tumor recurrence still occur. Designing CAR-T cells that target multiple antigens simultaneously is a viable approach for inhibiting tumor immune escape, and promising findings have been reported. In this study, we designed new CD19 and CD38 dual-target CAR-T cells that are strongly cytotoxic to target cells expressing CD19 or CD38. In vitro studies, compared with single-target CAR-T cells or CD19/CD38 tandem (Tan) CAR-T cells, CD38/CD19 Tan CAR-T cells presented similar CAR expression, superior cytotoxicity and antigen-stimulated T-cell proliferation. In vivo studies, CD38/CD19 Tan CAR-T cells demonstrated the same efficacy and safety as single-target CAR-T. These CD19/CD38 Tan CAR-T cells are fully compatible with existing clinical-grade T-cell manufacturing procedures and can be implemented using current clinical protocols. In summary, our findings provide an effective solution to the challenge of tumor immune escape in anti-CD19 CAR-T-cell therapy.
Collapse
Affiliation(s)
- Xiuying Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yaru Feng
- Junjo Biopharmaceutical Co., Ltd., Zhongshan 528437, China
| | - Zhiru Song
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jingjing Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhiqiang Luo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guohua Yu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jianxun Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China; Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen 518118, China.
| |
Collapse
|
3
|
Lim KJC, Wellard C, Moore E, Ninkovic S, Chng WJ, Spencer A, Mollee P, Hocking J, Ho PJ, Janowski W, Kim K, McCaughan G, Dun K, McQuilten ZK, Chen F, Quach H. Presence of 1q21 Gain and Amplification May be Associated With Poorer Outcomes in Daratumumab-treated Multiple Myeloma Patients. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:243-248. [PMID: 39616008 DOI: 10.1016/j.clml.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 03/18/2025]
Affiliation(s)
- Kenneth J C Lim
- Department of Haematology, St Vincent's Hospital Melbourne, Melbourne, Australia; Victorian Cancer Cytogenetics Service, St Vincent's Hospital Melbourne, Melbourne Australia.
| | - Cameron Wellard
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Elizabeth Moore
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Slavisa Ninkovic
- Department of Haematology, St Vincent's Hospital Melbourne, Melbourne, Australia; Victorian Cancer Cytogenetics Service, St Vincent's Hospital Melbourne, Melbourne Australia; Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Wee Joo Chng
- National University Cancer Institute, National University Health System, Singapore; Cancer Science Institute of Singapore, and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Andrew Spencer
- Department of Haematology, The Alfred Hospital, Melbourne Australia; Department of Haematology, Monash Health, Melbourne Australia
| | - Peter Mollee
- Department of Haematology, Princess Alexandra Hospital, Brisbane Australia
| | - Jay Hocking
- Department of Haematology, Austin Health, Melbourne Australia
| | - Phoebe Joy Ho
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, Australia; University of Sydney, Sydney, Australia
| | - Wojt Janowski
- Calvary Mater Newcastle Hospital, Newcastle, Australia
| | - Kihyun Kim
- Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Georgia McCaughan
- Department of Haematology, St Vincent's Hospital Sydney, Sydney, Australia; Garvan Institute of Medical Research, Sydney, Australia; Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Karen Dun
- Victorian Cancer Cytogenetics Service, St Vincent's Hospital Melbourne, Melbourne Australia
| | - Zoe K McQuilten
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia; Department of Haematology, Monash Health, Melbourne Australia
| | - Fiona Chen
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Hang Quach
- Department of Haematology, St Vincent's Hospital Melbourne, Melbourne, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia
| |
Collapse
|
4
|
Lin L, Roccuzzo G, Yakymiv Y, Marchisio S, Ortolan E, Funaro A, Senetta R, Pala V, Bagot M, de Masson A, Battistella M, Guenova E, Ribero S, Quaglino P. The CD39/CD73/Adenosine and NAD/CD38/CD203a/CD73 Axis in Cutaneous T-Cell Lymphomas. Cells 2025; 14:309. [PMID: 39996780 PMCID: PMC11854806 DOI: 10.3390/cells14040309] [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: 01/05/2025] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
Cutaneous T-cell lymphoma (CTCL), characterized by malignant T-cell proliferation primarily in the skin, includes subtypes such as mycosis fungoides (MF) and Sézary syndrome (SS). The tumor microenvironment (TME) is central to their pathogenesis, with flow cytometry and histology being the gold standards for detecting malignant T cells within the TME. Alongside emerging molecular markers, particularly clonality analysis, these tools are indispensable for accurate diagnosis and treatment planning. Of note, adenosine signaling within the TME has been shown to suppress immune responses, affecting various cell types. The expression of CD39, CD73, and CD38, enzymes involved in adenosine production, can be elevated in MF and SS, contributing to immune suppression. Conversely, the expression of CD26, part of the adenosine deaminase/CD26 complex, that degrades adenosine, is often lost by circulating tumoral cells. Flow cytometry has demonstrated increased levels of CD39 and CD73 on Sézary cells, correlating with disease progression and prognosis, while CD38 shows a variable expression, with its prognostic significance remaining under investigation. Understanding these markers' roles in the complexity of TME-mediated immune evasion mechanisms might enhance diagnostic precision and offer new therapeutic targets in CTCL.
Collapse
Affiliation(s)
- Liyun Lin
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Gabriele Roccuzzo
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (V.P.); (P.Q.)
| | - Yuliya Yakymiv
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Sara Marchisio
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Erika Ortolan
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Ada Funaro
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Rebecca Senetta
- Pathology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy;
| | - Valentina Pala
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (V.P.); (P.Q.)
| | - Martine Bagot
- Dermatology Department, Saint-Louis Hospital, AP-HP, Université Paris Cité, Inserm U976, 75010 Paris, France; (M.B.); (A.d.M.)
| | - Adèle de Masson
- Dermatology Department, Saint-Louis Hospital, AP-HP, Université Paris Cité, Inserm U976, 75010 Paris, France; (M.B.); (A.d.M.)
| | - Maxime Battistella
- Department of Pathology, Hôpital Saint-Louis, AP-HP, Université Paris Cité, Inserm U976, 75010 Paris, France;
| | - Emmanuella Guenova
- Department of Dermatology, Lausanne University Hospital (CHUV) and Faculty of Biology and Medicine, University of Lausanne, 1007 Lausanne, Switzerland;
- University Institute and Clinic for Immunodermatology, Medical Faculty, Johannes Kepler University, 4020 Linz, Austria
| | - Simone Ribero
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (V.P.); (P.Q.)
| | - Pietro Quaglino
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (V.P.); (P.Q.)
| |
Collapse
|
5
|
Wan X, Yu T, Yu T, Cai H. The impact of multiple myeloma drugs treatments on autologous stem cell transplantation in the era of new drugs. Front Oncol 2025; 15:1479164. [PMID: 40027122 PMCID: PMC11868118 DOI: 10.3389/fonc.2025.1479164] [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: 08/11/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Autologous stem cell transplantation (ASCT) is the standard treatment recommended by the National Comprehensive Cancer Network (NCCN) for newly diagnosed multiple myeloma (MM) patients who are eligible for transplantation. This procedure follows response achieved through induction therapy. The key to the success of ASCT lies in the quantity and quality of hematopoietic stem cells collected after mobilization. Studies have shown a positive correlation between the number of hematopoietic stem cells collected and the engraftment time of absolute neutrophil count (ANC) and platelet count (PLT). However, the advent of novel therapeutic agents that have significantly improved the survival of MM patients has also impacted hematopoietic stem cell mobilization, potentially delaying hematopoietic recovery, a process referred to as hematopoietic remodeling. In this paper, we will retrospectively analyze and summarise the research progress related to the effects of previous chemotherapeutic agents on hematopoietic stem cell mobilization and hematopoietic remodeling, to further improve the prognosis and quality of survival of MM patients who are eligible for transplantation.
Collapse
Affiliation(s)
- Xixi Wan
- Department of Hematology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, China
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Tian Yu
- Department of Hematology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, China
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Tao Yu
- Department of Hematology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, China
| | - Huili Cai
- Department of Hematology, the First Clinical Medical College of China Three Gorges University, Yichang, China
| |
Collapse
|
6
|
Caroni F, Sammartano V, Pacelli P, Sicuranza A, Malchiodi M, Dragomir A, Ciofini S, Raspadori D, Bocchia M, Gozzetti A. Minimal Residual Disease Significance in Multiple Myeloma Patients Treated with Anti-CD38 Monoclonal Antibodies. Pharmaceuticals (Basel) 2025; 18:159. [PMID: 40005973 PMCID: PMC11858645 DOI: 10.3390/ph18020159] [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: 12/14/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Minimal residual disease (MRD) evaluation is a recognized endpoint in clinical trials. Both next-generation flow and sequencing could be used as complementary techniques to detect myeloma cells after therapy to measure the depth of response and novel drug efficacy. Anti-CD38 monoclonal antibodies combined with proteasome inhibitors and immunomodulatory drugs have increased the quality of response in myeloma patients, and MRD evaluation is also entering routine clinical practice in many hematological centers. This review analyzes updated results from recent clinical trials utilizing anti-CD38 monoclonal antibodies such as isatuximab and daratumumab in terms of their responses and MRD data. MRD-driven therapy appears promising for the future of MM patients, and emerging minimally invasive techniques to assess MRD are under investigation as novel potential methods to replace or integrate traditional MRD evaluation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Alessandro Gozzetti
- AOUS Policlinico Le Scotte, University of Siena, 53100 Siena, Italy; (F.C.); (V.S.); (P.P.); (A.S.); (M.M.); (A.D.); (S.C.); (D.R.); (M.B.)
| |
Collapse
|
7
|
Galusic D, Krecak I, Blaslov V, Krstulovic Opara A, Valkovic T, Basic Kinda S. Is There a Role for Daratumumab Retreatment in Patients with Relapsed/Refractory Multiple Myeloma? Biomedicines 2025; 13:207. [PMID: 39857790 PMCID: PMC11762825 DOI: 10.3390/biomedicines13010207] [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: 12/23/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Multiple myeloma (MM) is a hematologic disease characterized by the clonal expansion of malignant plasma cells that accumulate in the bone marrow, leading to osteolytic bone disease, hypercalcemia, anemia, and renal dysfunction. Daratumumab was the first monoclonal anti-CD38 antibody approved for the treatment of MM, initially in relapse/refractory settings and, more recently, for newly diagnosed patients. Increased first-line usage of daratumumab will also substantially change treatment approaches for patients with relapsed/refractory disease. Due to the cost and availability of bispecific T cell redirecting antibodies (BsAbs) and chimeric antigen receptor T cell therapy (CAR-T) in real-life settings in many countries, retreatment with daratumumab in subsequent lines of therapy might be a reasonable choice. Data regarding efficacy and optimal combinations of daratumumab retreatment are lacking, and here we provide a short literature review of available data. We identified only a small number of articles based on retrospective analysis of medical records in real-life settings. A strong consistency in results regarding response rates and treatment duration was noticed among mainly heavily pre-treated MM patients, with approximately half of patients achieving at least partial remission (PR) after retreatment with daratumumab-based protocol. The duration of treatment and time to the next treatment for retreatment episodes were considerable and consistent with clinical expectations for later lines of therapy. The analysis of data in this literature review indicates that daratumumab retreatment may provide meaningful clinical benefit to some patients with relapsed/refractory MM despite having prior exposure. However, further research is needed to identify clinical and biological parameters that may predict favorable responses to daratumumab retreatment.
Collapse
Affiliation(s)
- Davor Galusic
- Division of Hematology, University Hospital of Split, 21000 Split, Croatia; (D.G.); (V.B.); (A.K.O.)
