1
|
Pirosa MC, Stathis A, Zucca E. Tafasitamab for the treatment of patients with diffuse large B-cell lymphoma. Hum Vaccin Immunother 2024; 20:2309701. [PMID: 38299612 PMCID: PMC10841029 DOI: 10.1080/21645515.2024.2309701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/21/2024] [Indexed: 02/02/2024] Open
Abstract
Patients with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) require additional treatments, especially those not eligible or not responding to high dose cytotoxic chemotherapy and stem cell transplantation. Over the last few years, several new treatments have been developed and approved for these patients, among them of particular relevance are those targeting CD19. Tafasitamab is a humanized monoclonal antibody targeting CD19, composed of a modified fragment crystallizable (Fc) region engineered with higher affinity for Fc gamma receptors (FcγR) receptors, leading to increased cytotoxicity through natural killer cells and macrophages (antibody-dependent cellular cytotoxicity and antibody-dependent cell-mediated phagocytosis). In this product review, we will discuss its mechanism of action, safety profile and efficacy results from clinical trials that led to its approval in combination with lenalidomide for patients with R/R DLBCL ineligible for high-dose chemotherapy and autologous transplantation.
Collapse
Affiliation(s)
- Maria Cristina Pirosa
- Clinic of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Science, Universita’ della Svizzera italiana, Lugano, Switzerland
| | - Anastasios Stathis
- Clinic of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Faculty of Biomedical Science, Universita’ della Svizzera italiana, Lugano, Switzerland
| | - Emanuele Zucca
- Clinic of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Science, Universita’ della Svizzera italiana, Lugano, Switzerland
| |
Collapse
|
2
|
Camus V, Houot R, Brisou G, Tessoulin B, Bailly S, Sesques P, Decroocq J, Krzisch D, Oberic L, Lemonnier F, Bouabdallah K, Campidelli A, Tounes L, Abraham J, Herbaux C, Morschhauser F, Damaj GL, Guidez S, Carras S, Fornecker LM, Choquet S, Hermine O, Paillassa J, Chauchet A, Casasnovas O, Drieu La Rochelle L, Castilla-Llorente C, Joris M, Dupont V, Marquet A, Le Gouill S, Jardin F. Outcome of patients with large B-cell lymphoma treated with tafasitamab plus lenalidomide either before or after CAR T-cell therapy. Blood Adv 2024; 8:5371-5381. [PMID: 39163620 DOI: 10.1182/bloodadvances.2024013726] [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: 05/28/2024] [Revised: 07/16/2024] [Accepted: 07/31/2024] [Indexed: 08/22/2024] Open
Abstract
ABSTRACT Tafasitamab plus lenalidomide (TAFA-LEN) treatment relevance pre- or post-anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is debated. We analyzed patients with large B-cell lymphoma in the DESCAR-T registry treated with axi[1]cel or tisa-cel in ≥3rd line and TAFA-LEN before (n = 15, "TL-pre-CAR-T" set) or directly after (n = 52, "TL-post-CAR-T" set) CAR T-cell therapy. We compared TAFA-LEN v. other treatments using inverse probability weighting in the TL-post-CAR[1]T set. In the TL-post-CAR-T set, the median progression-free survival (mPFS), overall survival (mOS), and duration of response (mDOR) since the first treatment for progression (mPFS2/mOS2/mDOR2) were 3, 4.7, and 8.1 months, respectively. The best overall response rate (bORR) and best complete response rate (bCRR) after TAFA-LEN were 13.5% and 7.7%, respectively. Outcomes were better for patients who relapsed >6 months after CAR T-cell therapy (mPFS2: 5.6 vs 2 months, P = .0138; mOS2: not reached vs 3.8 months, P = .0034). The bORR and bCRR between TAFA-LEN and other treatments were 20.6% vs 24.9% and 11.6% vs 15.6%, respectively. Outcomes were similar between TAFA-LEN and other treatments (mPFS2: 2.9 vs 2.4 months, P = .91; mOS2: 3.3 vs 5.5 months, P = .06). In an exploratory analysis of the TL-pre-CAR-T set, the median TAFA-LEN treatment duration before CAR-T was 3.7 months with no patient becoming CD19 negative. The bORR, bCRR, 6- month PFS, and OS rates after CAR T-cell infusion were 45.5%, 36.4%, 20.1%, and 58.2%, respectively. Neither TAFA-LEN nor comparative salvage treatment improved outcomes for patients relapsing after CAR T-cell therapy.
Collapse
Affiliation(s)
- Vincent Camus
- Department of Hematology, Centre Henri Becquerel, Rouen, France
| | - Roch Houot
- Department of Hematology, Centre Hospitalier Universitaire Rennes, University of Rennes, INSERM U1236, Etablissement Français du Sang, Rennes, France
| | - Gabriel Brisou
- Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Benoit Tessoulin
- Department of Hematology, Nantes University Hospital, Nantes, France
| | - Sébastien Bailly
- Department of Hematology, Centre Hospitalier Universitaire Estaing, Clermont-Ferrand, France
| | - Pierre Sesques
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Pierre-Benite, France
| | - Justine Decroocq
- Department of Hematology, Cochin University Hospital, Paris, France
| | - Daphné Krzisch
- Department of Hemato-oncology, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Université de Paris, Paris, France
| | - Lucie Oberic
- Department of Hematology, Institut Universitaire du Cancer, Toulouse-Oncopole, Toulouse, France
| | - François Lemonnier
- Department of Hematology, Henri Mondor University Hospital, Créteil, France
| | - Krimo Bouabdallah
- Department of Hematology and Cellular Therapy, Bordeaux University Hospital, Bordeaux, France
| | - Arnaud Campidelli
- Department of Hematology, Hôpital de Brabois, Nancy University Hospital, Nancy, France
| | - Ledraa Tounes
- Department of Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Antoine, Université de Paris, Paris, France
| | - Julie Abraham
- Department of Hematology, Dupuytren Hospital, Limoges University Hospital, Limoges, France
| | - Charles Herbaux
- Department of Hematology, Montpellier University Hospital, Montpellier, France
| | - Franck Morschhauser
- Department of Hematology, Claude Huriez Hospital, Lille University Hospital, Lille, France
| | | | - Stéphanie Guidez
- Department of Hematology, Poitiers University Hospital, Poitiers, France
| | - Sylvain Carras
- Department of Hematology, Albert Michallon University Hospital, Grenoble, France
| | | | - Sylvain Choquet
- Department of Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Université de Paris, Paris, France
| | - Olivier Hermine
- Department of Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Université de Paris, Paris, France
| | - Jérome Paillassa
- Department of Hematology, Angers University Hospital, Angers, France
| | - Adrien Chauchet
- Department of Hematology, Besançon University Hospital, Besançon, France
| | | | | | | | - Magalie Joris
- Department of Hematology, Amiens University Hospital, Amiens, France
| | - Vivien Dupont
- Lymphoma Academic Research Organisation, Lyon-Sud Hospital, Pierre-Bénite, France
| | - Alexandra Marquet
- Lymphoma Academic Research Organisation, Lyon-Sud Hospital, Pierre-Bénite, France
| | | | - Fabrice Jardin
- Department of Hematology, Centre Henri Becquerel, Rouen, France
| |
Collapse
|
3
|
Toledo-Stuardo K, Ribeiro CH, González-Herrera F, Matthies DJ, Le Roy MS, Dietz-Vargas C, Latorre Y, Campos I, Guerra Y, Tello S, Vásquez-Sáez V, Novoa P, Fehring N, González M, Rodríguez-Siza J, Vásquez G, Méndez P, Altamirano C, Molina MC. Therapeutic antibodies in oncology: an immunopharmacological overview. Cancer Immunol Immunother 2024; 73:242. [PMID: 39358613 PMCID: PMC11448508 DOI: 10.1007/s00262-024-03814-2] [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: 06/22/2024] [Accepted: 08/16/2024] [Indexed: 10/04/2024]
Abstract
The biotechnological development of monoclonal antibodies and their immunotherapeutic use in oncology have grown exponentially in the last decade, becoming the first-line therapy for some types of cancer. Their mechanism of action is based on the ability to regulate the immune system or by interacting with targets that are either overexpressed in tumor cells, released into the extracellular milieu or involved in processes that favor tumor growth. In addition, the intrinsic characteristics of each subclass of antibodies provide specific effector functions against the tumor by activating antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent cellular phagocytosis, among other mechanisms. The rational design and engineering of monoclonal antibodies have improved their pharmacokinetic and pharmacodynamic features, thus optimizing the therapeutic regimens administered to cancer patients and improving their clinical outcomes. The selection of the immunoglobulin G subclass, modifications to its crystallizable region (Fc), and conjugation of radioactive substances or antineoplastic drugs may all improve the antitumor effects of therapeutic antibodies. This review aims to provide insights into the immunological and pharmacological aspects of therapeutic antibodies used in oncology, with a rational approach at molecular modifications that can be introduced into these biological tools, improving their efficacy in the treatment of cancer.
Collapse
Affiliation(s)
- Karen Toledo-Stuardo
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Carolina H Ribeiro
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Fabiola González-Herrera
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Douglas J Matthies
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - María Soledad Le Roy
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Claudio Dietz-Vargas
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Yesenia Latorre
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Ivo Campos
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Yuneisy Guerra
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Samantha Tello
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Valeria Vásquez-Sáez
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Pedro Novoa
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Nicolás Fehring
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Mauricio González
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Jose Rodríguez-Siza
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Gonzalo Vásquez
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Pamela Méndez
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- Centro Regional de Estudio en Alimentos Saludables, Valparaíso, Chile
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Santiago, Chile
| | - María Carmen Molina
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile.
| |
Collapse
|
4
|
Saha A, Chavez JC. Recent developments in CD19-targeted therapies for follicular lymphoma. Expert Opin Biol Ther 2024; 24:1049-1055. [PMID: 39291554 DOI: 10.1080/14712598.2024.2404100] [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: 06/10/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION CD19 has emerged as an important and novel therapeutic target in follicular lymphoma. CD19-directed therapies, including monoclonal antibodies, bispecific antibodies, and CAR T-cell therapies, offer promising avenues for treating follicular lymphoma and improving outcomes. AREAS COVERED We review the role and rationale of targeting CD19 in follicular lymphoma and different interventions of CD19 targeting, such as cell therapy, bispecific antibodies, antibody-drug conjugates, and monoclonal antibodies. We finalize with a discussion on how these therapies may influence the treatment landscape of follicular lymphoma. EXPERT OPINION CD19 is an attractive target for therapeutic development in follicular lymphoma. Given its effectiveness, it will continue to move forward as a promising therapy for this disease.
Collapse
Affiliation(s)
- Aditi Saha
- Department of Medicine/Hematology Oncology, University of South Florida, Tampa, FL, USA
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
5
|
Duell J, Westin J. The future of immunotherapy for diffuse large B-cell lymphoma. Int J Cancer 2024. [PMID: 39319495 DOI: 10.1002/ijc.35156] [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: 08/22/2023] [Revised: 06/21/2024] [Accepted: 07/15/2024] [Indexed: 09/26/2024]
Abstract
With the introduction of anti-CD19 chimeric antigen receptor (CAR) T-cell (CAR T) therapies, bispecific CD3/CD20 antibodies and anti-CD19 antibodies, immunotherapy continues to transform the treatment of diffuse large B-cell lymphoma (DLBCL). A number of novel immunotherapeutic strategies are under investigation to build upon current clinical benefit and offer further options to those patients who cannot tolerate conventional intensive therapies due to their age and/or state of health. Alongside immunotherapies that leverage the adaptive immune response, natural killer (NK) cell and myeloid cell-engaging therapies can utilize the innate immune system. Monoclonal antibodies engineered for greater recognition by the patient's immune system can enhance antitumor cytotoxic mechanisms mediated by NK cells and macrophages. In addition, CAR technology is extending into NK cells and macrophages and investigational immune checkpoint inhibitors targeting macrophage/myeloid cell checkpoints via the CD47/SIRPα axis are in development. Regimens that engage both innate and adaptive immune responses may help to overcome resistance to current immunotherapies. Furthermore, combinations of immunotherapy and oncogenic pathway inhibitors to reprogram the immunosuppressive tumor microenvironment of DLBCL may also potentiate antitumor responses. As immunotherapy treatment options continue to expand, both in the first-line setting and further lines of therapy, understanding how to harness these immunotherapies and the potential for combination approaches will be important for the development of future DLBCL treatment approaches.
Collapse
Affiliation(s)
- Johannes Duell
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Jason Westin
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
6
|
Zekri L, Hagelstein I, Märklin M, Klimovich B, Christie M, Lindner C, Kämereit S, Prakash N, Müller S, Stotz S, Maurer A, Greve C, Schmied B, Atar D, Rammensee HG, Jung G, Salih HR. Immunocytokines with target cell-restricted IL-15 activity for treatment of B cell malignancies. Sci Transl Med 2024; 16:eadh1988. [PMID: 38446900 DOI: 10.1126/scitranslmed.adh1988] [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: 02/16/2023] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
Despite the advances in cancer treatment achieved, for example, by the CD20 antibody rituximab, an urgent medical need remains to optimize the capacity of such antibodies to induce antibody-dependent cellular cytotoxicity (ADCC) that determines therapeutic efficacy. The cytokine IL-15 stimulates proliferation, activation, and cytolytic capacity of NK cells, but broad clinical use is prevented by short half-life, poor accumulation at the tumor site, and severe toxicity due to unspecific immune activation. We here report modified immunocytokines consisting of Fc-optimized CD19 and CD20 antibodies fused to an IL-15 moiety comprising an L45E-E46K double mutation (MIC+ format). The E46K mutation abrogated binding to IL-15Rα, thereby enabling substitution of physiological trans-presentation by target binding and thus conditional IL-15Rβγ stimulation, whereas the L45E mutation optimized IL-15Rβγ agonism and producibility. In vitro analysis of NK activation, anti-leukemia reactivity, and toxicity using autologous and allogeneic B cells confirmed target-dependent function of MIC+ constructs. Compared with Fc-optimized CD19 and CD20 antibodies, MIC+ constructs mediated superior target cell killing and NK cell proliferation. Mouse models using luciferase-expressing human NALM-6 lymphoma cells, patient acute lymphoblastic leukemia (ALL) cells, and murine EL-4 lymphoma cells transduced with human CD19/CD20 as targets and human and murine NK cells as effectors, respectively, confirmed superior and target-dependent anti-leukemic activity. In summary, MIC+ constructs combine the benefits of Fc-optimized antibodies and IL-15 cytokine activity and mediate superior NK cell immunity with potentially reduced side effects. They thus constitute a promising new immunotherapeutic approach shown here for B cell malignancies.
Collapse
Affiliation(s)
- Latifa Zekri
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
- Department of Immunology, Institute for Cell Biology, Eberhard Karls Universität Tübingen, Germany
- DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), 72076 Tübingen, Germany
| | - Ilona Hagelstein
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
| | - Melanie Märklin
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
| | - Boris Klimovich
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
| | - Mary Christie
- School of Medical Sciences, University of Sydney, 2050 NSW, Australia
| | - Cornelia Lindner
- Department of Immunology, Institute for Cell Biology, Eberhard Karls Universität Tübingen, Germany
- DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), 72076 Tübingen, Germany
| | - Sofie Kämereit
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Department of Immunology, Institute for Cell Biology, Eberhard Karls Universität Tübingen, Germany
- DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), 72076 Tübingen, Germany
| | - Nisha Prakash
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
- Department of Immunology, Institute for Cell Biology, Eberhard Karls Universität Tübingen, Germany
- DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), 72076 Tübingen, Germany
| | - Stefanie Müller
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
| | - Sophie Stotz
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
- Department for Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Andreas Maurer
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
- Department for Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Carsten Greve
- Department of Immunology, Institute for Cell Biology, Eberhard Karls Universität Tübingen, Germany
- DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), 72076 Tübingen, Germany
| | - Bastian Schmied
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
| | - Daniel Atar
- Childrens University Hospital, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Hans-Georg Rammensee
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
- Department of Immunology, Institute for Cell Biology, Eberhard Karls Universität Tübingen, Germany
- DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), 72076 Tübingen, Germany
| | - Gundram Jung
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
- Department of Immunology, Institute for Cell Biology, Eberhard Karls Universität Tübingen, Germany
- DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), 72076 Tübingen, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
7
|
Duell J, Abrisqueta P, Andre M, Gaidano G, Gonzales-Barca E, Jurczak W, Kalakonda N, Liberati AM, Maddocks KJ, Menne T, Nagy Z, Tournilhac O, Kuffer C, Bakuli A, Amin A, Gurbanov K, Salles G. Tafasitamab for patients with relapsed or refractory diffuse large B-cell lymphoma: final 5-year efficacy and safety findings in the phase II L-MIND study. Haematologica 2024; 109:553-566. [PMID: 37646664 PMCID: PMC10828760 DOI: 10.3324/haematol.2023.283480] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
Tafasitamab, an anti-CD19 immunotherapy, is used with lenalidomide for patients with autologous stem cell transplant-ineligible relapsed/refractory diffuse large B-cell lymphoma based on the results of the phase II L-MIND study (NCT02399085). We report the final 5-year analysis of this study. Eighty patients ≥18 years who had received one to three prior systemic therapies, and had Eastern Cooperative Oncology Group performance status 0-2 received up to 12 cycles of co-administered tafasitamab and lenalidomide, followed by tafasitamab monotherapy until disease progression or unacceptable toxicity. The primary endpoint was the best objective response rate. Secondary endpoints included duration of response, progression-free survival, overall survival, and safety. Exploratory analyses evaluated efficacy endpoints by prior lines of therapy. At data cutoff on November 14, 2022, the objective response rate was 57.5%, with a complete response rate of 41.3% (n=33), which was consistent with prior analyses. With a median follow-up of 44.0 months, the median duration of response was not reached. The median progression-free survival was 11.6 months (95% confidence interval [95% CI]: 5.7-45.7) with a median follow-up of 45.6 months. The median overall survival was 33.5 months (95% CI: 18.3-not reached) with a median follow-up of 65.6 months. Patients who had received one prior line of therapy (n=40) showed a higher objective response rate (67.5%; 52.5% complete responses) compared to patients who had received two or more prior lines of therapy (n=40; 47.5%; 30% complete responses), but the median duration of response was not reached in either subgroup. Other exploratory analyses revealed consistent long-term efficacy results across subgroups. Adverse events were consistent with those described in previous reports, were manageable, and their frequency decreased during tafasitamab monotherapy, with no new safety concerns. This final 5-year analysis of L-MIND demonstrates that the immunotherapy combination of tafasitamab and lenalidomide is well tolerated and has long-term clinical benefit with durable responses.
Collapse
Affiliation(s)
- Johannes Duell
- Medizinische Klinik und Poliklinik II, Universitätsklinik Würzburg, Würzburg.
| | - Pau Abrisqueta
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona
| | - Marc Andre
- Centre Hospitalier Universitaire CHU UCL Namur, Belgium
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine University of Eastern Piedmont and Ospedale Maggiore della Carità, Novara
| | - Eva Gonzales-Barca
- Department of Hematology, Institut Català d'Oncologia, Hospitalet de Llobregat, IDIBELL, Univeristat de Barcelona, Barcelona
| | - Wojciech Jurczak
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków
| | - Nagesh Kalakonda
- Department of Molecular and Clinical Cancer University of Liverpool, Liverpool, United Kingdom
| | - Anna Marina Liberati
- Università degli Studi di Perugia, Azienda Ospedaliera Santa Maria di Terni, Terni
| | - Kami J Maddocks
- Department of Internal Medicine, Arthur G James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, OH
| | - Tobias Menne
- Freeman Hospital, The Newcastle upon Tyne Hospitals, Newcastle upon Tyne, United Kingdom
| | - Zsolt Nagy
- Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | - Gilles Salles
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
8
|
Sakemura RL, Manriquez Roman C, Horvei P, Siegler EL, Girsch JH, Sirpilla OL, Stewart CM, Yun K, Can I, Ogbodo EJ, Adada MM, Bezerra ED, Kankeu Fonkoua LA, Hefazi M, Ruff MW, Kimball BL, Mai LK, Huynh TN, Nevala WK, Ilieva K, Augsberger C, Patra-Kneuer M, Schanzer J, Endell J, Heitmüller C, Steidl S, Parikh SA, Ding W, Kay NE, Nowakowski GS, Kenderian SS. CD19 occupancy with tafasitamab increases therapeutic index of CART19 cell therapy and diminishes severity of CRS. Blood 2024; 143:258-271. [PMID: 37879074 PMCID: PMC10808250 DOI: 10.1182/blood.2022018905] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/27/2023] Open
Abstract
ABSTRACT In the development of various strategies of anti-CD19 immunotherapy for the treatment of B-cell malignancies, it remains unclear whether CD19 monoclonal antibody therapy impairs subsequent CD19-targeted chimeric antigen receptor T-cell (CART19) therapy. We evaluated the potential interference between the CD19-targeting monoclonal antibody tafasitamab and CART19 treatment in preclinical models. Concomitant treatment with tafasitamab and CART19 showed major CD19 binding competition, which led to CART19 functional impairment. However, when CD19+ cell lines were pretreated with tafasitamab overnight and the unbound antibody was subsequently removed from the culture, CART19 function was not affected. In preclinical in vivo models, tafasitamab pretreatment demonstrated reduced incidence and severity of cytokine release syndrome and exhibited superior antitumor effects and overall survival compared with CART19 alone. This was associated with transient CD19 occupancy with tafasitamab, which in turn resulted in the inhibition of CART19 overactivation, leading to diminished CAR T apoptosis and pyroptosis of tumor cells.
Collapse
Affiliation(s)
- R. Leo Sakemura
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Claudia Manriquez Roman
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN
| | - Paulina Horvei
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Pediatric Bone Marrow Transplant and Cellular Therapy, UPMC Children’s Hospital of Pittsburgh, PA
| | - Elizabeth L. Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - James H. Girsch
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN
| | - Olivia L. Sirpilla
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Carli M. Stewart
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Kun Yun
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN
| | - Ismail Can
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Ekene J. Ogbodo
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Mohamad M. Adada
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | | | - Mehrdad Hefazi
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Michael W. Ruff
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Department of Neurology, Mayo Clinic, Rochester, MN
| | - Brooke L. Kimball
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Long K. Mai
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Truc N. Huynh
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | | | | | | - Wei Ding
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Neil E. Kay
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - Saad S. Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN
- Department of Immunology, Mayo Clinic, Rochester, MN
| |
Collapse
|
9
|
Damelang T, Brinkhaus M, van Osch TLJ, Schuurman J, Labrijn AF, Rispens T, Vidarsson G. Impact of structural modifications of IgG antibodies on effector functions. Front Immunol 2024; 14:1304365. [PMID: 38259472 PMCID: PMC10800522 DOI: 10.3389/fimmu.2023.1304365] [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: 09/29/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Immunoglobulin G (IgG) antibodies are a critical component of the adaptive immune system, binding to and neutralizing pathogens and other foreign substances. Recent advances in molecular antibody biology and structural protein engineering enabled the modification of IgG antibodies to enhance their therapeutic potential. This review summarizes recent progress in both natural and engineered structural modifications of IgG antibodies, including allotypic variation, glycosylation, Fc engineering, and Fc gamma receptor binding optimization. We discuss the functional consequences of these modifications to highlight their potential for therapeutical applications.