- School of Medicine, University of Split, 21000 Split, Croatia
| | - Ivan Krecak
- Department of Internal Medicine, General Hospital of Sibenik, 22000 Sibenik, Croatia
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
- University of Applied Sciences Sibenik, 22000 Sibenik, Croatia
| | - Viktor Blaslov
- Division of Hematology, University Hospital of Split, 21000 Split, Croatia; (D.G.); (V.B.); (A.K.O.)
| | - Andela Krstulovic Opara
- Division of Hematology, University Hospital of Split, 21000 Split, Croatia; (D.G.); (V.B.); (A.K.O.)
| | - Toni Valkovic
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
- Department of Internal Medicine, Pula General Hospital, 52100 Pula, Croatia
| | - Sandra Basic Kinda
- Division of Hematology, University Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia
| |
Collapse
|
8
|
Zaiema SEGE, Hafez HMS, El-Ela DEDMSA, Saad RAAAM. Illuminating the impact of CD38-induced adenosine formation in B-cell lymphoma. Sci Rep 2025; 15:1807. [PMID: 39805878 PMCID: PMC11731001 DOI: 10.1038/s41598-024-82800-1] [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: 07/18/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
The expression of CD38 by cancer cells may mediate an immune-suppressive effect by producing Extracellular Adenosine (ADO) acting through G-protein-coupled cell surface receptors on cellular components and tumor cells. This can increase PD-1 expression and interaction with PD-L1, suppressing CD8 + cytotoxic T cells. This study examines the impact of heightened CD38 expression and extracellular ADO on various hematological and clinical parameters in patients with mature B-cell lymphoma, alongside their correlation with the soluble counterparts of the PD-1/PD-L1 axis. Our study was conducted on 90 patients, CD38-positive and CD38-negative (measured by flow cytometry), with mature B-cell lymphoma divided into CLL and B-NHL subtypes. Their serum ADO, soluble PD-1, and PD-L1 levels were measured using a sandwich ELISA. Our study revealed a positive correlation between CD38 expression, sADO, sPD-1, and sPD-L1 in mature B-cell lymphoma patients. CD38-positive patients had higher sADO, sPD-1, and sPD-L1 levels. Higher CD38 expression and extracellular ADO negatively affected HB level and PLT count and positively correlated with the higher risk stratification in mature B-cell lymphoma patients. This study explored the potential impact of CD38 expression and elevated extracellular ADO on B-cell lymphoma alongside their link with the PD-1/PD-L1 axis. Our findings underscore the influence of extracellular ADO on the neoplastic process of mature B-cell lymphoma. We also propose targeting the CD38-induced-ADO formation pathway, which could serve as a promising therapeutic immune target with multifaceted effects within mature B-cell neoplasms.
Collapse
|
9
|
Yiğit Kaya S, Vatani M, Akil R, Cakir T, Maral S, Kaynar L, Sevindik ÖG. A complete response with daratumumab, venetoclax, azacitidine and dexamethasone in a heavily pre-treated, chemo-refractory early T-precursor acute lymphoblastic leukemia/lymphoma patient. Ann Hematol 2025; 104:829-833. [PMID: 39847114 DOI: 10.1007/s00277-024-06118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/21/2024] [Indexed: 01/24/2025]
Abstract
Early T-precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) is a rare and aggressive subtype of T-cell leukemia with poor prognosis and resistance to standard treatments. We report a 21-year-old male with ETP-ALL/LBL who, after an initial complete remission with the HOELZER protocol, experienced early relapse and was refractory to subsequent FLEND and BFM protocols. Following disease progression and complications, he was treated with a combination of daratumumab, venetoclax, azacitidine, and dexamethasone. This regimen achieved complete remission after one cycle. This case highlights the potential of this combination therapy as an effective treatment for refractory ETP-ALL/LBL, suggesting further research is warranted to validate its efficacy and safety.
Collapse
Affiliation(s)
- Süreyya Yiğit Kaya
- Department of Hematology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Mehrad Vatani
- Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Rima Akil
- Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Tansel Cakir
- Department of Nuclear Medicine, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Senem Maral
- Department of Hematology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Leylagül Kaynar
- Department of Hematology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ömür Gökmen Sevindik
- Department of Hematology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
10
|
Song S, Gan J, Long Q, Gao Z, Zheng Y. Decoding NAD+ Metabolism in COVID-19: Implications for Immune Modulation and Therapy. Vaccines (Basel) 2024; 13:1. [PMID: 39852780 PMCID: PMC11768799 DOI: 10.3390/vaccines13010001] [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: 11/22/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/26/2025] Open
Abstract
The persistent threat of COVID-19, particularly with the emergence of new variants, underscores the urgency for innovative therapeutic strategies beyond conventional antiviral treatments. Current immunotherapies, including IL-6/IL-6R monoclonal antibodies and JAK inhibitors, exhibit suboptimal efficacy, necessitating alternative approaches. Our review delves into the significance of NAD+ metabolism in COVID-19 pathology, marked by decreased NAD+ levels and upregulated NAD+-consuming enzymes such as CD38 and poly (ADP-ribose) polymerases (PARPs). Recognizing NAD+'s pivotal role in energy metabolism and immune modulation, we propose modulating NAD+ homeostasis could bolster the host's defensive capabilities against the virus. The article reviews the scientific rationale behind targeting NAD+ pathways for therapeutic benefit, utilizing strategies such as NAD+ precursor supplementation and enzyme inhibition to modulate immune function. While preliminary data are encouraging, the challenge lies in optimizing these interventions for clinical use. Future research should aim to unravel the intricate roles of key metabolites and enzymes in NAD+ metabolism and to elucidate their specific mechanisms of action. This will be essential for developing targeted NAD+ therapies, potentially transforming the management of COVID-19 and setting a precedent for addressing other infectious diseases.
Collapse
Affiliation(s)
- Shixu Song
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
| | - Jialing Gan
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
| | - Qiuyue Long
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
| | - Zhancheng Gao
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
- Department of Respiratory and Critical Care Medicine, Peking University People’s Hospital, Beijing 100044, China
| | - Yali Zheng
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
| |
Collapse
|
11
|
Lv R, Wang D, Wang T, Li R, Zhuang A. Causality between gut microbiota, immune cells, and breast cancer: Mendelian randomization analysis. Medicine (Baltimore) 2024; 103:e40815. [PMID: 39654239 PMCID: PMC11630993 DOI: 10.1097/md.0000000000040815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024] Open
Abstract
The association between gut microbiota (GM) and breast cancer (BC) has been studied. Nevertheless, the causal relationship between them and the potential mediating factors have not been clearly defined. Therefore, in this study, Mendelian randomization analysis (MR) was employed to explore the causal relationship between 473 GM and BC, as well as the mediating effect of potential immune cells. In this investigation, we availed ourselves of the publicly accessible summary statistics from the genome-wide association study to undertake two-sample and reverse Mendelian randomization analyses on GM and BC, with the intention of clarifying the causal association between GM and BC. Subsequently, through the application of the two-step Mendelian randomization analysis, it was revealed that the relationship between GM and BC was mediated by immune cells. The stability of the research outcomes was verified via sensitivity analysis. Mendelian randomization analysis elucidated the protective impacts of 8 genera on BC (such as Phylum Actinobacteriota, Species Bacteroides A plebeius A, Species Bifidobacterium adolescentis, Species CAG-841 sp002479075, Family Fibrobacteraceae, Order Fibrobacterales, Class Fibrobacteria, and Species Phascolarctobacterium sp003150755). Additionally, there are 23 immune cell traits related to BC. Our research findings showed that the species Megamonas funiformis was associated with an increased risk of BC, and 11.20% of this effect was mediated by CD38 on IgD+ CD24-. Likewise, HLA DR on CD33br HLA DR+ CD14- mediated the causal relationship between Species Prevotellamassilia and BC, having a mediating ratio of 7.89%. This study clarifies a potential causal relationship between GM, immune cells, and BC and provides genetic evidence for this causal connection. It offers research directions for the subsequent prevention and treatment of BC through the interaction between GM and immune cells, and provides a reference for future mechanistic and clinical studies in this field.
Collapse
Affiliation(s)
- Rui Lv
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Danyan Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhuji Second People’s Hospital, Zhuji, China
| | - Tengyue Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rongqun Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Aiwen Zhuang
- Institute of TCM Literature and Information, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
12
|
Pitoy A, Desmée S, Riglet F, Thai HT, Klippel Z, Semiond D, Veyrat-Follet C, Bertrand J. Isatuximab-dexamethasone-pomalidomide combination effects on serum M protein and PFS in myeloma: Development of a joint model using phase I/II data. CPT Pharmacometrics Syst Pharmacol 2024; 13:2087-2101. [PMID: 39607833 DOI: 10.1002/psp4.13206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/26/2024] [Accepted: 07/07/2024] [Indexed: 11/30/2024] Open
Abstract
This study aimed at leveraging data from phase I/II clinical trials to build a nonlinear joint model of serum M-protein kinetics and progression-free survival (PFS) accounting for the effects of isatuximab (Isa), pomalidomide (Pom), and dexamethasone (Dex) in patients with relapsed and/or refractory multiple myeloma. Serum M-protein levels and PFS data from 203 evaluable patients, included either in a phase I/II study (n = 173) or in a phase I study (n = 30), were used to build the model. First, we independently developed a longitudinal model and a PFS model. Then, we linked them in a nonlinear joint model by selecting the link function that best captured the association between serum M-protein kinetics and PFS. A Claret tumor growth-inhibition model accounting for the additive effects of Isa, with an Emax function, Pom, and Dex on serum M-protein elimination was selected to describe serum M-protein kinetics. PFS was best described with a log-logistic model and associations with baseline beta-2 microglobulin level, age, and coadministration of Dex were identified. The instantaneous change in serum M-protein level was found to be associated with PFS in the final joint model. Using model simulations, we retrospectively supported the Isa 10 mg/kg weekly for 4 weeks, then biweekly (QW/Q2W) dosing regimen of the ICARIA-MM phase III pivotal study, and validated it using the same phase III pivotal study data.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Female
- Humans
- Male
- Middle Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Dexamethasone/administration & dosage
- Dexamethasone/therapeutic use
- Dexamethasone/pharmacology
- Models, Biological
- Multiple Myeloma/drug therapy
- Multiple Myeloma/blood
- Myeloma Proteins/analysis
- Progression-Free Survival
- Thalidomide/analogs & derivatives
- Thalidomide/administration & dosage
- Thalidomide/therapeutic use
Collapse
Affiliation(s)
- Antoine Pitoy
- Sanofi Data and Data Sciences, Translational Disease Modeling, Gentilly, France
- INSERM, SPHERE, U1246, Tours University, Nantes University, Tours, France
- INSERM, IAME, Université Paris Cité, Paris, France
| | - Solène Desmée
- INSERM, SPHERE, U1246, Tours University, Nantes University, Tours, France
| | - François Riglet
- Sanofi Data and Data Sciences, Translational Disease Modeling, Gentilly, France
- INSERM, IAME, Université Paris Cité, Paris, France
- Clinical Pharmacometrics, Quantitative Pharmacology, Servier, Saclay, France
| | - Hoai-Thu Thai
- Sanofi Data and Data Sciences, Translational Disease Modeling, Gentilly, France
| | - Zandra Klippel
- Sanofi Translational Medicine and Early Development, Cambridge, Massachusetts, USA
| | - Dorothée Semiond
- Sanofi Translational Medicine and Early Development, Cambridge, Massachusetts, USA
| | | | | |
Collapse
|
13
|
Huang P, Zhang C, Zhang A, Mao J, Liu G, Hu C, Zhu H. CD56 briCD38 + as a novel neutrophil-specific marker in chronic myeloid leukemia. Heliyon 2024; 10:e39465. [PMID: 39559206 PMCID: PMC11570290 DOI: 10.1016/j.heliyon.2024.e39465] [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: 06/02/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
Background Chronic myeloid leukemia (CML) is classified as a subtype of myeloproliferative neoplasm, and bone marrow flow cytometry does not reveal any specific immunophenotype. Interestingly, aberrant expression of cluster of differentiation (CD) markers such as CD56 and CD38 has been observed on neutrophils in CML patients. Therefore, we investigated the abnormal expression of CD56 and CD38 in CML neutrophils to explore their diagnostic value in identifying CML through flow cytometry. Methods We have developed a multi-parameter flow cytometry assay to identify aberrant immunophenotypes in CML neutrophils among bone marrow nucleated cells. Results Compared to healthy donors and patients with a reactive neutrophilia or other hematological malignancies, the percentage of CD56briCD38+ neutrophil subsets in CML patients exhibits a distinctive increase (cut-off value, 2.0 %). The specificity and sensitivity associated with the learning cohort (168 samples) were 90.8 % and 84.9 %, respectively, while in the validation cohort (194 samples), they were 90.7 % and 84.7 %. The accumulation of CD56briCD38+ neutrophil subsets, which demonstrate are abnormal characteristics, is independent of the neutrophil count, BCR/ABL1 fusions and risk stratification but associated with blast cells immunophenotype. Moreover, this increase disappears in CML patients after treatment with tyrosine kinase inhibitors when the curative effect was satisfactory. Conclusions We conclude that an increase in the proportion of CD56briCD38+ neutrophil subsets exceeding 2.0 % of total neutrophils serves as a highly sensitive and specific flow cytometry marker, enabling rapid and accurate identification of CML.