Collapse
Affiliation(s)
- Timon Damelang
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Sanquin Research, Department of Immunopathology, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Maximilian Brinkhaus
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Thijs L. J. van Osch
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Janine Schuurman
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Aran F. Labrijn
- Department of Antibody Research & Technologies’, Genmab, Utrecht, Netherlands
| | - Theo Rispens
- Sanquin Research, Department of Immunopathology, Amsterdam, Netherlands
| | - Gestur Vidarsson
- Sanquin Research, Department of Experimental Immunohematology and Landsteiner Laboratory, Amsterdam, Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
10
|
Brooks TR, Caimi PF. A paradox of choice: Sequencing therapy in relapsed/refractory diffuse large B-cell lymphoma. Blood Rev 2024; 63:101140. [PMID: 37949705 DOI: 10.1016/j.blre.2023.101140] [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: 04/18/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023]
Abstract
The available treatments for relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) have experienced a dramatic change since 2017. Incremental advances in basic and translational science over several decades have led to innovations in immune-oncology. These innovations have culminated in eight separate approvals by the US Food and Drug Administration for the treatment of patients with R/R DLBCL over the last 10 years. High-dose therapy and autologous stem cell transplant (HDT-ASCT) remains the standard of care for transplant-eligible patients who relapse after an initial remission. For transplant-ineligible patients or for those who relapse following HDT-ASCT, multiple options exist. Monoclonal antibodies targeting CD19, antibody-drug conjugates, bispecific antibodies, immune effector cell products, and other agents with novel mechanisms of action are now available for patients with R/R DLBCL. There is increasing use of chimeric antigen receptor (CAR) T-cells as second-line therapy for patients with early relapse of DLBCL or those who are refractory to initial chemoimmunotherapy. The clinical benefits of these strategies vary and are influenced by patient and disease characteristics, as well as the type of prior therapy administered. Therefore, there are multiple clinical scenarios that clinicians might encounter when treating R/R DLBCL. An optimal sequence of drugs has not been established, and there is no evidence-based consensus on how to best order these agents. This abundance of choices introduces a paradox: proliferating treatment options are initially a boon to patients and providers, but as choices grow further they no longer liberate. Rather, more choices make the management of R/R DLBCL more challenging due to lack of direct comparisons among agents and a desire to maximize patient outcomes. Here, we provide a review of recently-approved second- and subsequent-line agents, summarize real-world data detailing the use of these medicines, and provide a framework for sequencing therapy in R/R DLBCL.
Collapse
Affiliation(s)
- Taylor R Brooks
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Center, Cleveland, OH, United States of America
| | - Paolo F Caimi
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Center, Cleveland, OH, United States of America; Case Comprehensive Cancer Center, Cleveland, OH, United States of America.
| |
Collapse
|
11
|
García-Sancho AM, Cabero A, Gutiérrez NC. Treatment of Relapsed or Refractory Diffuse Large B-Cell Lymphoma: New Approved Options. J Clin Med 2023; 13:70. [PMID: 38202077 PMCID: PMC10779497 DOI: 10.3390/jcm13010070] [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: 10/30/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Overall, around 40% of patients with diffuse large B-cell lymphoma (DLBCL) have refractory disease or relapse after the first line of treatment. Until relatively recently, the prognosis of patients with relapsed or refractory DLBCL was very poor and treatment options were very limited. In recent years, several novel therapies have been approved that provide more effective options than conventional chemotherapy and that have manageable toxicity profiles. CAR-T cell therapy has become the new standard treatment for patients with refractory or early relapsed DLBCL, based on the positive results of the phase 3 ZUMA-7 and TRANSFORM clinical trials. This review addresses the role of CAR-T therapy and autologous stem cell transplantation in the treatment of these patients and other approved options for patients who are not candidates for transplant, such as the combinations of polatuzumab vedotin with bendamustine and rituximab, and tafasitamab with lenalidomide.
Collapse
Affiliation(s)
- Alejandro Martín García-Sancho
- Hematology Department, University Hospital of Salamanca, IBSAL (Instituto de Investigación Biomédica de Salamanca), CIBERONC (Centro de Investigación Biomédica en Red en Cáncer ), University of Salamanca, 37007 Salamanca, Spain; (A.C.); (N.C.G.)
| | | | | |
Collapse
|
12
|
Ilieva K, Eberl M, Jaehrling J, Blair D, Patra-Kneuer M, Boxhammer R, Alvarez Arias D, Heitmüller C. Preclinical study of CD19 detection methods post tafasitamab treatment. Front Immunol 2023; 14:1274556. [PMID: 37928552 PMCID: PMC10622958 DOI: 10.3389/fimmu.2023.1274556] [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: 08/08/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction Several CD19 targeted antibody-based therapeutics are currently available for patients with diffuse large B-cell lymphoma (DLBCL), including the Fc-modified antibody immunotherapy tafasitamab. This therapeutic landscape warrants the evaluation of potential sequencing approaches. Prior to a subsequent CD19-targeted therapy, CD19 expression on tafasitamab-treated patient biopsy samples may be assessed. However, no standardized methods for its detection are currently available. In this context, selecting a tafasitamab-competing CD19 detection antibody for immunohistochemistry (IHC) or flow cytometry (FC) may lead to misinterpreting epitope masking by tafasitamab as antigen loss or downregulation. Methods We analyzed a comprehensive panel of commercially available CD19 detection antibody clones for IHC and FC using competition assays on tafasitamab pre-treated cell lines. To remove bound tafasitamab from the cell surface, an acidic dissociation protocol was used. Antibody affinities for CD19 were measured using Surface Plasmon Resonance (SPR) or Bio-Layer Interferometry (BLI). Results While CD19 was successfully detected on tafasitamab pre-treated samples using all 7 tested IHC antibody clones, all 8 tested FC antibody clones were confirmed to compete with tafasitamab. An acidic dissociation was demonstrated essential to circumvent CD19 masking by tafasitamab and avoid false negative FC results. Discussion The current study highlights the importance of selecting appropriate CD19 detection tools and techniques for correct interpretation of CD19 expression. The findings presented herein can serve as a guideline to investigators and may help navigate treatment strategies in the clinical setting.
Collapse
Affiliation(s)
- Kristina Ilieva
- Department of Translational Research, Translational Sciences, MorphoSys AG, Planegg, Germany
| | - Markus Eberl
- Department of Translational Research, Translational Sciences, MorphoSys AG, Planegg, Germany
| | - Jan Jaehrling
- Department of Analytical Sciences, Translational Sciences, MorphoSys AG, Planegg, Germany
| | - Derek Blair
- Department of Clinical Biomarkers & Companion Diagnostics, Translational Sciences, MorphoSys AG, Planegg, Germany
| | - Maria Patra-Kneuer
- Department of Translational Research, Translational Sciences, MorphoSys AG, Planegg, Germany
| | - Rainer Boxhammer
- Department of Clinical Biomarkers & Companion Diagnostics, Translational Sciences, MorphoSys AG, Planegg, Germany
| | | | - Christina Heitmüller
- Department of Translational Research, Translational Sciences, MorphoSys AG, Planegg, Germany
| |
Collapse
|
13
|
Belada D, Kopeckova K, Bergua Burgues JM, Stevens D, André M, Persona EP, Pichler P, Staber PB, Trneny M, Duell J, Waldron-Lynch M, Wagner S, Mukhopadhyay A, Dirnberger-Hertweck M, Burke JM, Nowakowski GS. Safety and efficacy of tafasitamab with or without lenalidomide added to first-line R-CHOP for DLBCL: the phase 1b First-MIND study. Blood 2023; 142:1348-1358. [PMID: 37369099 PMCID: PMC10651865 DOI: 10.1182/blood.2023020637] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/08/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Anti-CD19 immunotherapy tafasitamab is used in combination with lenalidomide in patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL) who are ineligible for autologous stem cell transplant. Open-label, phase 1b, First-MIND study assessed safety and preliminary efficacy of tafasitamab + R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) ± lenalidomide as first-line therapy in patients with DLBCL. From December 2019 to August 2020, 83 adults with untreated DLBCL (International Prognostic Index 2-5) were screened and 66 were randomly assigned (33 per arm) to R-CHOP-tafasitamab (arm T) or R-CHOP-tafasitamab-lenalidomide (arm T/L) for 6 cycles. Primary end point was safety; secondary end points included end-of-treatment (EoT) overall response rate (ORR) and complete response (CR) rate. All patients had ≥1 treatment-emergent adverse event, mostly grade 1 or 2. Grade ≥3 neutropenia and thrombocytopenia occurred, respectively, in 57.6% and 12.1% (arm T) and 84.8% and 36.4% (arm T/L) of patients. Nonhematologic toxicities occurred at similar rates among arms. R-CHOP mean relative dose intensity was ≥89% in both arms. EoT ORR was 75.8% (CR 72.7%) in arm T and 81.8% (CR 66.7%) in arm T/L; best ORR across visits was 90.0% and 93.9%. Eighteen-month duration of response and of CR rates were 72.7% and 74.5% (arm T) and 78.7% and 86.5% (arm T/L); 24-month progression-free and overall survival rates were 72.7% and 90.3% (arm T) and 76.8% and 93.8% (arm T/L). Manageable safety and promising signals of efficacy were observed in both arms. Potential benefit of adding tafasitamab + lenalidomide to R-CHOP is being investigated in phase 3 frontMIND (NCT04824092). This study is registered at www.clinicaltrials.gov as #NCT04134936.
Collapse
Affiliation(s)
- David Belada
- 4th Department of Internal Medicine—Hematology, Charles University, Hospital and Faculty of Medicine, Hradec Králové, Czech Republic
| | - Katerina Kopeckova
- Department of Oncology of the 2nd Faculty of Medicine of Charles University and University Hospital in Motol, Prague, Czech Republic
| | | | - Don Stevens
- Norton Cancer Institute—St. Matthews Campus, Louisville, KY
| | - Marc André
- Department of Hematology, Université Catholique de Louvain, CHU UCL Namur, Yvoir, Belgium
| | - Ernesto Perez Persona
- Bioaraba (Onco-hematology Research Group), Vitoria-Gasteiz, Spain
- Department of Hematology, Osakidetza Basque Health Service, Araba University Hospital, Vitoria-Gasteiz, Spain
| | - Petra Pichler
- Department of Internal Medicine, University Hospital of St. Pölten, St. Pölten, Austria
- Karl Landsteiner University of Health Sciences, Karl Landsteiner Institute for Nephrology and Hemato Oncology, St. Pölten, Austria
| | - Philipp B. Staber
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Marek Trneny
- Charles University General Hospital, Prague, Czech Republic
| | - Johannes Duell
- Medizinische Klinik und Poliklinik II, Universitätsklinik Würzburg, Würzburg, Germany
| | | | | | | | | | - John M. Burke
- US Oncology Research and Rocky Mountain Cancer Centers, Aurora, CO
| | | |
Collapse
|
14
|
Lucero OM, Lee JA, Bowman J, Johnson K, Sapparapu G, Thomas JK, Fan G, Chang BH, Thiel-Klare K, Eide CA, Okada C, Palazzolo M, Lind E, Kosaka Y, Druker BJ, Lydon N, Bowers PM. Patient-Specific Targeting of the T-Cell Receptor Variable Region as a Therapeutic Strategy in Clonal T-Cell Diseases. Clin Cancer Res 2023; 29:4230-4241. [PMID: 37199721 PMCID: PMC10592575 DOI: 10.1158/1078-0432.ccr-22-0906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 05/19/2023]
Abstract
PURPOSE Targeted therapeutics are a goal of medicine. Methods for targeting T-cell lymphoma lack specificity for the malignant cell, leading to elimination of healthy cells. The T-cell receptor (TCR) is designed for antigen recognition. T-cell malignancies expand from a single clone that expresses one of 48 TCR variable beta (Vβ) genes, providing a distinct therapeutic target. We hypothesized that a mAb that is exclusive to a specific Vβ would eliminate the malignant clone while having minimal effects on healthy T cells. EXPERIMENTAL DESIGN We identified a patient with large granular T-cell leukemia and sequenced his circulating T-cell population, 95% of which expressed Vβ13.3. We developed a panel of anti-Vβ13.3 antibodies to test for binding and elimination of the malignant T-cell clone. RESULTS Therapeutic antibody candidates bound the malignant clone with high affinity. Antibodies killed engineered cell lines expressing the patient TCR Vβ13.3 by antibody-dependent cellular cytotoxicity and TCR-mediated activation-induced cell death, and exhibited specific killing of patient malignant T cells in combination with exogenous natural killer cells. EL4 cells expressing the patient's TCR Vβ13.3 were also killed by antibody administration in an in vivo murine model. CONCLUSIONS This approach serves as an outline for development of therapeutics that can treat clonal T-cell-based malignancies and potentially other T-cell-mediated diseases. See related commentary by Varma and Diefenbach, p. 4024.
Collapse
Affiliation(s)
- Olivia M Lucero
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Ji-Ann Lee
- Clinical and Translational Science Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Jenna Bowman
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Kara Johnson
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Gopal Sapparapu
- Clinical and Translational Science Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - John K Thomas
- Clinical and Translational Science Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Guang Fan
- Department of Pathology and Clinical Laboratory Medicine, Oregon Health & Science University, Portland, Oregon
| | - Bill H Chang
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Division of Pediatric Hematology and Oncology, Oregon Health & Science University, Portland, Oregon
| | - Karina Thiel-Klare
- Division of Pediatric Hematology and Oncology, Oregon Health & Science University, Portland, Oregon
| | - Christopher A Eide
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Craig Okada
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Mike Palazzolo
- Clinical and Translational Science Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Evan Lind
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Yoko Kosaka
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Division of Pediatric Hematology and Oncology, Oregon Health & Science University, Portland, Oregon
| | - Brian J Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
- VB Therapeutics LLC, Jackson, Wyoming
| | | | - Peter M Bowers
- Therapeutic Antibody Laboratory, Department of Pulmonology and Critical Care, David Geffen School of Medicine, Los Angeles, California
| |
Collapse
|
15
|
Lu T, Zhang J, Xu-Monette ZY, Young KH. The progress of novel strategies on immune-based therapy in relapsed or refractory diffuse large B-cell lymphoma. Exp Hematol Oncol 2023; 12:72. [PMID: 37580826 PMCID: PMC10424456 DOI: 10.1186/s40164-023-00432-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/30/2023] [Indexed: 08/16/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) can be cured with standard front-line immunochemotherapy, whereas nearly 30-40% of patients experience refractory or relapse. For several decades, the standard treatment strategy for fit relapsed/refractory (R/R) DLBCL patients has been high-dose chemotherapy followed by autologous hematopoietic stem cell transplant (auto-SCT). However, the patients who failed in salvage treatment or those ineligible for subsequent auto-SCT have dismal outcomes. Several immune-based therapies have been developed, including monoclonal antibodies, antibody-drug conjugates, bispecific T-cell engaging antibodies, chimeric antigen receptor T-cells, immune checkpoint inhibitors, and novel small molecules. Meanwhile, allogeneic SCT and radiotherapy are still necessary for disease control for fit patients with certain conditions. In this review, to expand clinical treatment options, we summarize the recent progress of immune-related therapies and prospect the future indirections in patients with R/R DLBCL.
Collapse
Affiliation(s)
- Tingxun Lu
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province, 214122, China
- Division of Hematopathology, Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jie Zhang
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province, 214122, China
| | - Zijun Y Xu-Monette
- Division of Hematopathology, Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Durham, NC, 27710, USA
| | - Ken H Young
- Division of Hematopathology, Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Cancer Institute, Durham, NC, 27710, USA.
| |
Collapse
|
16
|
Patra-Kneuer M, Chang G, Xu W, Augsberger C, Grau M, Zapukhlyak M, Ilieva K, Landgraf K, Mangelberger-Eberl D, Yousefi K, Berning P, Kurz KS, Ott G, Klener P, Khandanpour C, Horna P, Schanzer J, Steidl S, Endell J, Heitmüller C, Lenz G. Activity of tafasitamab in combination with rituximab in subtypes of aggressive lymphoma. Front Immunol 2023; 14:1220558. [PMID: 37600821 PMCID: PMC10433160 DOI: 10.3389/fimmu.2023.1220558] [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: 05/10/2023] [Accepted: 07/14/2023] [Indexed: 08/22/2023] Open
Abstract
Background Despite recent advances in the treatment of aggressive lymphomas, a significant fraction of patients still succumbs to their disease. Thus, novel therapies are urgently needed. As the anti-CD20 antibody rituximab and the CD19-targeting antibody tafasitamab share distinct modes of actions, we investigated if dual-targeting of aggressive lymphoma B-cells by combining rituximab and tafasitamab might increase cytotoxic effects. Methods Antibody single and combination efficacy was determined investigating different modes of action including direct cytotoxicity, antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) in in vitro and in vivo models of aggressive B-cell lymphoma comprising diffuse large B-cell lymphoma (DLBCL) and Burkitt lymphoma (BL). Results Three different sensitivity profiles to antibody monotherapy or combination treatment were observed in in vitro models: while 1/11 cell lines was primarily sensitive to tafasitamab and 2/11 to rituximab, the combination resulted in enhanced cell death in 8/11 cell lines in at least one mode of action. Treatment with either antibody or the combination resulted in decreased expression of the oncogenic transcription factor MYC and inhibition of AKT signaling, which mirrored the cell line-specific sensitivities to direct cytotoxicity. At last, the combination resulted in a synergistic survival benefit in a PBMC-humanized Ramos NOD/SCID mouse model. Conclusion This study demonstrates that the combination of tafasitamab and rituximab improves efficacy compared to single-agent treatments in models of aggressive B-cell lymphoma in vitro and in vivo.
Collapse
Affiliation(s)
| | - Gaomei Chang
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | - Wendan Xu
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | | | - Michael Grau
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | - Myroslav Zapukhlyak
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | | | | | | | - Kasra Yousefi
- Translational Research, MorphoSys AG, Planegg, Germany
| | - Philipp Berning
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| | - Katrin S. Kurz
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Stuttgart, Germany
| | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Stuttgart, Germany
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University Prague, Prague, Czechia
- First Medical Department, Department of Hematology, Charles University General Hospital Prague, Prague, Czechia
| | - Cyrus Khandanpour
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
- Hematology and Oncology Clinic, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Pedro Horna
- Division of Hematopathology, Mayo Clinic, Rochester, MN, United States
| | | | - Stefan Steidl
- Translational Research, MorphoSys AG, Planegg, Germany
| | - Jan Endell
- Translational Research, MorphoSys AG, Planegg, Germany
| | | | - Georg Lenz
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster, Germany
| |
Collapse
|
17
|
Abdulhaq H, Hwang A, Mahmood O. Targeted Treatment of Adults with Relapsed or Refractory Diffuse Large B-Cell Lymphoma (DLBCL): Tafasitamab in Context. Onco Targets Ther 2023; 16:617-629. [PMID: 37492075 PMCID: PMC10364833 DOI: 10.2147/ott.s372783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/06/2023] [Indexed: 07/27/2023] Open
Abstract
The outcomes of Relapsed/Refractory (R/R) Diffuse Large B-cell lymphoma have been historically poor. The recent development of several novel therapies including CD19 directed agents has improved the prognosis of this disease significantly. Chimeric antigen receptor (CAR) T-cell therapy has drastically changed the treatment of R/R DLBCL, but it is still associated with significant barriers and limited access. Tafasitamab (an anti-CD19 engineered monoclonal antibody), in addition to lenalidomide, has shown significant efficacy with exceptionally durable responses in patients with R/R DLBCL who are ineligible for autologous stem cell transplantation (ASCT). Tafasitamab-lenalidomide and certain other therapies (ie, antibody-drug conjugates and bispecific antibodies) are important treatment options for patients who are ineligible for CAR-T due to co-morbidities or lack of access, and patients with rapid progression of disease who are unable to wait for manufacturing of CAR-T. This review will thus discuss currently approved and recently studied targeted treatment options for patients with R/R DLBCL with an emphasis on CAR-T alternative options, particularly Tafasitamab-lenalidomide.
Collapse
Affiliation(s)
- Haifaa Abdulhaq
- Division of Hematology/Oncology, University of California San Francisco, Fresno, CA, USA
| | - Andrew Hwang
- Division of Hematology/Oncology, University of California San Francisco, Fresno, CA, USA
| | - Omar Mahmood
- Division of Hematology/Oncology, University of California San Francisco, Fresno, CA, USA
| |
Collapse
|
18
|
Bhattacharyya P, Christopherson RI, Skarratt KK, Chen JZ, Balle T, Fuller SJ. Combination of High-Resolution Structures for the B Cell Receptor and Co-Receptors Provides an Understanding of Their Interactions with Therapeutic Antibodies. Cancers (Basel) 2023; 15:2881. [PMID: 37296844 PMCID: PMC10251933 DOI: 10.3390/cancers15112881] [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: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
B cells are central to the adaptive immune response, providing long lasting immunity after infection. B cell activation is mediated by a cell surface B cell receptor (BCR) following recognition of an antigen. BCR signaling is modulated by several co-receptors including CD22 and a complex that contains CD19 and CD81. Aberrant signaling through the BCR and co-receptors promotes the pathogenesis of several B cell malignancies and autoimmune diseases. Treatment of these diseases has been revolutionized by the development of monoclonal antibodies that bind to B cell surface antigens, including the BCR and its co-receptors. However, malignant B cells can escape targeting by several mechanisms and until recently, rational design of antibodies has been limited by the lack of high-resolution structures of the BCR and its co-receptors. Herein we review recently determined cryo-electron microscopy (cryo-EM) and crystal structures of the BCR, CD22, CD19 and CD81 molecules. These structures provide further understanding of the mechanisms of current antibody therapies and provide scaffolds for development of engineered antibodies for treatment of B cell malignancies and autoimmune diseases.