Collapse
Affiliation(s)
- Panpan Huang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Cuiping Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Aimei Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Ju Mao
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Gan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Chaojie Hu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| | - Huaiping Zhu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, PR China
| |
Collapse
|
14
|
Pilon C, Joher N, Usureau C, Boutin E, Boueilh A, Taupin JL, Thiolat A, Cohen JL, Kheav VD, Canoui-Poitrine F, Carmagnat M, Grimbert P, Matignon M. Open-Label Phase 1/2 Study of Daratumumab-Based Desensitization Before Kidney Transplantation. Kidney Int Rep 2024; 9:3250-3264. [PMID: 39534185 PMCID: PMC11551132 DOI: 10.1016/j.ekir.2024.08.020] [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: 05/30/2024] [Revised: 07/15/2024] [Accepted: 08/20/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction The safety and benefit of the anti-CD38 monoclonal antibody daratumumab, which induces lysis of antibody-producing plasma cells in sensitized patients prior to kidney transplantation, remain to be determined. Methods A 2-phase (1 and 2), monocentric open-label study was conducted to evaluate the month 6 (M6) safety and efficacy of daratumumab in kidney transplant candidates with calculated panel reactive antibody (cPRA) > 95%. In the first (safety) phase, we used 4-weekly escalating doses of daratumumab. Phase 2 tested desensitization with 8 weekly infusions of 16 mg/kg daratumumab. cPRA 10,000 was calculated considering only human leukocyte antigen (HLA) antibodies with mean fluorescence intensity (MFI) of > 10,000. Results Nine patients were enrolled in phase 1 and 14 in phase 2. Safety analysis showed 4 serious non-treatment-emergent adverse events (non-TEAEs), 36 mild TEAEs, mostly infusion-related reactions, grade 1 and 2 (causing 2 temporary drug discontinuations), but no serious TEAEs. Significant reductions in anti-HLA antibodies were observed at month 3 (M3), with cPRA 10,000 (P = 0.003), number of anti-HLA (P < 0.001), maximum MFI (MFI max) (P = 0.053), and the sum of MFI (MFI sum) (P < 0.001), with complete return to baseline levels at month 12 (M12). At M6, 46.15% (19.22%-74.87%) and 76.92% (46.19%-94.96%) of patients showed sustained response (1% decrease in cPRA) for cPRA 2000 and 10,000, respectively. At month 1 (M1), immune cells (T-reg, CD8 + TEMRA, CD19 + CD138 + B cells, and NK cells) significantly decreased. At M3, other antibodies decreased significantly, but returned to baseline levels at M12, except for gamma globulins, without any infectious complications. Conclusion The first use of daratumumab in desensitization demonstrated infusion-related adverse (AEs) events and rapid, albeit transient, reductions in anti-HLA antibodies, with less than 40% of durable responders, limiting its potential clinical use.
Collapse
Affiliation(s)
- Caroline Pilon
- Universite Paris Est Creteil, INSERM IMRB U955, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Groupe hospitalo-universitaire Chenevier Mondor, Centre d’Investigation Clinique Biotherapy, Fédération hospitalo-Universitaire, Innovative therapy for immune disorders, Créteil, France
| | - Nizar Joher
- Department of Nephrology and Renal Transplantation, Assistance Publique-Hôpitaux de Paris, Groupe hospitalo-universitaire Chenevier Mondor, Fédération Hospitalo-Universitaire, Innovative therapy for immune disorders, Créteil, France
| | - Cédric Usureau
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
- INSERM UMR976, Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Emmanuelle Boutin
- Universite Paris Est Creteil, INSERM IMRB U955, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor-Albert Chenevier, Public Health Department and URC, Créteil, France
| | - Anna Boueilh
- Department of Nephrology and Renal Transplantation, Assistance Publique-Hôpitaux de Paris, Groupe hospitalo-universitaire Chenevier Mondor, Fédération Hospitalo-Universitaire, Innovative therapy for immune disorders, Créteil, France
| | - Jean-Luc Taupin
- INSERM UMR976, Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor-Albert Chenevier, Public Health Department and URC, Créteil, France
| | - Allan Thiolat
- Universite Paris Est Creteil, INSERM IMRB U955, Créteil, France
| | - José L. Cohen
- Universite Paris Est Creteil, INSERM IMRB U955, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Groupe hospitalo-universitaire Chenevier Mondor, Centre d’Investigation Clinique Biotherapy, Fédération hospitalo-Universitaire, Innovative therapy for immune disorders, Créteil, France
| | - Vissal David Kheav
- INSERM UMR976, Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Florence Canoui-Poitrine
- Universite Paris Est Creteil, INSERM IMRB U955, Créteil, France
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
| | - Maryvonnick Carmagnat
- INSERM UMR976, Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Philippe Grimbert
- Universite Paris Est Creteil, INSERM IMRB U955, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Groupe hospitalo-universitaire Chenevier Mondor, Centre d’Investigation Clinique Biotherapy, Fédération hospitalo-Universitaire, Innovative therapy for immune disorders, Créteil, France
- Department of Nephrology and Renal Transplantation, Assistance Publique-Hôpitaux de Paris, Groupe hospitalo-universitaire Chenevier Mondor, Fédération Hospitalo-Universitaire, Innovative therapy for immune disorders, Créteil, France
| | - Marie Matignon
- Universite Paris Est Creteil, INSERM IMRB U955, Créteil, France
- Department of Nephrology and Renal Transplantation, Assistance Publique-Hôpitaux de Paris, Groupe hospitalo-universitaire Chenevier Mondor, Fédération Hospitalo-Universitaire, Innovative therapy for immune disorders, Créteil, France
| |
Collapse
|
15
|
Garbayo E, El Moukhtari SH, Rodríguez-Nogales C, Agirre X, Rodriguez-Madoz JR, Rodriguez-Marquez P, Prósper F, Couvreur P, Blanco-Prieto MJ. RNA-loaded nanoparticles for the treatment of hematological cancers. Adv Drug Deliv Rev 2024; 214:115448. [PMID: 39303823 DOI: 10.1016/j.addr.2024.115448] [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: 02/19/2024] [Revised: 06/07/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
Hematological cancers encompass a diverse group of malignancies affecting the blood, bone marrow, lymph nodes, and spleen. These disorders present unique challenges due to their complex etiology and varied clinical manifestations. Despite significant advancements in understanding and treating hematological malignancies, innovative therapeutic approaches are continually sought to enhance patient outcomes. This review highlights the application of RNA nanoparticles (RNA-NPs) in the treatment of hematological cancers. We delve into detailed discussions on in vitro and preclinical studies involving RNA-NPs for adult patients, as well as the application of RNA-NPs in pediatric hematological cancer. The review also addresses ongoing clinical trials involving RNA-NPs and explores the emerging field of CAR-T therapy engineered by RNA-NPs. Finally, we discuss the challenges still faced in translating RNA-NP research to clinics.
Collapse
Affiliation(s)
- Elisa Garbayo
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain
| | - Souhaila H El Moukhtari
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Carlos Rodríguez-Nogales
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain
| | - Xabier Agirre
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Juan R Rodriguez-Madoz
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Paula Rodriguez-Marquez
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Felipe Prósper
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain; Departmento de Hematología and CCUN, Clínica Universidad de Navarra, University of Navarra, Avenida Pío XII 36, 31008 Pamplona, Spain
| | - Patrick Couvreur
- Institut Galien Paris-Sud, UMR CNRS 8612, Université Paris-Saclay, Orsay Cedex, France.
| | - María J Blanco-Prieto
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain.
| |
Collapse
|
16
|
Dai Y, Luo L, Wei Z, Cheng P, Luo J, Li J. The clinical efficacy of a daratumumab-based regimen in relapsed/refractory acute leukemia: a single-center experience. Ann Hematol 2024; 103:4057-4063. [PMID: 39046511 PMCID: PMC11512853 DOI: 10.1007/s00277-024-05892-9] [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: 05/21/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
Relapsed/refractory acute leukemia (R/R-AL) is associated with a low remission rate, short survival rate, and poor prognosis. Treating R/R-AL remains challenging as there is no standardized effective regimen; hence, there is a need for efficient therapies. CD38 expression has been observed in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL). Daratumumab is a humanized anti-CD38 monoclonal antibody used to treat multiple myeloma and has been reported to treat R/R-AL safely and effectively. The clinical data of 10 adult patients with R/R-AL who were treated with a daratumumab-based salvage regimen between July 2018 and May 2023 at our center were analyzed retrospectively. Seven AML and three ALL cases were included in the analysis. Seven (70%) patients showed responses to the treatments (complete response [CR], 60%; partial response [PR], 10%). Of the seven responders, three underwent allogenic stem cell transplantation (ASCT), including one who underwent a second ASCT. Among the five patients with R/R AML who had prior exposure to venetoclax, three achieved a therapeutic response (two CR and one PR) when re-treated with venetoclax in combination with daratumumab. The median follow-up time was 6.15 months (0.9-21 months). Overall survival and event-free survival rates at 12 months were 68.6% and 40.0%, respectively. The main adverse events included grade 3 febrile neutropenia (20%) and grade 3 hematological toxicities (60%). The daratumumab-based salvage regimen offers patients with R/R-AL the opportunity of remission with acceptable tolerability, creating the possibility of bridging ASCT.
Collapse
Affiliation(s)
- Yi Dai
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Lin Luo
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zhenbin Wei
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Peng Cheng
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jun Luo
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jing Li
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
17
|
Rabelink TJ, de Vries APJ. CD38 - a new target in renal immune disease. Nat Rev Nephrol 2024; 20:641-642. [PMID: 39044006 DOI: 10.1038/s41581-024-00874-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Affiliation(s)
- Ton J Rabelink
- Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands.
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands.
| | - Aiko P J de Vries
- Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
18
|
Ziccheddu B, Giannotta C, D'Agostino M, Bertuglia G, Saraci E, Oliva S, Genuardi E, Papadimitriou M, Diamond B, Corradini P, Coffey D, Landgren O, Bolli N, Bruno B, Boccadoro M, Massaia M, Maura F, Larocca A. Genomic and immune determinants of resistance to daratumumab-based therapy in relapsed refractory multiple myeloma. Blood Cancer J 2024; 14:117. [PMID: 39030183 PMCID: PMC11271515 DOI: 10.1038/s41408-024-01096-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Targeted immunotherapy combinations, including the anti-CD38 monoclonal antibody (MoAb) daratumumab, have shown promising results in patients with relapsed/refractory multiple myeloma (RRMM), leading to a considerable increase in progression-free survival. However, a large fraction of patients inevitably relapse. To understand this, we investigated 32 relapsed MM patients treated with daratumumab, lenalidomide, and dexamethasone (Dara-Rd; NCT03848676). We conducted an integrated analysis using whole-genome sequencing (WGS) and flow cytometry in patients with RRMM. WGS before and after treatment pinpointed genomic drivers associated with early progression, including RPL5 loss, APOBEC mutagenesis, and gain of function structural variants involving MYC and chromothripsis. Flow cytometry on 202 blood samples, collected every 3 months until progression for 31 patients, revealed distinct immune changes significantly impacting clinical outcomes. Progressing patients exhibited significant depletion of CD38-positive NK cells, persistence of T-cell exhaustion, and reduced depletion of regulatory T cells over time. These findings underscore the influence of immune composition and daratumumab-induced immune changes in promoting MM resistance. Integrating genomics and flow cytometry unveiled associations between adverse genomic features and immune patterns. Overall, this study sheds light on the intricate interplay between genomic complexity and the immune microenvironment driving resistance to Dara-Rd in patients with RRMM.
Collapse
Affiliation(s)
- Bachisio Ziccheddu
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Claudia Giannotta
- Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, Università di Torino, Torino, Italy
| | - Mattia D'Agostino
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Giuseppe Bertuglia
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Elona Saraci
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Stefania Oliva
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Elisa Genuardi
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Marios Papadimitriou
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Benjamin Diamond
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Paolo Corradini
- Division of Hematology and Bone Marrow Transplant, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - David Coffey
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Ola Landgren
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Niccolò Bolli
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Benedetto Bruno
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | | | - Massimo Massaia
- Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, Università di Torino, Torino, Italy
- SC Ematologia, AO S. Croce e Carle, Cuneo, Italy
| | - Francesco Maura
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA.
| | - Alessandra Larocca
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| |
Collapse
|
19
|
Bhowmick K, von Suskil M, Al-Odat OS, Elbezanti WO, Jonnalagadda SC, Budak-Alpdogan T, Pandey MK. Pathways to therapy resistance: The sheltering effect of the bone marrow microenvironment to multiple myeloma cells. Heliyon 2024; 10:e33091. [PMID: 39021902 PMCID: PMC11252793 DOI: 10.1016/j.heliyon.2024.e33091] [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: 02/16/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Multiple Myeloma (MM) is a malignant expansion of plasma cells in the bone marrow (BM), resulting in a disease characterized by symptoms of end organ damage from light chain secretion, crowding of the BM, and bone lesions. Although the past two decades have been characterized by numerous novel therapies emerging, the disease remains incurable due to intrinsic or acquired drug resistance. A major player in MM's drug resistance arises from its intimate relationship with the BM microenvironment (BMME). Through stress-inducing conditions, soluble messengers, and physical adhesion to BM elements, the BMME activates numerous pathways in the myeloma cell. This not only propagates myeloma progression through survival and growth signals, but also specific mechanisms to circumvent therapeutic actions. In this review, we provide an overview of the BMME, the role of individual components in MM survival, and various therapy-specific resistance mechanisms reported in the literature.