Collapse
Affiliation(s)
- Puja Bhattacharyya
- Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW 2750, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | | | - Kristen K. Skarratt
- Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW 2750, Australia
- Nepean Hospital, Kingswood, NSW 2747, Australia
| | - Jake Z. Chen
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Brain and Mind Centre, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Thomas Balle
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Brain and Mind Centre, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Stephen J. Fuller
- Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW 2750, Australia
- Nepean Hospital, Kingswood, NSW 2747, Australia
| |
Collapse
|
19
|
Mohammadi M, Jeddi-Tehrani M, Golsaz-Shirazi F, Arjmand M, Torkashvand F, Bahadori T, Judaki MA, Shiravi F, Ahmadi Zare H, Notash Haghighat F, Mobini M, Shokri F, Amiri MM. A Novel Fc-Engineered Anti-HER2 Bispecific Antibody With Enhanced Antitumor Activity. J Immunother 2023; 46:121-131. [PMID: 36939675 DOI: 10.1097/cji.0000000000000464] [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: 10/09/2022] [Accepted: 02/17/2023] [Indexed: 03/21/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression has been demonstrated in a variety of cancers. Targeted therapy with anti-HER2 monoclonal antibodies (mAbs) has been approved as a therapeutic modality. Despite the efficacy of mAbs in tumor treatment, many patients do not benefit from this therapeutic platform. Fragment crystallizable (Fc) engineering is a common approach to improve the efficacy of therapeutic mAbs. Five Fc-engineered mAbs have so far been approved by FDA. We have recently developed an anti-HER2 bispecific mAb, BiHT, constructed from variable domains of trastuzumab, and our novel humanized anti-HER2 mAb, hersintuzumab. BiHT displayed promising antitumor activity as potently as the combination of the parental mAbs. Here, we aimed to modify the Fc of BiHT to improve its therapeutic efficacy. The Fc-engineered BiHT (MBiHT) bound to recombinant HER2 and its subdomains with an affinity similar to BiHT. It also recognized native HER2 on different cell lines, inhibited their proliferation, downregulated HER2 expression, and suppressed downstream signaling pathways similar to BiHT. Compared with BiHT, MBiHT displayed enhanced antibody-dependent cellular cytotoxicity activity against various tumor cell lines. It also inhibited the growth of ovarian xenograft tumors in nude mice more potently than BiHT. Our findings suggest that MBiHT could be a potent therapeutic candidate for the treatment of HER2-overexpressing cancer types.
Collapse
Affiliation(s)
- Mehdi Mohammadi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences
| | | | - Forough Golsaz-Shirazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences
| | | | | | - Tannaz Bahadori
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences
| | - Mohammad Ali Judaki
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences
| | - Fariba Shiravi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences
| | | | | | - Maryam Mobini
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences
| | - Fazel Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences
| |
Collapse
|
20
|
Nedved A, Maddocks K, Nowakowski GS. Clinical Treatment Guidelines for Tafasitamab Plus Lenalidomide in Patients with Relapsed or Refractory Diffuse Large B-Cell Lymphoma. Oncologist 2023; 28:199-207. [PMID: 36648324 PMCID: PMC10020798 DOI: 10.1093/oncolo/oyac256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 10/27/2022] [Indexed: 01/18/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) accounts for approximately 24% of new cases of B-cell non-Hodgkin lymphoma in the US each year. Up to 50% of patients relapse or are refractory (R/R) to the standard first-line treatment option, R-CHOP. The anti-CD19 monoclonal antibody tafasitamab, in combination with lenalidomide (LEN), is an NCCN preferred regimen for transplant-ineligible patients with R/R DLBCL and received accelerated approval in the US (July 2020) and conditional marketing authorization in Europe (August 2021) and other countries, based on data from the L-MIND study. The recommended dose of tafasitamab is 12 mg/kg by intravenous infusion, administered in combination with LEN 25 mg for 12 cycles, followed by tafasitamab monotherapy until disease progression or unacceptable toxicity. Tafasitamab + LEN is associated with durable responses in patients with R/R DLBCL. The majority of clinically significant treatment-associated adverse events are attributable to LEN and can be managed with dose modification and supportive therapy. We provide guidelines for the management of patients with R/R DLBCL treated with tafasitamab and LEN in routine clinical practice, including elderly patients and those with renal and hepatic impairment, and advice regarding patient education as part of a comprehensive patient engagement plan. Our recommendations include LEN administration at a reduced dose if required in patients unable to tolerate the recommended dose. No dose modification is required for tafasitamab in special patient populations.
Collapse
Affiliation(s)
| | - Kami Maddocks
- Arthur G. James Comprehensive Cancer Center, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Grzegorz S Nowakowski
- Corresponding author: Grzegorz S. Nowakowski, MD, Division of Hematology, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA.
| |
Collapse
|
21
|
Aguilar OA, Gonzalez-Hinojosa MD, Arakawa-Hoyt JS, Millan AJ, Gotthardt D, Nabekura T, Lanier LL. The CD16 and CD32b Fc-gamma receptors regulate antibody-mediated responses in mouse natural killer cells. J Leukoc Biol 2023; 113:27-40. [PMID: 36822164 PMCID: PMC10197019 DOI: 10.1093/jleuko/qiac003] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Indexed: 01/12/2023] Open
Abstract
Natural killer (NK) cells are innate lymphocytes capable of mediating immune responses without prior sensitization. NK cells express Fc-gamma receptors (FcγRs) that engage the Fc region of IgG. Studies investigating the role of FcγRs on mouse NK cells have been limited due to lack specific reagents. In this study, we characterize the expression and biological consequences of activating mouse NK cells through their FcγRs. We demonstrate that most NK cells express the activating CD16 receptor, and a subset of NK cells also expresses the inhibitory CD32b receptor. Critically, these FcγRs are functional on mouse NK cells and can modulate antibody-mediated responses. We also characterized mice with conditional knockout alleles of Fcgr3 (CD16) or Fcgr2b (CD32b) in the NK and innate lymphoid cell (ILC) lineage. NK cells in these mice did not reveal any developmental defects and were responsive to cross-linking activating NK receptors, cytokine stimulation, and killing of YAC-1 targets. Importantly, CD16-deficient NK cells failed to induce antibody-directed cellular cytotoxicity of antibody-coated B-cell lymphomas in in vitro assays. In addition, we demonstrate the important role of CD16 on NK cells using an in vivo model of cancer immunotherapy using anti-CD20 antibody treatment of B-cell lymphomas.
Collapse
Affiliation(s)
- Oscar A. Aguilar
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Maria D.R. Gonzalez-Hinojosa
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Janice S. Arakawa-Hoyt
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Alberto J. Millan
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Dagmar Gotthardt
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Present Address: Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Tsukasa Nabekura
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki, Japan
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| |
Collapse
|
22
|
Wang ZH, Li W, Dong H, Han F. Current state of NK cell-mediated immunotherapy in chronic lymphocytic leukemia. Front Oncol 2023; 12:1077436. [PMID: 37078002 PMCID: PMC10107371 DOI: 10.3389/fonc.2022.1077436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) has become one of the most common hematological diseases in western countries, with an annual incidence of 42/100,000. Conventional chemotherapy and targeted therapeutic drugs showed limitations in prognosis or in efficiency in high-risk patients. Immunotherapy represented is one of the most effective therapeutic approaches with the potential of better effect and prognosis. Natural killer (NK) cells are good options for immunotherapy as they can effectively mediate anti-tumor activity of immune system by expressing activating and inhibiting receptors and recognizing specific ligands on various tumor cells. NK cells are critical in the immunotherapy of CLL by enhancing self-mediated antibody-dependent cytotoxicity (ADCC), allogeneic NK cell therapy and chimeric antigen receptor-natural killer (CAR-NK) cell therapy. In this article, we reviewed the features, working mechanisms, and receptors of NK cells, and the available evidence of the advantages and disadvantages of NK cell-based immunotherapies, and put forward future study directions in this field.
Collapse
Affiliation(s)
- Zong-Han Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Li
- Department of General Surgery, Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Hao Dong
- Department of Gastrointestinal Nutrition and Surgical Surgery, The Second Affiliated Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Hao Dong, ; Fujun Han,
| | - Fujun Han
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Hao Dong, ; Fujun Han,
| |
Collapse
|
23
|
McCurry D, Flowers CR, Bermack C. Immune-based therapies in diffuse large B-cell lymphoma. Expert Opin Investig Drugs 2023; 32:479-493. [PMID: 37394970 DOI: 10.1080/13543784.2023.2230137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/23/2023] [Indexed: 07/04/2023]
Abstract
INTRODUCTION Diffuse large B-cell lymphoma (DLBCL) is an aggressive and clinically heterogeneous malignancy originating from B-cells with up to 40% of patients experiencing primary refractory disease or relapse after first-line treatment. However, the past 5 years have seen a flurry of new drug approvals for DLBCL anchored upon new immune therapies, including chimeric antigen receptor (CAR) T-cells and antibody-based therapies. AREAS COVERED This article summarizes recent advances in the treatment of DLBCL, including in the first line and relapsed and refractory setting (second-line and beyond). A literature search was conducted for publications relevant to the immunotherapeutic approach to DLBCL from 2000 through March 2023 within PubMed and articles were reviewed. The search terms were immunotherapy, monoclonal antibodies, chimeric antigen receptor modified T-cell (CAR-T), and classification of DLBCL. Relevant clinical trials and pre-clinical studies exploring the strengths and weaknesses of current immune therapies against DLBCL were chosen. We additionally explored how intrinsic differences amongst DLBCL subtype biology and endogenous host immune recruitment contribute to variable therapeutic efficacy. EXPERT OPINION Future treatments will minimize chemotherapy exposure and be chosen by underlying tumor biology, paving the way for the promise of chemotherapeutic free regimens and improved outcomes for poor-risk subgroups.
Collapse
Affiliation(s)
- Dustin McCurry
- Oncology Fellow, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Christopher R Flowers
- Division Head Ad Interim of Cancer Medicine, Chair and Professor of the Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Casey Bermack
- Oncology Fellow, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
24
|
Sermer D, Elavalakanar P, Abramson JS, Palomba ML, Salles G, Arnason J. Targeting CD19 for diffuse large B cell lymphoma in the era of CARs: Other modes of transportation. Blood Rev 2023; 57:101002. [PMID: 35989138 DOI: 10.1016/j.blre.2022.101002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 01/28/2023]
Abstract
CD19 is nearly ubiquitously expressed on B-lymphocytes and in B-cell malignancies. Although CD19-directed CAR T cells have greatly improved outcomes in B-cell malignancies, there are significant limitations with this therapy. CD19 can also be effectively targeted by other drug classes, such as monoclonal antibodies, antibody-drug conjugates, and bispecific T cell engagers or antibodies. However, the optimal patient selection and sequencing of these novel therapies has not yet been established. In this review, we discuss the utilization of CD19 as a target for the treatment of DLBCL, focusing on tafasitamab, loncastuximab tesirine, and blinatumomab. We provide a comprehensive review of the pivotal clinical trials, discussing the strength and limitations of the data for each agent. We explore the emerging evidence that CD19 expression is retained following exposure to these agents and that patients can be successfully re-challenged with anti-CD19 therapies of a different drug class upon disease relapse post-CAR T cells. Finally, we discuss how these drugs potentially fit into the most current treatment paradigm for DLBCL.