Collapse
Affiliation(s)
- Kuntal Bhowmick
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Max von Suskil
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Omar S. Al-Odat
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Weam Othman Elbezanti
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Hematology, MD Anderson Cancer Center at Cooper, Cooper University Health Care, Camden, NJ, USA
| | - Subash C. Jonnalagadda
- Department of Chemistry and Biochemistry, College of Science and Mathematics, Rowan University, Glassboro, NJ, USA
| | - Tulin Budak-Alpdogan
- Department of Hematology, MD Anderson Cancer Center at Cooper, Cooper University Health Care, Camden, NJ, USA
| | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| |
Collapse
|
20
|
Choudhery MS, Arif T, Mahmood R, Harris DT. CAR-T-Cell-Based Cancer Immunotherapies: Potentials, Limitations, and Future Prospects. J Clin Med 2024; 13:3202. [PMID: 38892913 PMCID: PMC11172642 DOI: 10.3390/jcm13113202] [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: 03/14/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer encompasses various elements occurring at the cellular and genetic levels, necessitating an immunotherapy capable of efficiently addressing both aspects. T cells can combat cancer cells by specifically recognizing antigens on them. This innate capability of T cells has been used to develop cellular immunotherapies, but most of them can only target antigens through major histocompatibility complexes (MHCs). New gene-editing techniques such as clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein 9 (CRISPR-cas9) can precisely edit the DNA sequences. CRISPR-cas9 has made it possible to generate genetically engineered chimeric antigen receptors (CARs) that can overcome the problems associated with old immunotherapies. In chimeric antigen receptor T (CAR-T) cell therapy, the patient's T cells are isolated and genetically modified to exhibit synthetic CAR(s). CAR-T cell treatment has shown remarkably positive clinical outcomes in cancers of various types. Nevertheless, there are various challenges that reduce CAR-T effectiveness in solid tumors. It is required to address these challenges in order to make CAR-T cell therapy a better and safer option. Combining CAR-T treatment with other immunotherapies that target multiple antigens has shown positive outcomes. Moreover, recently generated Boolean logic-gated advanced CARs along with artificial intelligence has expanded its potential to treat solid tumors in addition to blood cancers. This review aims to describe the structure, types, and various methods used to develop CAR-T cells. The clinical applications of CAR-T cells in hematological malignancies and solid tumours have been described in detail. In addition, this discussion has addressed the limitations associated with CAR-T cells, explored potential strategies to mitigate CAR-T-related toxicities, and delved into future perspectives.
Collapse
Affiliation(s)
- Mahmood S. Choudhery
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 54600, Pakistan;
| | - Taqdees Arif
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 54600, Pakistan;
| | - Ruhma Mahmood
- Jinnah Hospital, Allama Iqbal Medical College, Lahore 54700, Pakistan;
| | - David T. Harris
- Department of Immunobiology, College of Medicine, University of Arizona Health Sciences Biorepository, The University of Arizona, Tucson, AZ 85724-5221, USA;
| |
Collapse
|
21
|
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
|
22
|
Major-Monfried H, Hosszu K, McAvoy DP, Vallone A, Shukla N, Gillio A, Spitzer B, Kung AL, Cancio M, Curran K, Scaradavou A, Oved JH, O'Reilly RJ, Boelens JJ, Harris AC. Two novel assays demonstrate persistent daratumumab exposure in a pediatric patient with delayed engraftment following allogeneic hematopoietic stem cell transplantation. Cytotherapy 2024; 26:466-471. [PMID: 38430078 DOI: 10.1016/j.jcyt.2024.01.005] [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: 03/31/2023] [Revised: 11/08/2023] [Accepted: 01/25/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND AIMS Daratumumab, a human IgG monoclonal antibody targeting CD38, is a promising treatment for pediatric patients with relapsed or refractory T-cell acute lymphoblastic leukemia (T-ALL). We describe a case of delayed engraftment following a mismatched, unrelated donor hematopoietic stem cell transplant (HSCT) in a 14-year-old female with relapsed T-ALL, treated with daratumumab and chemotherapy. By Day 28 post-HSCT, the patient had no neutrophil engraftment but full donor myeloid chimerism. METHODS We developed two novel, semi-quantitative, antibody-based assays to measure the patient's bound and plasma daratumumab levels to determine if prolonged drug exposure may have contributed to her slow engraftment. RESULTS Daratumumab levels were significantly elevated more than 30 days after the patient's final infusion, and levels inversely correlated with her white blood cell counts. To clear daratumumab, the patient underwent several rounds of plasmapheresis and subsequently engrafted. CONCLUSIONS This is the first report of both delayed daratumumab clearance and delayed stem cell engraftment following daratumumab treatment in a pediatric patient. Further investigation is needed to elucidate the optimal dosing of daratumumab for treatment of acute leukemias in pediatric populations as well as daratumumab's potential effects on hematopoietic stem cells and stem cell engraftment following allogenic HSCT.
Collapse
Affiliation(s)
- Hannah Major-Monfried
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.
| | - Kinga Hosszu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Devin P McAvoy
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alexander Vallone
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alfred Gillio
- Pediatric Blood and Marrow Transplantation, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Barbara Spitzer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Pediatric Blood and Marrow Transplantation, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maria Cancio
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kevin Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andromachi Scaradavou
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joseph H Oved
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jaap Jan Boelens
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andrew C Harris
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
23
|
Lee HE, Cho AH, Hwang JH, Kim JW, Yang HR, Ryu T, Jung Y, Lee S. Development, High-Throughput Profiling, and Biopanning of a Large Phage Display Single-Domain Antibody Library. Int J Mol Sci 2024; 25:4791. [PMID: 38732011 PMCID: PMC11083953 DOI: 10.3390/ijms25094791] [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: 03/20/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Immunoglobulin G-based monoclonal antibodies (mAbs) have been effective in treating various diseases, but their large molecular size can limit their penetration of tissue and efficacy in multifactorial diseases, necessitating the exploration of alternative forms. In this study, we constructed a phage display library comprising single-domain antibodies (sdAbs; or "VHHs"), known for their small size and remarkable stability, using a total of 1.6 × 109 lymphocytes collected from 20 different alpacas, resulting in approximately 7.16 × 1010 colonies. To assess the quality of the constructed library, next-generation sequencing-based high-throughput profiling was performed, analyzing approximately 5.65 × 106 full-length VHH sequences, revealing 92% uniqueness and confirming the library's diverse composition. Systematic characterization of the library revealed multiple sdAbs with high affinity for three therapeutically relevant antigens. In conclusion, our alpaca sdAb phage display library provides a versatile resource for diagnostics and therapeutics. Furthermore, the library's vast natural VHH antibody repertoire offers insights for generating humanized synthetic sdAb libraries, further advancing sdAb-based therapeutics.
Collapse
Affiliation(s)
- Hee Eon Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (H.E.L.); (A.H.C.); (J.H.H.); (J.W.K.); (H.R.Y.)
| | - Ah Hyun Cho
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (H.E.L.); (A.H.C.); (J.H.H.); (J.W.K.); (H.R.Y.)
| | - Jae Hyeon Hwang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (H.E.L.); (A.H.C.); (J.H.H.); (J.W.K.); (H.R.Y.)
| | - Ji Woong Kim
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (H.E.L.); (A.H.C.); (J.H.H.); (J.W.K.); (H.R.Y.)
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (H.E.L.); (A.H.C.); (J.H.H.); (J.W.K.); (H.R.Y.)
| | - Taehoon Ryu
- ATG Lifetech Inc., Seoul 08507, Republic of Korea; (T.R.); (Y.J.)
| | - Yushin Jung
- ATG Lifetech Inc., Seoul 08507, Republic of Korea; (T.R.); (Y.J.)
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea; (H.E.L.); (A.H.C.); (J.H.H.); (J.W.K.); (H.R.Y.)
- Department of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
24
|
Parrondo RD, Ailawadhi S, Cerchione C. Bispecific antibodies for the treatment of relapsed/refractory multiple myeloma: updates and future perspectives. Front Oncol 2024; 14:1394048. [PMID: 38660139 PMCID: PMC11039948 DOI: 10.3389/fonc.2024.1394048] [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: 02/29/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Patients with relapsed/refractory multiple myeloma (RRMM) that are refractory to the five most active anti-MM drugs, so-called penta-refractory MM, have historically had dismal outcomes with subsequent therapies. Progressive immune dysfunction, particularly of the T-cell repertoire, is implicated in the development of disease progression and refractory disease. However, the advent of novel immunotherapies such as bispecific antibodies are rapidly changing the treatment landscape and improving the survival outcomes of patients with RRMM. Bispecific antibodies are antibodies that are engineered to simultaneously engage cytotoxic immune effector cells (T cells or NK cells) and malignant plasma cells via binding to immune effector cell antigens and extracellular plasma cell antigens leading to immune effector cell activation and malignant plasma cell destruction. Currently, bispecific antibodies that bind CD3 on T cells and plasma cell epitopes such as B-cell maturation antigen (BCMA), G-protein coupled receptor family C group 5 member D (GPRC5d), and Fc receptor homologue 5 (FcRH5) are the most advanced in clinical development and are showing unprecedented response rates in patients with RRMM, including patients with penta-refractory disease. In this review article, we explore the available clinical data of bispecific antibodies in RRMM and summarize the efficacy, safety, toxicity, clinical outcomes, mechanisms of resistance, and future directions of these therapies in patients with RRMM.
Collapse
Affiliation(s)
- Ricardo D. Parrondo
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, and Cellular Therapies, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| | - Sikander Ailawadhi
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, and Cellular Therapies, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| | - Claudio Cerchione
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
25
|
Nolan RP, Printz MA. Modeling the subcutaneous pharmacokinetics of antibodies co-administered with rHuPH20. Clin Transl Sci 2024; 17:e13788. [PMID: 38561908 PMCID: PMC10985223 DOI: 10.1111/cts.13788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Predicting the subcutaneous (SC) pharmacokinetics (PK) of antibodies in humans is challenging, with clinical data currently being the only reliable data source for modeling SC absorption and bioavailability. Recombinant human hyaluronidase PH20 (rHuPH20) is an enzyme that facilitates SC delivery of high-dose, high-volume therapeutics. Numerous monoclonal antibodies have been co-administered SC with rHuPH20 in a clinical setting, establishing an extensive PK database. The goal of this work is to demonstrate how aggregated clinical data can be leveraged in a universal modeling framework for characterizing SC antibody PK, resulting in parameterization that can be used in predictive simulations of new antibodies. Data for 10 individual antibodies co-administered SC with rHuPH20 were obtained from publicly available sources. PK modeling of each antibody was conducted using the same model structure, but uniquely parameterized. The model structure consisted of a two-compartment model to capture linear kinetics, plus a target-binding mechanism to accommodate nonlinear kinetics driven by antibody-target complex formation and elimination. The clinical PK profiles for all antibodies were accurately described using the universal modeling framework. The SC PK parameters of absorption and bioavailability were consistent across the range of antibody and target properties evaluated. SC administration with rHuPH20 yielded a 30% increase in absorption rate on average and similar or better bioavailability. These parameter values can serve as initial conditions for model-based PK predictions for new antibodies co-administered SC with rHuPH20 to enable evaluation of optimal SC dose and schedule regimens prior to and during clinical development.
Collapse
|
26
|
Grandclément C, Estoppey C, Dheilly E, Panagopoulou M, Monney T, Dreyfus C, Loyau J, Labanca V, Drake A, De Angelis S, Rubod A, Frei J, Caro LN, Blein S, Martini E, Chimen M, Matthes T, Kaya Z, Edwards CM, Edwards JR, Menoret E, Kervoelen C, Pellat-Deceunynck C, Moreau P, Mbow ML, Srivastava A, Dyson MR, Zhukovsky EA, Perro M, Sammicheli S. Development of ISB 1442, a CD38 and CD47 bispecific biparatopic antibody innate cell modulator for the treatment of multiple myeloma. Nat Commun 2024; 15:2054. [PMID: 38448430 PMCID: PMC10917784 DOI: 10.1038/s41467-024-46310-y] [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: 07/31/2023] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
Antibody engineering can tailor the design and activities of therapeutic antibodies for better efficiency or other advantageous clinical properties. Here we report the development of ISB 1442, a fully human bispecific antibody designed to re-establish synthetic immunity in CD38+ hematological malignancies. ISB 1442 consists of two anti-CD38 arms targeting two distinct epitopes that preferentially drive binding to tumor cells and enable avidity-induced blocking of proximal CD47 receptors on the same cell while preventing on-target off-tumor binding on healthy cells. The Fc portion of ISB 1442 is engineered to enhance complement dependent cytotoxicity, antibody dependent cell cytotoxicity and antibody dependent cell phagocytosis. ISB 1442 thus represents a CD47-BsAb combining biparatopic targeting of a tumor associated antigen with engineered enhancement of antibody effector function to overcome potential resistance mechanisms that hamper treatment of myeloma with monospecific anti-CD38 antibodies. ISB 1442 is currently in a Phase I clinical trial in relapsed refractory multiple myeloma.
Collapse
Affiliation(s)
| | - C Estoppey
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - E Dheilly
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | | | - T Monney
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - C Dreyfus
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - J Loyau
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - V Labanca
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - A Drake
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - S De Angelis
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - A Rubod
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - J Frei
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - L N Caro
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - S Blein
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - E Martini
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - M Chimen
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - T Matthes
- Haematology Service, Department of Oncology and Clinical Pathology Service, Department of Diagnostics, University Hospital Geneva, 1211, Geneva, Switzerland
| | - Z Kaya
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, UK
| | - C M Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, UK
| | - J R Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, UK
| | - E Menoret
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - C Kervoelen
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - C Pellat-Deceunynck
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
- SIRIC ILIAD, Angers, Nantes, France
| | - P Moreau
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
- SIRIC ILIAD, Angers, Nantes, France
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU, Nantes, France
| | - M L Mbow
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - A Srivastava
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - M R Dyson
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - E A Zhukovsky
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - M Perro
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland.
| | - S Sammicheli
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland.
| |
Collapse
|
27
|
Ziccheddu B, Giannotta C, D'Agostino M, Bertuglia G, Saraci E, Oliva S, Genuardi E, Papadimitriou M, Diamond B, Corradini P, Coffey D, Landgren O, Bolli N, Bruno B, Boccadoro M, Massaia M, Maura F, Larocca A. Genomic and immune determinants of resistance to anti-CD38 monoclonal antibody-based therapy in relapsed refractory multiple myeloma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.04.23299287. [PMID: 38106151 PMCID: PMC10723485 DOI: 10.1101/2023.12.04.23299287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Anti-CD38 antibody therapies have transformed multiple myeloma (MM) treatment. However, a large fraction of patients inevitably relapses. To understand this, we investigated 32 relapsed MM patients treated with daratumumab, lenalidomide, and dexamethasone (Dara-Rd; NCT03848676 ). Whole genome sequencing (WGS) before and after treatment pinpointed genomic drivers associated with early progression, including RPL5 loss and APOBEC mutagenesis. Flow cytometry on 202 blood samples, collected every three months until progression for 31 patients, revealed distinct immune changes significantly impacting clinical outcomes. Progressing patients exhibited significant depletion of CD38+ NK cells, persistence of T cell exhaustion, and reduced depletion of T-reg cells over time. These findings underscore the influence of immune composition and daratumumab-induced immune changes in promoting MM resistance. Integrating genomics and flow cytometry unveiled associations between adverse genomic features and immune patterns. Overall, this study sheds light on the intricate interplay between genomic complexity and the immune microenvironment driving resistance to Dara-Rd.
Collapse
|
28
|
Majeed J, Sabbagh MN, Kang MH, Lawrence JJ, Pruitt K, Bacus S, Reyna E, Brown M, Decourt B. Cancer drugs with high repositioning potential for Alzheimer's disease. Expert Opin Emerg Drugs 2023; 28:311-332. [PMID: 38100555 PMCID: PMC10877737 DOI: 10.1080/14728214.2023.2296079] [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: 08/25/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Despite the recent full FDA approval of lecanemab, there is currently no disease modifying therapy (DMT) that can efficiently slow down the progression of Alzheimer's disease (AD) in the general population. This statement emphasizes the need to identify novel DMTs in the shortest time possible to prevent a global epidemic of AD cases as the world population experiences an increase in lifespan. AREAS COVERED Here, we review several classes of anti-cancer drugs that have been or are being investigated in Phase II/III clinical trials for AD, including immunomodulatory drugs, RXR agonists, sex hormone therapies, tyrosine kinase inhibitors, and monoclonal antibodies. EXPERT OPINION Given the overall course of brain pathologies during the progression of AD, we express a great enthusiasm for the repositioning of anti-cancer drugs as possible AD DMTs. We anticipate an increasing number of combinatorial therapy strategies to tackle AD symptoms and their underlying pathologies. However, we strongly encourage improvements in clinical trial study designs to better assess target engagement and possible efficacy over sufficient periods of drug exposure.
Collapse
Affiliation(s)
- Jad Majeed
- University of Arizona Honors College, Tucson, Arizona, USA
| | - Marwan N. Sabbagh
- Alzheimer’s and Memory Disorders Division, Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Min H. Kang
- Department of Pediatrics, Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - J. Josh Lawrence
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kevin Pruitt
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Ellie Reyna
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Maddy Brown
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
- Roseman University of Health Sciences, Las Vegas, Nevada, USA
| |
Collapse
|
29
|
van de Donk NWCJ, O'Neill C, de Ruijter MEM, Verkleij CPM, Zweegman S. T-cell redirecting bispecific and trispecific antibodies in multiple myeloma beyond BCMA. Curr Opin Oncol 2023; 35:601-611. [PMID: 37501530 PMCID: PMC10566598 DOI: 10.1097/cco.0000000000000983] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
PURPOSE OF REVIEW B-cell maturation antigen (BCMA)-directed T-cell immunotherapies, such as chimeric antigen receptor T-cells (CAR T-cells) and bispecific antibodies (BsAbs) have markedly improved the survival of triple-class refractory multiple myeloma (MM). However, the majority of patients still develops disease progression, underlining the need for new agents for these patients. RECENT FINDINGS Novel T-cell redirecting BsAbs targeting alternative tumor-associated antigens have shown great promise in heavily pretreated MM, including patients previously exposed to BCMA-directed therapies. This includes the G-protein-coupled receptor class 5 member D (GPRC5D)-targeting BsAbs talquetamab and forimtamig, as well as the Fc receptor-homolog 5 (FcRH5)-targeting BsAb cevostamab. Toxicity associated with these BsAbs includes cytokine-release syndrome, cytopenias, and infections. In addition, GPRC5D-targeting BsAbs are associated with specific 'on target/off tumor' toxicities including rash, nail disorders, and dysgeusia. Trispecifc antibodies targeting two different MM-associated antigens to prevent antigen escape are in early clinical development, as well as trispecific antibodies (TsAbs) that provide an additional co-stimulatory signal to T-cells to prevent their exhaustion. SUMMARY Various T-cell redirecting BsAbs are in advanced stages of clinical development with promising activity and a manageable toxicity profile. Ongoing studies are evaluating combination strategies, fixed-duration treatment, and use of BsAbs in earlier lines of therapy. TsAbs hold great promise for the future.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Chloe O'Neill
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Maaike E M de Ruijter
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Bisht K, Fukao T, Chiron M, Richardson P, Atanackovic D, Chini E, Chng WJ, Van De Velde H, Malavasi F. Immunomodulatory properties of CD38 antibodies and their effect on anticancer efficacy in multiple myeloma. Cancer Med 2023; 12:20332-20352. [PMID: 37840445 PMCID: PMC10652336 DOI: 10.1002/cam4.6619] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND CD38 has been established as an important therapeutic target for multiple myeloma (MM), for which two CD38 antibodies are currently approved-daratumumab and isatuximab. CD38 is an ectoenzyme that degrades NAD and its precursors and is involved in the production of adenosine and other metabolites. AIM Among the various mechanisms by which CD38 antibodies can induce MM cell death is immunomodulation, including multiple pathways for CD38-mediated T-cell activation. Patients who respond to anti-CD38 targeting treatment experience more marked changes in T-cell expansion, activity, and clonality than nonresponders. IMPLICATIONS Resistance mechanisms that undermine the immunomodulatory effects of CD38-targeting therapies can be tumor intrinsic, such as the downregulation of CD38 surface expression and expression of complement inhibitor proteins, and immune microenvironment-related, such as changes to the natural killer (NK) cell numbers and function in the bone marrow niche. There are numerous strategies to overcome this resistance, which include identifying and targeting other therapeutic targets involved in, for example, adenosine production, the activation of NK cells or monocytes through immunomodulatory drugs and their combination with elotuzumab, or with bispecific T-cell engagers.
Collapse
Affiliation(s)
| | - Taro Fukao
- Sanofi OncologyCambridgeMassachusettsUSA
| | | | - Paul Richardson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma CenterDana Farber Cancer Institute, Harvard Medical SchoolBostonMassachusettsUSA
| | - Djordje Atanackovic
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer CenterBaltimoreMarylandUSA
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Eduardo Chini
- Department of Anesthesiology and Perioperative MedicineMayo ClinicJacksonvilleFloridaUSA
| | - Wee Joo Chng
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | | | - Fabio Malavasi
- Department of Medical SciencesUniversity of TurinTorinoItaly
- Fondazione Ricerca MolinetteTorinoItaly
| |
Collapse
|
31
|
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
|
32
|
Chemlal D, Varlet E, Machura A, Ovejero S, Requirand G, Robert N, Cartron G, Alaterre E, Bret C, Vincent L, Herbaux C, Cavalli G, Bruyer A, De Boussac H, Moreaux J. EZH2 targeting induces CD38 upregulation and response to anti-CD38 immunotherapies in multiple myeloma. Leukemia 2023; 37:1925-1928. [PMID: 37532787 PMCID: PMC10457196 DOI: 10.1038/s41375-023-01983-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/12/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Affiliation(s)
- Djamila Chemlal
- Diag2Tec, Montpellier, France
- Institute of Human Genetics, UMR CNRS-UM, 9002, Montpellier, France
| | - Emmanuel Varlet
- Institute of Human Genetics, UMR CNRS-UM, 9002, Montpellier, France
| | | | - Sara Ovejero
- Institute of Human Genetics, UMR CNRS-UM, 9002, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Guilhem Requirand
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Nicolas Robert
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Guillaume Cartron
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
- University of Montpellier, UFR Medicine, Montpellier, France
| | - Elina Alaterre
- Institute of Human Genetics, UMR CNRS-UM, 9002, Montpellier, France
| | - Caroline Bret
- Institute of Human Genetics, UMR CNRS-UM, 9002, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
- University of Montpellier, UFR Medicine, Montpellier, France
| | - Laure Vincent
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
| | - Charles Herbaux
- Institute of Human Genetics, UMR CNRS-UM, 9002, Montpellier, France
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
- University of Montpellier, UFR Medicine, Montpellier, France
| | - Giacomo Cavalli
- Institute of Human Genetics, UMR CNRS-UM, 9002, Montpellier, France
| | | | | | - Jerome Moreaux
- Institute of Human Genetics, UMR CNRS-UM, 9002, Montpellier, France.
- Department of Biological Hematology, CHU Montpellier, Montpellier, France.
- University of Montpellier, UFR Medicine, Montpellier, France.
- Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
33
|
Sunami K, Fuchida SI, Suzuki K, Ri M, Matsumoto M, Shimazaki C, Asaoku H, Shibayama H, Ishizawa K, Takamatsu H, Ikeda T, Maruyama D, Imada K, Uchiyama M, Kiguchi T, Iyama S, Murakami H, Onishi R, Tada K, Iida S. Anti-CD38 antibody isatuximab monotherapy for Japanese individuals with relapsed/refractory multiple myeloma: An update of the phase 1/2 ISLANDs study. Hematol Oncol 2023; 41:442-452. [PMID: 36433829 DOI: 10.1002/hon.3105] [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: 08/23/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/28/2022]
Abstract
The primary analysis of the phase 1/2 ISLANDs study in Japanese individuals with relapsed/refractory multiple myeloma (RRMM) showed that isatuximab monotherapy was well tolerated and effective, even in participants with high-risk cytogenetic abnormalities. Here, we report a prespecified second analysis conducted 20 months after the first dosing of the last participant (ClinicalTrials.gov identifier: NCT02812706). The primary objectives were to evaluate the safety and tolerability of isatuximab in phase 1 and to evaluate the efficacy of isatuximab, including assessment of overall response rate (ORR) at the recommended dose (RD), in phase 2. In phase 1, three participants received isatuximab 10 mg/kg every week (QW) for 4 weeks/cycle followed by every 2 weeks (Q2W) and five participants received 20 mg/kg QW/Q2W. Since no dose-limiting toxicities occurred in phase 1, 20 mg/kg QW/Q2W was identified as the RD for the phase 2 study (n = 28). At the time of data cut-off, three participants (one in phase 1 and two in phase 2) continued to receive isatuximab; disease progression and treatment-related adverse events were the most common reasons for treatment discontinuation. The overall safety profile was consistent with the primary analysis. One death, not related to isatuximab treatment, was reported since the first analysis. The ORR and clinical benefit rate remained unchanged from the primary analysis at 36.4% (95% confidence interval [CI]: 20.4%-54.9%) and 54.5% (95% CI: 36.4%-71.9%), respectively. The median progression-free survival (PFS) was 5.6 months, longer than the median PFS reported in the primary analysis (4.7 months), whereas median overall survival was not reached. Overall, isatuximab 20 mg/kg QW/Q2W had an acceptable safety and tolerability profile and showed promising antitumor activity in Japanese individuals with RRMM.