Collapse
Affiliation(s)
- David Sermer
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | | | - Jeremy S Abramson
- Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - M Lia Palomba
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Weill-Cornell Medical College, New York, NY, USA
| | - Gilles Salles
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Weill-Cornell Medical College, New York, NY, USA
| | - Jon Arnason
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Iwata Y, Katada H, Okuda M, Doi Y, Ching TJ, Harada A, Takeiri A, Honda M, Mishima M. Preclinical in vitro evaluation of immune suppression induced by GYM329, Fc-engineered sweeping antibody. J Toxicol Sci 2023; 48:399-409. [PMID: 37394653 DOI: 10.2131/jts.48.399] [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: 07/04/2023]
Abstract
Fc-engineering is commonly used to improve the therapeutic potency of antibody (Ab) treatments. Because FcγRIIb is the only inhibitory FcγR that contains an immunoreceptor tyrosine-based inhibition motif (ITIM), Fc-engineered Abs with enhanced binding affinity to FcγRIIb might provide immune suppression in clinical contexts. GYM329 is an anti-latent myostatin Fc-engineered Ab with increased affinity to FcγRIIb which is expected to improve muscle strength in patients with muscular disorders. Cross-linking of FcγRIIb by immune complex (IC) results in phosphorylation of ITIM to inhibit immune activation and apoptosis in B cells. We examined whether the IC of Fc-engineered Abs with enhanced binding affinity to FcγRIIb causes phosphorylation of ITIM or B cell apoptosis using GYM329 and its Fc variant Abs in human and cynomolgus-monkey (cyno) immune cells in vitro. IC of GYM329 with enhanced binding affinity to human FcγRIIb (×5) induced neither ITIM phosphorylation nor B cell apoptosis. As for GYM329, FcγRIIb should work as an endocytic receptor of small IC to sweep latent myostatin, so it is preferable that GYM329 induces neither ITIM phosphorylation nor B cell apoptosis to prevent immune suppression. In contrast, IC of myo-HuCy2b, the Ab with enhanced binding affinity to human FcγRIIb (×4), induced ITIM phosphorylation and B cell apoptosis. The result of the present study demonstrated that Fc-engineered Abs with similar binding affinity to FcγRIIb had different effects. Thus, it is important to also investigate FcγR-mediated immune functions other than binding to fully understand the biological effects of Fc-engineered Abs.
Collapse
Affiliation(s)
- Yoshika Iwata
- Translational Research Division, Chugai Pharmaceutical Co., Ltd
| | | | | | - Yoshiaki Doi
- Research Division, Chugai Pharmaceutical Co., Ltd
| | | | - Asako Harada
- Translational Research Division, Chugai Pharmaceutical Co., Ltd
| | - Akira Takeiri
- Translational Research Division, Chugai Pharmaceutical Co., Ltd
| | - Masaki Honda
- Translational Research Division, Chugai Pharmaceutical Co., Ltd
| | | |
Collapse
|
26
|
Kubacz M, Kusowska A, Winiarska M, Bobrowicz M. In Vitro Diffuse Large B-Cell Lymphoma Cell Line Models as Tools to Investigate Novel Immunotherapeutic Strategies. Cancers (Basel) 2022; 15:cancers15010235. [PMID: 36612228 PMCID: PMC9818372 DOI: 10.3390/cancers15010235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Despite the high incidence of diffuse large B-cell lymphoma (DLBCL), its management constitutes an ongoing challenge. The most common DLBCL variants include activated B-cell (ABC) and germinal center B-cell-like (GCB) subtypes including DLBCL with MYC and BCL2/BCL6 rearrangements which vary among each other with sensitivity to standard rituximab (RTX)-based chemoimmunotherapy regimens and lead to distinct clinical outcomes. However, as first line therapies lead to resistance/relapse (r/r) in about half of treated patients, there is an unmet clinical need to identify novel therapeutic strategies tailored for these patients. In particular, immunotherapy constitutes an attractive option largely explored in preclinical and clinical studies. Patient-derived cell lines that model primary tumor are indispensable tools that facilitate preclinical research. The current review provides an overview of available DLBCL cell line models and their utility in designing novel immunotherapeutic strategies.
Collapse
Affiliation(s)
- Matylda Kubacz
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Aleksandra Kusowska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Magdalena Winiarska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Małgorzata Bobrowicz
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
- Correspondence:
| |
Collapse
|
27
|
Tannoury M, Garnier D, Susin SA, Bauvois B. Current Status of Novel Agents for the Treatment of B Cell Malignancies: What's Coming Next? Cancers (Basel) 2022; 14:6026. [PMID: 36551511 PMCID: PMC9775488 DOI: 10.3390/cancers14246026] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Resistance to death is one of the hallmarks of human B cell malignancies and often contributes to the lack of a lasting response to today's commonly used treatments. Drug discovery approaches designed to activate the death machinery have generated a large number of inhibitors of anti-apoptotic proteins from the B-cell lymphoma/leukemia 2 family and the B-cell receptor (BCR) signaling pathway. Orally administered small-molecule inhibitors of Bcl-2 protein and BCR partners (e.g., Bruton's tyrosine kinase and phosphatidylinositol-3 kinase) have already been included (as monotherapies or combination therapies) in the standard of care for selected B cell malignancies. Agonistic monoclonal antibodies and their derivatives (antibody-drug conjugates, antibody-radioisotope conjugates, bispecific T cell engagers, and chimeric antigen receptor-modified T cells) targeting tumor-associated antigens (TAAs, such as CD19, CD20, CD22, and CD38) are indicated for treatment (as monotherapies or combination therapies) of patients with B cell tumors. However, given that some patients are either refractory to current therapies or relapse after treatment, novel therapeutic strategies are needed. Here, we review current strategies for managing B cell malignancies, with a focus on the ongoing clinical development of more effective, selective drugs targeting these molecules, as well as other TAAs and signaling proteins. The observed impact of metabolic reprogramming on B cell pathophysiology highlights the promise of targeting metabolic checkpoints in the treatment of these disorders.
Collapse
Affiliation(s)
| | | | | | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
| |
Collapse
|
28
|
Khosravi M, Khazaeil K, KhademiMoghadam F. Triggering of the immune response to MCF7 cell line using conjugated antibody with bacterial antigens: In-vitro and in-vivo study. PLoS One 2022; 17:e0275776. [PMID: 36206297 PMCID: PMC9543947 DOI: 10.1371/journal.pone.0275776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/22/2022] [Indexed: 11/07/2022] Open
Abstract
The current study intended to trigger the immune response to cancer cells by using antibodies conjugated with bacterial antigens. The protein membrane of the MCF7 cell line was extracted and specific antibodies against cell membrane antigens was produced in rabbits. The specific antibodies were purified using chromatography methods and linked to E. coli antigens or doxorubicin using Diethylenetriamine pentaacetate (DTPA) linker. After confirmation of the conjugation process using SDS-PAGE and ATR-FTIR methods, the MCF7 and HUVEC cells were treated with various concentrations of the prepared conjugated antibodies along with human serum. The toxicity of each treatment against MCF7 and HUVEC cells was evaluated using the MTT assay. Also, polylactic acid scaffolds that contain 10×104 MCF7 cells were surgically placed in the peritoneal cavity of the rats. After treatment of each group, induction of the inflammatory responses was evaluated on stained histological sections of the scaffolds. The lowest cytotoxic doses of the antigen conjugated-antibody, doxorubicin-conjugated-antibody was 4 and 1 μg/mL, respectively. Doxorubicin conjugated antibodies displayed greater toxicity on both MCF7 and HUVEC cells. The in vivo finding revealed that the inflammatory cells were significantly higher in treating animals with antigen conjugated-antibody. The current synthetic agent stimulated the serum toxicity and induced an inflammatory response to MCF7 cell lines. Targeting of the bacterial antigens on tumor sites by immune system elements, could limit the growth of the tumor cells.
Collapse
Affiliation(s)
- Mohammad Khosravi
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- * E-mail:
| | - Kaveh Khazaeil
- Department of Basic sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | | |
Collapse
|
29
|
Gehlert CL, Rahmati P, Boje AS, Winterberg D, Krohn S, Theocharis T, Cappuzzello E, Lux A, Nimmerjahn F, Ludwig RJ, Lustig M, Rösner T, Valerius T, Schewe DM, Kellner C, Klausz K, Peipp M. Dual Fc optimization to increase the cytotoxic activity of a CD19-targeting antibody. Front Immunol 2022; 13:957874. [PMID: 36119088 PMCID: PMC9471254 DOI: 10.3389/fimmu.2022.957874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
Targeting CD19 represents a promising strategy for the therapy of B-cell malignancies. Although non-engineered CD19 antibodies are poorly effective in mediating complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP), these effector functions can be enhanced by Fc-engineering. Here, we engineered a CD19 antibody with the aim to improve effector cell-mediated killing and CDC activity by exchanging selected amino acid residues in the Fc domain. Based on the clinically approved Fc-optimized antibody tafasitamab, which triggers enhanced ADCC and ADCP due to two amino acid exchanges in the Fc domain (S239D/I332E), we additionally added the E345K amino acid exchange to favor antibody hexamerization on the target cell surface resulting in improved CDC. The dual engineered CD19-DEK antibody bound CD19 and Fcγ receptors with similar characteristics as the parental CD19-DE antibody. Both antibodies were similarly efficient in mediating ADCC and ADCP but only the dual optimized antibody was able to trigger complement deposition on target cells and effective CDC. Our data provide evidence that from a technical perspective selected Fc-enhancing mutations can be combined (S239D/I332E and E345K) allowing the enhancement of ADCC, ADCP and CDC with isolated effector populations. Interestingly, under more physiological conditions when the complement system and FcR-positive effector cells are available as effector source, strong complement deposition negatively impacts FcR engagement. Both effector functions were simultaneously active only at selected antibody concentrations. Dual Fc-optimized antibodies may represent a strategy to further improve CD19-directed cancer immunotherapy. In general, our results can help in guiding optimal antibody engineering strategies to optimize antibodies’ effector functions.
Collapse
Affiliation(s)
- Carina Lynn Gehlert
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Pegah Rahmati
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Ammelie Svea Boje
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Dorothee Winterberg
- Department of Pediatrics I, University Hospital Schleswig-Holstein and Christian-Albrechts-University Kiel, Kiel, Germany
| | - Steffen Krohn
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Thomas Theocharis
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Elisa Cappuzzello
- Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Anja Lux
- Division of Genetics, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ralf J. Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Marta Lustig
- Division of Stem Cell Transplantation and Immunotherapy Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Thies Rösner
- Division of Stem Cell Transplantation and Immunotherapy Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Denis Martin Schewe
- Department of Pediatrics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Christian Kellner
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, Ludwig-Maximilians-University (LMU) University Hospital Munich, Munich, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
- *Correspondence: Matthias Peipp,
| |
Collapse
|
30
|
Harnessing natural killer cells for cancer immunotherapy: dispatching the first responders. Nat Rev Drug Discov 2022; 21:559-577. [PMID: 35314852 PMCID: PMC10019065 DOI: 10.1038/s41573-022-00413-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
Abstract
Natural killer (NK) cells have crucial roles in the innate immunosurveillance of cancer and viral infections. They are 'first responders' that can spontaneously recognize abnormal cells in the body, rapidly eliminate them through focused cytotoxicity mechanisms and potently produce pro-inflammatory cytokines and chemokines that recruit and activate other immune cells to initiate an adaptive response. From the initial discovery of the diverse cell surface receptors on NK cells to the characterization of regulatory events that control their function, our understanding of the basic biology of NK cells has improved dramatically in the past three decades. This advanced knowledge has revealed increased mechanistic complexity, which has opened the doors to the development of a plethora of exciting new therapeutics that can effectively manipulate and target NK cell functional responses, particularly in cancer patients. Here, we summarize the basic mechanisms that regulate NK cell biology, review a wide variety of drugs, cytokines and antibodies currently being developed and used to stimulate NK cell responses, and outline evolving NK cell adoptive transfer approaches to treat cancer.