Collapse
Affiliation(s)
- Kazutaka Sunami
- Department of Hematology, National Hospital Organization, Okayama Medical Center, Okayama, Japan
| | - Shin-Ichi Fuchida
- Department of Hematology, Japan Community Health Care Organization Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Kenshi Suzuki
- Myeloma/Amyloidosis Center, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Masaki Ri
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| | - Morio Matsumoto
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Shibukawa, Japan
| | - Chihiro Shimazaki
- Department of Hematology, Japan Community Health Care Organization Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | | | - Hirohiko Shibayama
- Department of Hematology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Kenichi Ishizawa
- Department of Third Internal Medicine, Division of Hematology and Cell Therapy, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Hiroyuki Takamatsu
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Takashi Ikeda
- Division of Hematology and Stem Cell Transplantation, Shizuoka Cancer Center, Shizuoka, Japan
| | - Dai Maruyama
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
| | - Kazunori Imada
- Department of Hematology, Japanese Red Cross Osaka Hospital, Osaka, Japan
| | - Michihiro Uchiyama
- Department of Hematology, Japanese Red Cross Society Suwa Hospital, Suwa, Japan
| | - Toru Kiguchi
- Department of Hematology, Chugoku Central Hospital, Fukuyama, Japan
- Division of Diabetes, Endocrinology and Hematology, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | - Satoshi Iyama
- Department of Hematology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirokazu Murakami
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan
| | | | | | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya, Japan
| |
Collapse
|
34
|
Morfino P, Aimo A, Castiglione V, Chianca M, Vergaro G, Cipolla CM, Fedele A, Emdin M, Fabiani I, Cardinale D. Cardiovascular toxicity from therapies for light chain amyloidosis. Front Cardiovasc Med 2023; 10:1212983. [PMID: 37476571 PMCID: PMC10354454 DOI: 10.3389/fcvm.2023.1212983] [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: 05/03/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023] Open
Abstract
Amyloid light-chain (AL) amyloidosis is a hematological disorder characterized by abnormal proliferation of a plasma cell clone producing monoclonal free light chains that misfold and aggregate into insoluble fibrils in various tissues. Cardiac involvement is a common feature leading to restrictive cardiomyopathy and poor prognosis. Current first-line treatments aim at achieving hematological response by targeting the plasma cell clones, and these have been adapted from multiple myeloma therapy. Patients with AL amyloidosis often exhibit multiorgan involvement, making them susceptible to cancer therapy-related cardiovascular toxicity. Managing AL amyloidosis is a complex issue that requires enhanced knowledge of the cardio-oncological implications of hematological treatments. Future research should focus on implementing and validating primary and secondary prevention strategies and understanding the biochemical basis of oncological therapy-related damage to mitigate cardiovascular toxicity.
Collapse
Affiliation(s)
- Paolo Morfino
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Alberto Aimo
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Vincenzo Castiglione
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michela Chianca
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Giuseppe Vergaro
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| | - Antonella Fedele
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| | - Michele Emdin
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Iacopo Fabiani
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Daniela Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| |
Collapse
|
35
|
Yadav S, Gundeti S, Bhave A, Deb U, Dixit J, Mishra K. Role of daratumumab in the frontline management of multiple myeloma: a narrative review. Expert Rev Hematol 2023; 16:743-760. [PMID: 37585685 DOI: 10.1080/17474086.2023.2246651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/31/2023] [Accepted: 08/07/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION The prevalence of multiple myeloma (MM) has gradually increased over the last few decades in India due to growing population, better disease awareness, and improved diagnostic procedures. Despite such advances, MM remains an incurable and relapsing disease due to its heterogeneity and genomic instability. With the inclusion of monoclonal antibodies, especially daratumumab in the frontline regimen, the management landscape of MM has improved significantly resulting in better disease control and patient outcomes. AREAS COVERED This review aims to provide an in-depth summary of efficacy and safety of frontline daratumumab therapy in treatment of MM including patients with high-risk cytogenetic profile. EXPERT OPINION Based on the review of literature, daratumumab in frontline therapy has demonstrated improved efficacy in terms of reduction in disease progression or death, and superior minimal residual disease (MRD)-negativity rates with an acceptable safety profile in patients with newly diagnosed MM (NDMM) including patients with high-risk cytogenetic profile. Daratumumab alone or in combination with other drugs has shown similar clinical outcomes in patients with relapsed/refractory MM. Hence, daratumumab can be used upfront in patients with MM.
Collapse
Affiliation(s)
- Sanjeev Yadav
- Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Sadashivudu Gundeti
- Department of Medical Oncology, Nizam's Institute of Medical Sciences, Hyderabad, India
| | - Abhay Bhave
- Department of Hematology, Lilavati Hospital and Research Centre, Mumbai, India
| | - Uttiya Deb
- Medical Affairs, Johnson and Johnson Private Limited, Mumbai, India
| | - Jitendra Dixit
- Medical Affairs, Johnson and Johnson Private Limited, Mumbai, India
| | - Kundan Mishra
- Department of Hematology, Command Hospital, Lucknow, India
| |
Collapse
|
36
|
Hiemstra IH, Santegoets KCM, Janmaat ML, De Goeij BECG, Ten Hagen W, van Dooremalen S, Boross P, van den Brakel J, Bosgra S, Andringa G, van Kessel-Welmers B, Verzijl D, Hibbert RG, Frerichs KA, Mutis T, van de Donk NWCJ, Ahmadi T, Satijn D, Sasser AK, Breij ECW. Preclinical anti-tumour activity of HexaBody-CD38, a next-generation CD38 antibody with superior complement-dependent cytotoxic activity. EBioMedicine 2023; 93:104663. [PMID: 37379657 DOI: 10.1016/j.ebiom.2023.104663] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND HexaBody®-CD38 (GEN3014) is a hexamerization-enhanced human IgG1 that binds CD38 with high affinity. The E430G mutation in its Fc domain facilitates the natural process of antibody hexamer formation upon binding to the cell surface, resulting in increased binding of C1q and potentiated complement-dependent cytotoxicity (CDC). METHODS Co-crystallization studies were performed to identify the binding interface of HexaBody-CD38 and CD38. HexaBody-CD38-induced CDC, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), trogocytosis, and apoptosis were assessed using flow cytometry assays using tumour cell lines, and MM patient samples (CDC). CD38 enzymatic activity was measured using fluorescence spectroscopy. Anti-tumour activity of HexaBody-CD38 was assessed in patient-derived xenograft mouse models in vivo. FINDINGS HexaBody-CD38 binds a unique epitope on CD38 and induced potent CDC in multiple myeloma (MM), acute myeloid leukaemia (AML), and B-cell non-Hodgkin lymphoma (B-NHL) cells. Anti-tumour activity was confirmed in patient-derived xenograft models in vivo. Sensitivity to HexaBody-CD38 correlated with CD38 expression level and was inversely correlated with expression of complement regulatory proteins. Compared to daratumumab, HexaBody-CD38 showed enhanced CDC in cell lines with lower levels of CD38 expression, without increasing lysis of healthy leukocytes. More effective CDC was also confirmed in primary MM cells. Furthermore, HexaBody-CD38 efficiently induced ADCC, ADCP, trogocytosis, and apoptosis after Fc-crosslinking. Moreover, HexaBody-CD38 strongly inhibited CD38 cyclase activity, which is hypothesized to relieve immune suppression in the tumour microenvironment. INTERPRETATION Based on these preclinical studies, a clinical trial was initiated to assess the clinical safety of HexaBody-CD38 in patients with MM. FUNDING Genmab.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Kristine A Frerichs
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tuna Mutis
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Niels W C J van de Donk
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | |
Collapse
|
37
|
Chen C, Yan W, Tao M, Fu Y. NAD + Metabolism and Immune Regulation: New Approaches to Inflammatory Bowel Disease Therapies. Antioxidants (Basel) 2023; 12:1230. [PMID: 37371959 DOI: 10.3390/antiox12061230] [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: 03/27/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Inflammatory bowel disease (IBD), which includes Crohn's disease (CD) and ulcerative colitis (UC), is a multifactorial systemic inflammatory immune response. Nicotinamide adenine dinucleotide (NAD+) is a co-enzyme involved in cell signaling and energy metabolism. Calcium homeostasis, gene transcription, DNA repair, and cell communication involve NAD+ and its degradation products. There is a growing recognition of the intricate relationship between inflammatory diseases and NAD+ metabolism. In the case of IBD, the maintenance of intestinal homeostasis relies on a delicate balance between NAD+ biosynthesis and consumption. Consequently, therapeutics designed to target the NAD+ pathway are promising for the management of IBD. This review discusses the metabolic and immunoregulatory processes of NAD+ in IBD to examine the molecular biology and pathophysiology of the immune regulation of IBD and to provide evidence and theoretical support for the clinical use of NAD+ in IBD.
Collapse
Affiliation(s)
- Chaoyue Chen
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Meihui Tao
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Fu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
38
|
Gerasimenko M, Higashida H. Remission of social behavior impairment by oral administration of a precursor of NAD in CD157, but not in CD38, knockout mice. Front Immunol 2023; 14:1166609. [PMID: 37215105 PMCID: PMC10192747 DOI: 10.3389/fimmu.2023.1166609] [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: 02/15/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a substrate of adenosine diphosphate (ADP)-ribosyl cyclase and is catalyzed to cyclic ADP-ribose (cADPR) by CD38 and/or CD157. cADPR, a Ca2+ mobilizing second messenger, is critical in releasing oxytocin from the hypothalamus into the brain. Although NAD precursors effectively play a role in neurodegenerative disorders, muscular dystrophy, and senescence, the beneficial effects of elevating NAD by NAD precursor supplementation on brain function, especially social interaction, and whether CD38 is required in this response, has not been intensely studied. Here, we report that oral gavage administration of nicotinamide riboside, a perspective NAD precursor with high bioavailability, for 12 days did not show any suppressive or increasing effects on sociability (mouse's interest in social targets compared to non-social targets) in both CD157KO and CD38KO male mice models in a three-chamber test. CD157KO and CD38KO mice displayed no social preference (that is, more interest towards a novel mouse than a familiar one) behavior. This defect was rescued after oral gavage administration of nicotinamide riboside for 12 days in CD157KO mice, but not in CD38KO mice. Social memory was not observed in CD157KO and CD38KO mice; subsequently, nicotinamide riboside administration had no effect on social memory. Together with the results that nicotinamide riboside had essentially no or little effect on body weight during treatment in CD157KO mice, nicotinamide riboside is less harmful and has beneficial effect on defects in recovery from social behavioral, for which CD38 is required in mice.
Collapse
Affiliation(s)
- Maria Gerasimenko
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Haruhiro Higashida
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
39
|
Braun T, Schrader A. Education and Empowering Special Forces to Eradicate Secret Defectors: Immune System-Based Treatment Approaches for Mature T- and NK-Cell Malignancies. Cancers (Basel) 2023; 15:cancers15092532. [PMID: 37173999 PMCID: PMC10177197 DOI: 10.3390/cancers15092532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Mature T- and NK-cell leukemia/lymphoma (MTCL/L) constitute a heterogeneous group of, currently, 30 distinct neoplastic entities that are overall rare, and all present with a challenging molecular markup. Thus, so far, the use of first-line cancer treatment modalities, including chemotherapies, achieve only limited clinical responses associated with discouraging prognoses. Recently, cancer immunotherapy has evolved rapidly, allowing us to help patients with, e.g., solid tumors and also relapsed/refractory B-cell malignancies to achieve durable clinical responses. In this review, we systematically unveiled the distinct immunotherapeutic approaches available, emphasizing the special impediments faced when trying to employ immune system defense mechanisms to target 'one of their own-gone mad'. We summarized the preclinical and clinical efforts made to employ the various platforms of cancer immunotherapies including antibody-drug conjugates, monoclonal as well as bispecific antibodies, immune-checkpoint blockades, and CAR T cell therapies. We emphasized the challenges to, but also the goals of, what needs to be done to achieve similar successes as seen for B-cell entities.
Collapse
Affiliation(s)
- Till Braun
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases, Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
| | - Alexandra Schrader
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases, Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
- Lymphoma Immuno Biology Team, Equipe Labellisée LIGUE 2023, Centre International de Recherche en Infectiologie, INSERM U1111-CNRS UMR5308, Faculté de Médecine Lyon-Sud, Hospices Civils de Lyon, Université Claude Bernard Lyon I-ENS de Lyon, 69921 Lyon, France
| |
Collapse
|
40
|
Kale A, Lech M, Anders HJ, Gaikwad AB. Lupus Nephritis: New and Emerging Biologic and Targeted Therapies. BioDrugs 2023:10.1007/s40259-023-00597-3. [PMID: 37093522 DOI: 10.1007/s40259-023-00597-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2023] [Indexed: 04/25/2023]
Abstract
Lupus nephritis (LN) is a severe complication of systemic lupus erythematosus (SLE), a polyclonal systemic autoimmunity directed against nuclear and other self-antigens. SLE/LN affects mostly females during childbearing age, which puts them at risk for the progression of chronic kidney disease (CKD), cardiovascular disease, and pregnancy complications. The current management of LN involves the use of drugs with significant toxicities, and despite many attempts at novel drug interventions, the overall treatment efficacy has remained low. In this article, we discuss recent drug approvals and the upcoming pipeline of novel medications tested in clinical trials to improve effectiveness in terms of LN disease activity, LN relapse, and progression of LN-related CKD. In this context, we discuss (1) drugs with the potential to achieve these treatment goals by modulating SLE activity as the driving force for LN (e.g., belimumab, obinutuzumab, anifrolumab, and others); (2) drugs with SLE-non specific renoprotective effects by targeting non-immune mechanisms of LN progression (dapagliflozin, empagliflozin); and (3) drugs with dual immunosuppressive and antiproteinuric effects (voclosporin). Increasing the number of possible drug options will help to improve the management of LN in terms of efficacy and safety, and enable a more personalized treatment approach.
Collapse
Affiliation(s)
- Ajinath Kale
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, Rajasthan, 333 031, India
| | - Maciej Lech
- Division of Nephrology, Department of Internal Medicine IV, Hospital of the Ludwig Maximilians University Munich, 80336, Munich, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Internal Medicine IV, Hospital of the Ludwig Maximilians University Munich, 80336, Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, Rajasthan, 333 031, India.
| |
Collapse
|
41
|
Deng L, Xu G. Update on the Application of Monoclonal Antibody Therapy in Primary Membranous Nephropathy. Drugs 2023; 83:507-530. [PMID: 37017915 DOI: 10.1007/s40265-023-01855-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
When first introduced, rituximab (RTX), a chimeric anti-CD20 monoclonal antibody, brought about an alternative therapeutic paradigm for primary membranous nephropathy (PMN). Rituximab was shown to be effective and safe in PMN patients with kidney dysfunction, with. patients receiving second-line rituximab therapy achieving remission as effectively as those patients who had not previously received immunotherapy. No safety issues were reported. The B cell-driven protocol seems to be as efficient as the 375 mg/m2 × 4 regimen or 1 g × 2 regimen in achieving B cell depletion and remission, but patients with high M-type phospholipase A2 receptor (PLA2R) antibody levels may benefit from a higher dose of rituximab. While rituximab added another therapeutic option to the treatment regimen, it does have limitations as 20 to 40% of patients do not respond. Not all patients respond to RTX therapy for lymphoproliferative disorders either, therefore further novel anti-CD20 monoclonal antibodies have been developed and these may provide alternative therapeutic options for PMN. Ofatumumab, a fully human monoclonal antibody, specifically recognizes an epitope encompassing both the small and large extracellular loops of the CD20 molecule, resulting in increased complement-dependent cytotoxic activity. Ocrelizumab binds an alternative but overlapping epitope region to rituximab and displays enhanced antibody-dependent cellular cytotoxic (ADCC) activities. Obinutuzumab is designed to have a modified elbow-hinge amino acid sequence, leading to increased direct cell death induction and ADCC activities. In PMN clinical studies, ocrelizumab and obinutuzumab showed promising results, while ofatumumab displayed mixed results. However, there is a lack of randomized controlled trials with large samples, especially direct head-to-head comparisons. Alternative molecular mechanisms have been suggested in this context to explore novel therapeutic strategies. B cell activator-targeted, plasma cell-targeted and complement-directed treatments may lead to novel therapy paradigms for PMN. Exploratory strategies for the use of drugs with different mechanisms, such as a combination of rituximab and cyclophosphamide and a steroid, a combination of rituximab and a calcineurin inhibitor, may provide more rapid and efficient remission, but the combination of standard immunosuppression with rituximab could increase infection risk.
Collapse
Affiliation(s)
- Le Deng
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
42
|
Ye ZH, Yu WB, Huang MY, Chen J, Lu JJ. Building on the backbone of CD47-based therapy in cancer: Combination strategies, mechanisms, and future perspectives. Acta Pharm Sin B 2023; 13:1467-1487. [PMID: 37139405 PMCID: PMC10149906 DOI: 10.1016/j.apsb.2022.12.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/10/2022] [Accepted: 11/18/2022] [Indexed: 12/27/2022] Open
Abstract
Described as a "don't eat me" signal, CD47 becomes a vital immune checkpoint in cancer. Its interaction with signal regulatory protein alpha (SIRPα) prevents macrophage phagocytosis. In recent years, a growing body of evidences have unveiled that CD47-based combination therapy exhibits a superior anti-cancer effect. Latest clinical trials about CD47 have adopted the regimen of collaborating with other therapies or developing CD47-directed bispecific antibodies, indicating the combination strategy as a general trend of the future. In this review, clinical and preclinical cases about the current combination strategies targeting CD47 are collected, their underlying mechanisms of action are discussed, and ideas from future perspectives are shared.
Collapse
Affiliation(s)
- Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Wei-Bang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Jun Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, University of Macau, Macao 999078, China
| |
Collapse
|
43
|
Mahadevia H, Ponvilawan B, Sharma P, Al-Obaidi A, Qasim H, Koyi J, Anwer F, Raza S. Advancements and future trends of immunotherapy in light-chain amyloidosis. Crit Rev Oncol Hematol 2023; 183:103917. [PMID: 36696931 DOI: 10.1016/j.critrevonc.2023.103917] [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: 08/30/2022] [Revised: 12/05/2022] [Accepted: 01/20/2023] [Indexed: 01/23/2023] Open
Abstract
Light-chain (AL) amyloidosis is a type of plasma cell neoplasm with abnormal monoclonal immunoglobulin light-chain production and their subsequent deposition in tissues causing end-organ damage. In addition to existing treatments including autologous stem cell transplantation, there is a need for other approaches for eradicating abnormal plasma cells and amyloid tissue deposits. Treatment strategies of AL amyloidosis are mostly based on medications that are effective in multiple myeloma due to similar cell of origin. Daratumumab along with proteasome inhibitors and corticosteroids has become standard of care for AL amyloidosis. Another appealing approach is disassembling amyloid deposits with hope to potentially reverse the damage done by the disease. This was met with promising results for CAEL-101 and birtamimab. Although still in early stages, novel treatment options in pipeline, including antibody-drug conjugates, bispecific T-cell engagers, and chimeric antigen receptor T cell therapy may diversify the treatment armamentarium of AL amyloidosis in the future.
Collapse
Affiliation(s)
- Himil Mahadevia
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Ben Ponvilawan
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Parth Sharma
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Ammar Al-Obaidi
- Department of Hematology/Oncology, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Hana Qasim
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Jagadish Koyi
- Department of Internal Medicine, University of Missouri-Kansas City, 2411 Holmes St, Kansas City, MO 64108, USA.
| | - Faiz Anwer
- Taussig Cancer Center, Cleveland Clinic, 10201 Carnegie Ave, Cleveland, OH 44106, USA.
| | - Shahzad Raza
- Taussig Cancer Center, Cleveland Clinic, 10201 Carnegie Ave, Cleveland, OH 44106, USA.
| |
Collapse
|
44
|
Desai A, Peters S. Immunotherapy-based combinations in metastatic NSCLC. Cancer Treat Rev 2023; 116:102545. [PMID: 37030062 DOI: 10.1016/j.ctrv.2023.102545] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023]
Abstract
Immuno-oncology has revolutionized the treatment of metastatic non-small cell lung cancer (mNSCLC) since the approval of immunotherapy by the U.S. FDA in 2015. Despite the advancements, outcomes for patients have room for further improvement. Combination therapies have shown promise in overcoming resistance and improving outcomes. This review focuses on current immunotherapy-based combination approaches, reported and ongoing trials, as well as novel combination strategies, challenges, and future directions for mNSCLC treatment. We summarize approaches in combination with chemotherapy, novel immune checkpoints, tyrosine kinase inhibitors and other strategies including vaccines, and radiation therapy. The promise of biomarker-driven studies to understand resistance and design multi-arm platform trials that evaluate novel therapies is becoming of increasing relevance with the ultimate goal of administering precision immunotherapy by identifying the right dose of the right combination for the right patient at the right time.
Collapse
|
45
|
Mills CM, Benton TZ, Piña I, Francis MJ, Reyes L, Dolloff NG, Peterson YK, Woster PM. Stimulation of natural killer cells with small molecule inhibitors of CD38 for the treatment of neuroblastoma. Chem Sci 2023; 14:2168-2182. [PMID: 36845935 PMCID: PMC9945084 DOI: 10.1039/d2sc05749b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/28/2023] [Indexed: 02/02/2023] Open
Abstract
High-risk neuroblastoma (NB) accounts for 15% of all pediatric cancer deaths. Refractory disease for high-risk NB patients is attributed to chemotherapy resistance and immunotherapy failure. The poor prognosis for high-risk NB patients demonstrates an unmet medical need for the development of new, more efficacious therapeutics. CD38 is an immunomodulating protein that is expressed constitutively on natural killer (NK) cells and other immune cells in the tumor microenvironment (TME). Furthermore, CD38 over expression is implicated in propagating an immunosuppressive milieu within the TME. Through virtual and physical screening, we have identified drug-like small molecule inhibitors of CD38 with low micromolar IC50 values. We have begun to explore structure activity relationships for CD38 inhibition through derivatization of our most effective hit molecule to develop a new compound with lead-like physicochemical properties and improved potency. We have demonstrated that our derivatized inhibitor, compound 2, elicits immunomodulatory effects in NK cells by increasing cell viability by 190 ± 36% in multiple donors and by significantly increasing interferon gamma. Additionally, we have illustrated that NK cells exhibited enhanced cytotoxicity toward NB cells (14% reduction of NB cells over 90 minutes) when given a combination treatment of our inhibitor and the immunocytokine ch14.18-IL2. Herein we describe the synthesis and biological evaluation of small molecule CD38 inhibitors and demonstrate their potential utility as a novel approach to NB immunotherapy. These compounds represent the first examples of small molecules that stimulate immune function for the treatment of cancer.
Collapse
Affiliation(s)
- Catherine M Mills
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Thomas Z Benton
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Ivett Piña
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Megan J Francis
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Leticia Reyes
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Nathan G Dolloff
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| | - Patrick M Woster
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina 70 President St Charleston SC 29425 USA
| |
Collapse
|
46
|
Abdallah AO, Mahmoudjafari Z, Ahmed N, Cui W, Shune L, McGuirk J, Mohan M, Mohyuddin GR, Afrough A, Alkharabsheh O, Atrash S. Clinical efficacy of retreatment of daratumumab-based therapy (D2) in daratumumab-refractory multiple myeloma. Eur J Haematol 2023; 110:626-632. [PMID: 36752328 DOI: 10.1111/ejh.13942] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023]
Abstract
Daratumumab demonstrates activity as a single agent and in combination with either immunomodulatory agents (IMiDs) or proteasome inhibitors (PIs) in relapsed refractory multiple myeloma (RRMM). However, little is known about the benefit of daratumumab retreatment in daratumumab-refractory MM. This study aimed to analyze the clinical efficacy of daratumumab-based retreatment (D2) in patients who are daratumumab refractory MM. Retrospectively, we identified 43 RRMM patients from a single-center database review. The median age was 65 years, 42% patients had high-risk cytogenetics, and 23% had an extramedullary disease, while the median time between D2 and prior daratumumab was 1 (0.25-39) month. All D2 patients received combination therapy with either pomalidomide, carfilzomib, bortezomib, or lenalidomide. The response rate, median progression-free, and overall survival were 49%, 7.97 and 32.6 months, respectively. Our study raises the possibility of re-utilizing daratumumab in combination with different classes of anti-myeloma drugs to generate responses in RRMM patients who are daratumumab-refractory.