Collapse
|
31
|
Poletto S, Novo M, Paruzzo L, Frascione PMM, Vitolo U. Treatment strategies for patients with Diffuse Large B-Cell Lymphoma. Cancer Treat Rev 2022; 110:102443. [DOI: 10.1016/j.ctrv.2022.102443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/30/2022] [Accepted: 07/27/2022] [Indexed: 11/02/2022]
|
32
|
Nuvvula S, Dahiya S, Patel SA. The Novel Therapeutic Landscape for Relapsed/Refractory Diffuse Large B Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:362-372. [PMID: 34922844 DOI: 10.1016/j.clml.2021.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/06/2021] [Accepted: 11/14/2021] [Indexed: 12/12/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL) is an aggressive malignancy that has been traditionally treated with anthracycline-based chemotherapy, but approximately one-third of patients relapse after first-line therapy or have primary refractoriness. In this focused review, we discuss the 7 novel Food & Drug Administration (FDA)-approved medications for relapsed/refractory (R/R) DLBCL. We describe 5 CD19-targeted therapies, 3 of which are chimeric antigen receptor (CAR)-T cell therapies. We also highlight novel non-cell-based targeted therapies and discuss optimal sequencing considerations based on the goal of treatment, with an emphasis on CAR-T cell therapy as curative intent. We consider the limited tolerability of certain novel agents, prospects for elderly patients, and financial aspects of these approaches. We discuss advantages and limitations of these targeted therapies based on seminal clinical trials. Finally, we summarize ongoing trials involving promising agents making their way into the pharmacologic pipeline. These therapies include allogeneic CAR-T treatments and multi-antigen targeting therapies such as the CD19/CD22 CAR-T and the CD3/CD20 bispecific antibodies mosunetuzumab and odronextamab. We summarize our approach based on the best available evidence as we enter 2022.
Collapse
Affiliation(s)
- Sri Nuvvula
- Department of Medicine - Division of Hematology/Oncology, University of Massachusetts Chan Medical School, UMass Memorial Medical Center, Worcester, MA
| | - Saurabh Dahiya
- Department of Medicine - Division of Hematology/Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| | - Shyam A Patel
- Department of Medicine - Division of Hematology/Oncology, University of Massachusetts Chan Medical School, UMass Memorial Medical Center, Worcester, MA.
| |
Collapse
|
33
|
Sawalha Y, Maddocks K. Novel treatments in B cell non-Hodgkin's lymphomas. BMJ 2022; 377:e063439. [PMID: 35443983 DOI: 10.1136/bmj-2020-063439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The improved understanding of lymphoma biology and recent advances in the field of cancer immunology have paved the way for the development of many effective small molecule inhibitors and immunotherapies in B cell non-Hodgkin's lymphomas. This article reviews novel treatments that have been approved recently by the US Food and Drug Administration and are now routinely used in clinical practice. It discusses their mechanisms of action, efficacy and safety, current therapeutic roles, and future directions in the treatment paradigm of different types of B cell non-Hodgkin's lymphoma. It also reviews other exciting novel treatments that are not yet approved but have unique mechanisms of action and have shown encouraging early results.
Collapse
Affiliation(s)
- Yazeed Sawalha
- Internal Medicine - Division of Hematology, Ohio State University Comprehensive Cancer Centre, Columbus, OH, USA
| | - Kami Maddocks
- Internal Medicine - Division of Hematology, Ohio State University Comprehensive Cancer Centre, Columbus, OH, USA
| |
Collapse
|
34
|
Blanco B, Ramírez-Fernández Á, Bueno C, Argemí-Muntadas L, Fuentes P, Aguilar-Sopeña Ó, Gutierrez-Agüera F, Zanetti SR, Tapia-Galisteo A, Díez-Alonso L, Segura-Tudela A, Castellà M, Marzal B, Betriu S, Harwood SL, Compte M, Lykkemark S, Erce-Llamazares A, Rubio-Pérez L, Jiménez-Reinoso A, Domínguez-Alonso C, Neves M, Morales P, Paz-Artal E, Guedan S, Sanz L, Toribio ML, Roda-Navarro P, Juan M, Menéndez P, Álvarez-Vallina L. Overcoming CAR-Mediated CD19 Downmodulation and Leukemia Relapse with T Lymphocytes Secreting Anti-CD19 T-cell Engagers. Cancer Immunol Res 2022; 10:498-511. [PMID: 35362043 DOI: 10.1158/2326-6066.cir-21-0853] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/06/2021] [Accepted: 02/09/2022] [Indexed: 11/16/2022]
Abstract
Chimeric antigen receptor (CAR)-modified T cells have revolutionized the treatment of CD19-positive hematologic malignancies. Although anti-CD19 CAR-engineered autologous T cells can induce remission in patients with B-cell acute lymphoblastic leukemia, a large subset relapse, most of them with CD19-positive disease. Therefore, new therapeutic strategies are clearly needed. Here, we report a comprehensive study comparing engineered T cells either expressing a second-generation anti-CD19 CAR (CAR-T19) or secreting a CD19/CD3-targeting bispecific T-cell engager antibody (STAb-T19). We found that STAb-T19 cells are more effective than CAR-T19 cells at inducing cytotoxicity, avoiding leukemia escape in vitro, and preventing relapse in vivo. We observed that leukemia escape in vitro is associated with rapid and drastic CAR-induced internalization of CD19 that is coupled with lysosome-mediated degradation, leading to the emergence of transiently CD19-negative leukemic cells that evade the immune response of engineered CAR-T19 cells. In contrast, engineered STAb-T19 cells induce the formation of canonical immunologic synapses and prevent the CD19 downmodulation observed in anti-CD19 CAR-mediated interactions. Although both strategies show similar efficacy in short-term mouse models, there is a significant difference in a long-term patient-derived xenograft mouse model, where STAb-T19 cells efficiently eradicated leukemia cells, but leukemia relapsed after CAR-T19 therapy. Our findings suggest that the absence of CD19 downmodulation in the STAb-T19 strategy, coupled with the continued antibody secretion, allows an efficient recruitment of the endogenous T-cell pool, resulting in fast and effective elimination of cancer cells that may prevent CD19-positive relapses frequently associated with CAR-T19 therapies.
Collapse
Affiliation(s)
- Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain.,Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (RICORS, RD21/0017/0029), Madrid, Spain
| | - Ángel Ramírez-Fernández
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Clara Bueno
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (RICORS, RD21/0017/0029), Madrid, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain.,Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Lidia Argemí-Muntadas
- Immunotherapy and Cell Engineering Laboratory, Department of Engineering, Aarhus University, Aarhus, Denmark
| | - Patricia Fuentes
- Centro de Biología Molecular Severo Ochoa CSIC-UAM, Madrid, Spain
| | - Óscar Aguilar-Sopeña
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, Madrid, Spain.,Lymphocyte Immunobiology Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Francisco Gutierrez-Agüera
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (RICORS, RD21/0017/0029), Madrid, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | | | - Antonio Tapia-Galisteo
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Laura Díez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Alejandro Segura-Tudela
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Maria Castellà
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
| | - Berta Marzal
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
| | - Sergi Betriu
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
| | - Seandean L Harwood
- Immunotherapy and Cell Engineering Laboratory, Department of Engineering, Aarhus University, Aarhus, Denmark
| | - Marta Compte
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Simon Lykkemark
- Immunotherapy and Cell Engineering Laboratory, Department of Engineering, Aarhus University, Aarhus, Denmark
| | - Ainhoa Erce-Llamazares
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Laura Rubio-Pérez
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain.,Chair for Immunology UFV/Merck, Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcón, Madrid, Spain
| | - Anaïs Jiménez-Reinoso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Carmen Domínguez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Maria Neves
- Centro de Biología Molecular Severo Ochoa CSIC-UAM, Madrid, Spain
| | - Pablo Morales
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Estela Paz-Artal
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Sonia Guedan
- Department of Hematology and Oncology, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - María L Toribio
- Centro de Biología Molecular Severo Ochoa CSIC-UAM, Madrid, Spain
| | - Pedro Roda-Navarro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, Madrid, Spain.,Lymphocyte Immunobiology Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Manel Juan
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain.,Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona, Spain.,Plataforma Immunoteràpia Hospital Sant Joan de Déu, Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - Pablo Menéndez
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (RICORS, RD21/0017/0029), Madrid, Spain.,Josep Carreras Leukemia Research Institute, Barcelona, Spain.,Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biomedicine, School of Medicine, Universitat de Barcelona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain.,Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (RICORS, RD21/0017/0029), Madrid, Spain.,Immunotherapy and Cell Engineering Laboratory, Department of Engineering, Aarhus University, Aarhus, Denmark
| |
Collapse
|
35
|
Her JH, Pretscher D, Patra-Kneuer M, Schanzer J, Cho SY, Hwang YK, Hoeres T, Boxhammer R, Heitmueller C, Wilhelm M, Steidl S, Endell J. Tafasitamab mediates killing of B-cell non-Hodgkin's lymphoma in combination with γδ T cell or allogeneic NK cell therapy. Cancer Immunol Immunother 2022; 71:2829-2836. [PMID: 35348812 PMCID: PMC9519642 DOI: 10.1007/s00262-022-03165-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/28/2022] [Indexed: 11/24/2022]
Abstract
Tafasitamab is an Fc-modified monoclonal antibody that binds to CD19, a cell-surface antigen that is broadly expressed on various types of B-cell non-Hodgkin’s lymphoma (NHL). Antibody-dependent cellular cytotoxicity (ADCC), a key mode of action of tafasitamab, is mediated through the binding of tafasitamab’s Fc region to FcγRIIIa receptors on immune effector cells and results in antitumor activity. Despite the proven clinical activity of tafasitamab in combination with lenalidomide in the treatment of diffuse large B-cell lymphoma (DLBCL), a higher number of immune cells in cancer patients may improve the activity of tafasitamab. Here, we characterized two ex vivo-expanded FcγRIIIa receptor—expressing cell types—γδ T and MG4101 natural killer (NK) cells—as effector cells for tafasitamab in vitro, and found that in the presence of these cells tafasitamab was able to induce ADCC against a range of NHL cell lines and patient-derived cells. We also explored the concept of effector cell supplementation during tafasitamab treatment in vivo by coadministering MG4101 NK cells in Raji and Ramos xenograft models of NHL. Combination treatment of tafasitamab and allogeneic MG4101 NK cells in these models demonstrated a survival benefit compared with tafasitamab or MG4101 monotherapy (Raji: 1.7- to 1.9-fold increase in lifespan; Ramos: 2.0- to 4.1-fold increase in lifespan). In conclusion, adoptive cell transfer of ex vivo-expanded allogeneic NK or autologous γδ T cells in combination with tafasitamab treatment may potentially be a promising novel approach to increase the number of immune effector cells and enhance the antitumor effect of tafasitamab.
Collapse
Affiliation(s)
- Jung Hyun Her
- Cell Therapy Research Center, GC LabCell, Yongin, Republic of Korea
| | - Dominik Pretscher
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | | | | | - Sung Yoo Cho
- Cell Therapy Research Center, GC LabCell, Yongin, Republic of Korea
| | - Yu Kyeong Hwang
- Cell Therapy Research Center, GC LabCell, Yongin, Republic of Korea
| | - Timm Hoeres
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | | | | | - Martin Wilhelm
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | | | | |
Collapse
|
36
|
Minson A, Tam C, Dickinson M, Seymour JF. Targeted Agents in the Treatment of Indolent B-Cell Non-Hodgkin Lymphomas. Cancers (Basel) 2022; 14:1276. [PMID: 35267584 PMCID: PMC8908980 DOI: 10.3390/cancers14051276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/24/2022] [Accepted: 02/27/2022] [Indexed: 02/01/2023] Open
Abstract
Targeted therapies continue to change the landscape of lymphoma treatment, resulting in improved therapy options and patient outcomes. Numerous agents are now approved for use in the indolent lymphomas and many others under development demonstrate significant promise. In this article, we review the landscape of targeted agents that apply to the indolent lymphomas, predominantly follicular lymphoma, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinaemia and marginal zone lymphoma. The review covers small molecule inhibitors, immunomodulators and targeted immunotherapies, as well as presenting emerging and promising combination therapies.