Collapse
Affiliation(s)
- Al-Ola Abdallah
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas, USA.,US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
| | - Zahra Mahmoudjafari
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA.,University of Kansas Medical Center, Westwood, Kansas, USA
| | - Nausheen Ahmed
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas, USA.,US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
| | - Wei Cui
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA.,University of Kansas Medical Center, Westwood, Kansas, USA
| | - Leyla Shune
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas, USA.,US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA
| | - Joseph McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas, USA
| | - Meera Mohan
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA.,Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ghulam Rehman Mohyuddin
- Division of Hematology and Hematologic Malignancies, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Aimaz Afrough
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA.,Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Omar Alkharabsheh
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA.,Division of Hematology/Oncology, University of South Alabama, Mobile, Alabama, USA
| | - Shebli Atrash
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, Kansas, USA.,Division of Hematology, Levine Cancer Institute, Carolinas Healthcare System, Charlotte, North Carolina, USA
| |
Collapse
|
47
|
Carlo‐Stella C, Zinzani PL, Sureda A, Araújo L, Casasnovas O, Carpio C, Yeh S, Bouabdallah K, Cartron G, Kim WS, Cordoba R, Koh Y, Re A, Alves D, Chamuleau M, Le Gouill S, López‐Guillermo A, Moreira I, van der Poel MWM, Abbadessa G, Meng R, Ji R, Lépine L, Saleem R, Ribrag V. A phase 1/2, open-label, multicenter study of isatuximab in combination with cemiplimab in patients with lymphoma. Hematol Oncol 2023; 41:108-119. [PMID: 36251503 PMCID: PMC10092787 DOI: 10.1002/hon.3089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/30/2022] [Accepted: 10/11/2022] [Indexed: 02/03/2023]
Abstract
Patients with relapsed or refractory lymphoma have limited treatment options, requiring newer regimens. In this Phase 1/2 study (NCT03769181), we assessed the safety, efficacy, and pharmacokinetics of isatuximab (Isa, anti-CD38 antibody) in combination with cemiplimab (Cemi, anti-programmed death-1 [PD-1] receptor antibody; Isa + Cemi) in patients with classic Hodgkin lymphoma (cHL), diffuse large B-cell lymphoma (DLBCL), and peripheral T-cell lymphoma (PTCL). In Phase 1, we characterized the safety and tolerability of Isa + Cemi with planned dose de-escalation to determine the recommended Phase 2 dose (RP2D). Six patients in each cohort were treated with a starting dose of Isa + Cemi to determine the RP2D. In Phase 2, the primary endpoints were complete response in Cohort A1 (cHL anti-PD-1/programmed death-ligand 1 [PD-L1] naïve), and objective response rate in Cohorts A2 (cHL anti-PD-1/PD-L1 progressors), B (DLBCL), and C (PTCL). An interim analysis was performed when the first 18 (Cohort A1), 12 (Cohort A2), 17 (Cohort B), and 11 (Cohort C) patients in Phase 2 had been treated and followed up for 24 weeks. Isa + Cemi demonstrated a manageable safety profile with no new safety signals. No dose-limiting toxicities were observed at the starting dose; thus, the starting dose of each drug was confirmed as the RP2D. Based on the Lugano 2014 criteria, 55.6% (Cohort A1), 33.3% (Cohort A2), 5.9% (Cohort B), and 9.1% (Cohort C) of patients achieved a complete or partial response. Pharmacokinetic analyses suggested no effect of Cemi on Isa exposure. Modest clinical efficacy was observed in patients with cHL regardless of prior anti-PD-1/PD-L1 exposure. In DLBCL or PTCL cohorts, interim efficacy analysis results did not meet prespecified criteria to continue enrollment in Phase 2 Stage 2. Isa + Cemi did not have a synergistic effect in these patient populations.
Collapse
Affiliation(s)
- Carmelo Carlo‐Stella
- Department of Biomedical SciencesHumanitas University and Department of Oncology and HematologyIRCCS Humanitas Research HospitalMilanoItaly
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero‐Universitaria di Bologna Istituto di Ematologia “Seràgnoli” and Dipartimento di Medicina SpecialisticaDiagnostica e Sperimentale Università di BolognaBolognaItaly
| | - Anna Sureda
- Institut Català D'Oncologia ‐ Hospital Duran i ReynalsIDIBELLUniversitat de BarcelonaBarcelonaSpain
| | | | | | - Cecilia Carpio
- Department of HematologyVall d'Hebron Institute of Oncology (VHIO)Hospital Universitari Vall d’HebronVall d’Hebron Barcelona Hospital CampusUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Su‐Peng Yeh
- China Medical University HospitalTaichungTaiwan
| | - Krimo Bouabdallah
- Hematology and Cellular Therapy DepartmentUniversity Hospital of BordeauxBordeauxFrance
| | - Guillaume Cartron
- Department of HematologyCentre Hospitalier Universitaire MontpellierMontpellierFrance
| | - Won Seog Kim
- Sungkyunkwan University School of MedicineSamsung Medical CenterSeoulKorea
| | - Raul Cordoba
- Fundación Jiménez Díaz University HospitalMadridSpain
| | - Youngil Koh
- Department of Internal MedicineSeoul National University HospitalSeoulKorea
| | - Alessandro Re
- Hematology DivisionASST Spedali Civili BresciaBresciaItaly
| | - Daniela Alves
- Hematology and Bone Marrow Transplant DepartmentHospital de Santa MariaCentro Hospitalar Universitário Lisboa Norte (CHULN)LisbonPortugal
| | - Martine Chamuleau
- Department of HematologyCancer Center Amsterdamon behalf of the LLPC (Lunenburg Lymphoma Phase I/II Consortium)Amsterdam University Medical CenterVU University AmsterdamAmsterdamThe Netherlands
| | | | | | - Ilídia Moreira
- Department of Onco‐HematologyPortuguese Institute of Oncology of PortoPortoPortugal
| | - Marjolein W. M. van der Poel
- Department of Internal MedicineDivision of HematologyGROW School for Oncology and Developmental Biologyon behalf of the LLPC (Lunenburg Lymphoma Phase I/II Consortium)Maastricht University Medical CenterMaastrichtthe Netherlands
| | | | | | - Ran Ji
- SanofiCambridgeMassachusettsUSA
| | | | | | - Vincent Ribrag
- Département d’Hématologie et Département des Essais Précoces (DITEP)Institut Gustave RoussyVillejuifFrance
| |
Collapse
|
48
|
Richter J, Lin PL, Garcia-Horton V, Guyot P, Singh E, Zhou ZY, Sievert M, Taiji R. Matching-adjusted indirect comparison of isatuximab plus carfilzomib and dexamethasone with daratumumab plus lenalidomide and dexamethasone in relapsed multiple myeloma. Cancer Med 2023; 12:8005-8017. [PMID: 36726287 PMCID: PMC10134287 DOI: 10.1002/cam4.5584] [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: 10/27/2022] [Revised: 11/28/2022] [Accepted: 12/17/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGOUND Lenalidomide-based regimens are commonly used for early relapse in patients with relapsed and/or refractory multiple myeloma (RRMM) receiving at least one prior line of therapy. In the absence of head-to-head comparison, matching-adjusted indirect comparison (MAIC) was conducted to demonstrate efficacy and safety of isatuximab+carfilzomib+dexamethasone (Isa-Kd) versus daratumumab + lenalidomide + dexamethasone (Dara-Rd) in RRMM. METHODS Patient-level data from IKEMA trial (Isa-Kd, n = 179) were matched to aggregate data from POLLUX (Dara-Rd, n = 286). Hazard ratios (HR) and 95% confidence intervals (CI) for progression-free survival (PFS) and overall survival (OS) were generated by weighted Cox proportional hazard models. Odds ratios (OR), 95% CI, and p-value were calculated for ≥very good partial response (≥VGPR) and treatment-emergent adverse events (TEAEs). RESULTS After matching, no significant differences were observed between Isa-Kd and Dara-Rd in baseline characteristics except for patients with >3 prior lines (0.0% vs. 4.9%). Isa-Kd showed significantly better PFS (HR [95% CI]: 0.46 [0.24-0.86]; p = 0.0155), statistically non-significant improvement favoring Isa-Kd in OS (0.47 [0.20-1.09]; 0.0798), and ≥VGPR (OR [95% CI]: 1.53 [0.89-2.64]; p = 0.1252) than Dara-Rd. Odds of occurrence were significantly lower for some all-grade and grade 3/4 TEAEs with Isa-Kd than Dara-Rd. CONCLUSION These results support Isa-Kd as an efficacious treatment for early relapse in non-lenalidomide refractory patients.
Collapse
Affiliation(s)
- Joshua Richter
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Zheng T, Chen P, Xu Y, Jia P, Li Y, Li Y, Cao J, Li W, Zhen Y, Zhang Y, Zhang S, Du J, Zhang J. Comprehensive analysis of thirteen-gene panel with prognosis value in Multiple Myeloma. Cancer Biomark 2023; 38:583-593. [PMID: 37980648 DOI: 10.3233/cbm-230115] [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] [Indexed: 11/21/2023]
Abstract
BACKGROUND Although there are many treatments for Multiple myeloma (MM), patients with MM still unable to escape the recurrence and aggravation of the disease. OBJECTIVE We constructed a risk model based on genes closely associated with MM prognosis to predict its prognostic value. METHODS Gene function enrichment and signal pathway enrichment analysis, Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis, univariate and multivariate Cox regression analysis, Kaplan-Meier (KM) survival analysis and Receiver Operating Characteristic (ROC) analysis were used to identify the prognostic gene signature for MM. Finally, the prognostic gene signature was validated using the Gene Expression Omnibus (GEO) database. RESULTS Thirteen prognostic genes were screened by univariate Cox analysis and LASSO regression analysis. Multivariate Cox analysis revealed risk score to be an independent prognostic factor for patients with MM [Hazard Ratio (HR) = 2.564, 95% Confidence Interval (CI) = 2.223-2.958, P< 0.001]. The risk score had a high level of predictive value according to ROC analysis, with an area under the curve (AUC) of 0.744. CONCLUSIONS The potential prognostic signature of thirteen genes were assessed and a risk model was constructed that significantly correlated with prognosis in MM patients.
Collapse
Affiliation(s)
- Tingting Zheng
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Panpan Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanlin Xu
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Peijun Jia
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yating Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiaming Cao
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Wanxin Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yazhe Zhen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shijie Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiangfeng Du
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jingxin Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
50
|
Zhang G, Guo C, Wang Y, Zhang X, Liu S, Qu W, Chen C, Yan L, Yang Z, Zhang Z, Jiang X, Chen X, Liu H, Lai Q, Wei X, Lu Y, Zhao S, Deng H, Wang Y, Yu L, Yu H, Wu Y, Su Z, Chen P, Ren Z, Yu M, Qu F, Luo Y, Gou L, Li Q, Huang Y, Ma F, Yang J. FTL004, an anti-CD38 mAb with negligible RBC binding and enhanced pro-apoptotic activity, is a novel candidate for treatments of multiple myeloma and non-Hodgkin lymphoma. J Hematol Oncol 2022; 15:177. [PMID: 36581954 PMCID: PMC9798557 DOI: 10.1186/s13045-022-01395-0] [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: 10/24/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
Anti-CD38 monoclonal antibodies (mAbs), daratumumab, and isatuximab have represented a breakthrough in the treatment of multiple myeloma (MM). Recently, CD38-based mAbs were expected to achieve increasing potential beyond MM, which encouraged us to develop new anti-CD38 mAbs to meet clinical needs. In this study, we developed a novel humanized anti-CD38 antibody, FTL004, which exhibited enhanced pro-apoptotic ability and negligible binding to red blood cells (RBCs). FTL004 presented a better ability to induce direct apoptosis independent of Fc-mediated cross-linking against lymphoma and MM cell lines as well as primary myeloma cells derived from MM patients. For instance, FTL004 induced RPMI 8226 cells with 55% early apoptosis cells compared with 20% in the isatuximab-treated group. Of interest, FTL004 showed ignorable binding to CD38 on human RBCs in contrast to tumor cells, even at concentrations up to 30 μg/mL. Furthermore, with an engineered Fc domain, FTL004 displayed stronger antibody-dependent cellular cytotoxicity (ADCC) against CD38+ malignant cells. In vivo MM and non-Hodgkin lymphoma tumor xenograft models showed that FTL004 possessed an effective anti-tumor effect. Cryo-electron microscopy structure resolved two epitope centers of FTL004 on CD38: one of which was unique while the other partly overlapped with that of isatuximab. Taken together, FTL004 distinguishes it from other CD38 targeting mAbs and represents a potential candidate for the treatment of MM and non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Guangbing Zhang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Cuiyu Guo
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Yan Wang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Xianda Zhang
- Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China
| | - Shuang Liu
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China ,Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China
| | - Wen Qu
- grid.490255.f0000 0004 7594 4364Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, People’s Republic of China
| | - Chunxia Chen
- grid.13291.380000 0001 0807 1581Department of Transfusion, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Lingli Yan
- grid.13291.380000 0001 0807 1581Department of Transfusion, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Zhouning Yang
- Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China
| | - Zhixiong Zhang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Xiaohua Jiang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Xiaofeng Chen
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Hong Liu
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Qinhuai Lai
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Xian Wei
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Ying Lu
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Shengyan Zhao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Han Deng
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Yuxi Wang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Lin Yu
- grid.490255.f0000 0004 7594 4364Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, People’s Republic of China
| | - Hongbin Yu
- grid.13291.380000 0001 0807 1581Department of Hematology, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yu Wu
- grid.13291.380000 0001 0807 1581Department of Hematology, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Zhaoming Su
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Pengyu Chen
- Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China
| | - Ziqing Ren
- Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China
| | - Meng Yu
- Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China
| | - Feng Qu
- Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China
| | - Yong Luo
- grid.13291.380000 0001 0807 1581Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Lantu Gou
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China
| | - Qing Li
- Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China ,grid.13291.380000 0001 0807 1581West China School of Public Health, Sichuan University, Chengdu, People’s Republic of China
| | - Ying Huang
- Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China
| | - Fanxin Ma
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China ,Sound Biopharmaceuticals Co., Ltd., Tianfu International Bio-Town, Huigu Dong 2nd Road 8, Chengdu, Sichuan 610200 People’s Republic of China
| | - Jinliang Yang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, 3-17 People Road, Chengdu, Sichuan 610041 People’s Republic of China ,grid.506261.60000 0001 0706 7839Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Chengdu, People’s Republic of China
| |
Collapse
|