Collapse
Affiliation(s)
- Adrian Minson
- Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (C.T.); (M.D.); (J.F.S.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Constantine Tam
- Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (C.T.); (M.D.); (J.F.S.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael Dickinson
- Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (C.T.); (M.D.); (J.F.S.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - John F. Seymour
- Peter MacCallum Cancer Centre & Royal Melbourne Hospital, Melbourne, VIC 3000, Australia; (C.T.); (M.D.); (J.F.S.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
37
|
SAR442085, a novel anti-CD38 antibody with enhanced antitumor activity against multiple myeloma. Blood 2022; 139:1160-1176. [PMID: 35201323 DOI: 10.1182/blood.2021012448] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/23/2021] [Indexed: 12/28/2022] Open
Abstract
Anti-CD38 monoclonal antibodies (mAbs) represent a breakthrough in the treatment of multiple myeloma (MM), yet some patients fail to respond or progress quickly with this therapy, highlighting the need for novel approaches. In this study we compared the preclinical efficacy of SAR442085, a next-generation anti-CD38 mAb with enhanced affinity for activating Fcγ receptors (FcγR), with first-generation anti-CD38 mAb daratumumab and isatuximab. In surface plasmon resonance and cellular binding assays, we found that SAR442085 had higher binding affinity than daratumumab and isatuximab for FcγRIIa (CD32a) and FcγRIIIa (CD16a). SAR442085 also exhibited better in vitro antibody-dependent cellular cytotoxicity (ADCC) against a panel of MM cells expressing variable CD38 receptor densities including MM patients' primary plasma cells. The enhanced ADCC of SAR442085 was confirmed using NK-92 cells bearing low and high affinity FcγRIIIa (CD16a)-158F/V variants. Using MM patients' primary bone marrow cells, we confirmed that SAR442085 had an increased ability to engage FcγRIIIa, resulting in higher natural killer (NK) cell activation and degranulation against primary plasma cells than preexisting Fc wild-type anti-CD38 mAbs. Finally, using huFcgR transgenic mice that express human Fcγ receptors under the control of their human regulatory elements, we demonstrated that SAR442085 had higher NK cell-dependent in vivo antitumor efficacy and better survival than daratumumab and isatuximab against EL4 thymoma or VK*MYC myeloma cells overexpressing human CD38. These results highlight the preclinical efficacy of SAR442085 and support the current evaluation of this next-generation anti-CD38 antibody in phase I clinical development in patients with relapsed/refractory MM.
Collapse
|
38
|
Gambella M, Carlomagno S, Raiola AM, Giannoni L, Ghiggi C, Setti C, Giordano C, Luchetti S, Serio A, Bo A, Falco M, Della Chiesa M, Angelucci E, Sivori S. CD19-Targeted Immunotherapies for Diffuse Large B-Cell Lymphoma. Front Immunol 2022; 13:837457. [PMID: 35280988 PMCID: PMC8911710 DOI: 10.3389/fimmu.2022.837457] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/07/2022] [Indexed: 12/15/2022] Open
Abstract
Surgical resection, chemotherapy and radiotherapy were, for many years, the only available cancer treatments. Recently, the use of immune checkpoint inhibitors and adoptive cell therapies has emerged as promising alternative. These cancer immunotherapies are aimed to support or harness the patient's immune system to recognize and destroy cancer cells. Preclinical and clinical studies, based on the use of T cells and more recently NK cells genetically modified with chimeric antigen receptors retargeting the adoptive cell therapy towards tumor cells, have already shown remarkable results. In this review, we outline the latest highlights and progress in immunotherapies for the treatment of Diffuse Large B-cell Lymphoma (DLBCL) patients, focusing on CD19-targeted immunotherapies. We also discuss current clinical trials and opportunities of using immunotherapies to treat DLBCL patients.
Collapse
Affiliation(s)
- Massimiliano Gambella
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Simona Carlomagno
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Anna Maria Raiola
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Livia Giannoni
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Ghiggi
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Setti
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Chiara Giordano
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Silvia Luchetti
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alberto Serio
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alessandra Bo
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michela Falco
- Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Emanuele Angelucci
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| |
Collapse
|
39
|
Serrano P, Wah Yuen H, Akdemir J, Hartmann M, Reinholz T, Peltier S, Ligensa T, Seiller C, Paraiso Le Bourhis A. Real-world data in drug development strategies for orphan drugs: tafasitamab in B cell lymphoma, a case study for approval based on a single-arm combination trial. Drug Discov Today 2022; 27:1706-1715. [PMID: 35218926 DOI: 10.1016/j.drudis.2022.02.017] [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: 11/05/2021] [Revised: 01/31/2022] [Accepted: 02/19/2022] [Indexed: 12/01/2022]
Abstract
Tafasitamab (TAF) plus lenalidomide (LEN) is a novel treatment option for patients with relapsed/refractory diffuse large B cell lymphoma (rrDLBCL) who are not eligible for autologous stem cell transplantation. The initial US/EU approvals for TAF represent precedents because this is the first time that approval of a novel combination therapy was granted based on a pivotal single-arm trial (SAT). Matching real world-data (RWD) helped to disentangle the contribution of individual agents. In this review, we present the TAF development strategy, the prospective incorporation of RWD within the clinical development plan, the corresponding regulatory hurdles of this strategy, and the prior regulatory actions for other cancer drugs that previously incorporated RWD and propensity score matching in EU and US regulatory submissions. We also outline how RWD could further advance and impact orphan drug development.
Collapse
Affiliation(s)
| | | | | | - Markus Hartmann
- European Consulting & Contracting in Oncology, Trier, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Kusowska A, Kubacz M, Krawczyk M, Slusarczyk A, Winiarska M, Bobrowicz M. Molecular Aspects of Resistance to Immunotherapies-Advances in Understanding and Management of Diffuse Large B-Cell Lymphoma. Int J Mol Sci 2022; 23:ijms23031501. [PMID: 35163421 PMCID: PMC8835809 DOI: 10.3390/ijms23031501] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 12/28/2022] Open
Abstract
Despite the unquestionable success achieved by rituximab-based regimens in the management of diffuse large B-cell lymphoma (DLBCL), the high incidence of relapsed/refractory disease still remains a challenge. The widespread clinical use of chemo-immunotherapy demonstrated that it invariably leads to the induction of resistance; however, the molecular mechanisms underlying this phenomenon remain unclear. Rituximab-mediated therapeutic effect primarily relies on complement-dependent cytotoxicity and antibody-dependent cell cytotoxicity, and their outcome is often compromised following the development of resistance. Factors involved include inherent genetic characteristics and rituximab-induced changes in effectors cells, the role of ligand/receptor interactions between target and effector cells, and the tumor microenvironment. This review focuses on summarizing the emerging advances in the understanding of the molecular basis responsible for the resistance induced by various forms of immunotherapy used in DLBCL. We outline available models of resistance and delineate solutions that may improve the efficacy of standard therapeutic protocols, which might be essential for the rational design of novel therapeutic regimens.
Collapse
Affiliation(s)
- Aleksandra Kusowska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Matylda Kubacz
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
| | - Marta Krawczyk
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Doctoral School of Translational Medicine, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Aleksander Slusarczyk
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, 02-005 Warsaw, Poland
| | - Magdalena Winiarska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Malgorzata Bobrowicz
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.K.); (M.K.); (M.K.); (A.S.); (M.W.)
- Correspondence:
| |
Collapse
|
41
|
Fitzgerald KN, Quesada AE, von Keudell G, Raj S, Lewis NE, Dogan A, Salles G, Palomba ML. CD19 epitope masking by tafasitamab leads to delays in subsequent use of CD19 CAR T-cell therapy in two patients with aggressive mature B-cell lymphomas. Leuk Lymphoma 2021; 63:751-754. [DOI: 10.1080/10428194.2021.1992622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Kelly N. Fitzgerald
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andres E. Quesada
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | | | - Sandeep Raj
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natasha E. Lewis
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Ahmet Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gilles Salles
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M. Lia Palomba
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
42
|
Wilkinson I, Anderson S, Fry J, Julien LA, Neville D, Qureshi O, Watts G, Hale G. Fc-engineered antibodies with immune effector functions completely abolished. PLoS One 2021; 16:e0260954. [PMID: 34932587 PMCID: PMC8691596 DOI: 10.1371/journal.pone.0260954] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/20/2021] [Indexed: 01/12/2023] Open
Abstract
Elimination of the binding of immunoglobulin Fc to Fc gamma receptors (FcγR) is highly desirable for the avoidance of unwanted inflammatory responses to therapeutic antibodies and fusion proteins. Many different approaches have been described in the literature but none of them completely eliminates binding to all of the Fcγ receptors. Here we describe a set of novel variants having specific amino acid substitutions in the Fc region at L234 and L235 combined with the substitution G236R. They show no detectable binding to Fcγ receptors or to C1q, are inactive in functional cell-based assays and do not elicit inflammatory cytokine responses. Meanwhile, binding to FcRn, manufacturability, stability and potential for immunogenicity are unaffected. These variants have the potential to improve the safety and efficacy of therapeutic antibodies and Fc fusion proteins.
Collapse
Affiliation(s)
- Ian Wilkinson
- Absolute Antibody Ltd, Wilton, United Kingdom
- mAbsolve Limited, Oxford, United Kingdom
| | | | - Jeremy Fry
- ProImmune Limited, Oxford, United Kingdom
| | | | - David Neville
- Reading Scientific Services Limited, Reading, United Kingdom
| | | | - Gary Watts
- Abzena Limited, Babraham, United Kingdom
| | - Geoff Hale
- mAbsolve Limited, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
Sawalha Y. Relapsed/Refractory Diffuse Large B-Cell Lymphoma: A Look at the Approved and Emerging Therapies. J Pers Med 2021; 11:1345. [PMID: 34945817 PMCID: PMC8708171 DOI: 10.3390/jpm11121345] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/12/2021] [Accepted: 11/28/2021] [Indexed: 12/11/2022] Open
Abstract
Approximately 40% of patients with diffuse large B cell lymphoma (DLBCL) do not respond or develop relapsed disease after first-line chemoimmunotherapy. A minority of these patients can be cured with autologous hematopoietic stem cell transplantation (AHCT). Although chimeric antigen receptor (CAR) T cells have transformed the treatment paradigm of relapsed/refractory DLBCL, only 30-40% of patients achieve durable remissions. In addition, many patients with relapsed/refractory DLBCL are ineligible to receive treatment with CAR T cells due to comorbidities or logistical limitations. Since 2019, the following four non-CAR T-cell treatments have been approved in relapsed/refractory DLBCL: polatuzumab in combination with bendamustine and rituximab, selinexor, tafasitamab plus lenalidomide, and loncastuximab. In this article, I review the data behind these four approvals and discuss important considerations on their use in clinical practice. I also review emerging therapies that have shown promising early results in relapsed/refractory DLBCL including the bispecific antibodies, antibody-drug conjugates, Bruton tyrosine kinase inhibitors, BCL2 inhibitors, immune checkpoint inhibitors, and epigenetic modifiers.
Collapse
Affiliation(s)
- Yazeed Sawalha
- Department of Internal Medicine, Division of Hematology, Arthur G. James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
44
|
The European Medicines Agency Review of Tafasitamab in Combination With Lenalidomide for the Treatment of Adult Patients With Relapsed/Refractory Diffuse Large B-cell Lymphoma. Hemasphere 2021; 5:e666. [PMID: 34805769 PMCID: PMC8601272 DOI: 10.1097/hs9.0000000000000666] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/01/2021] [Indexed: 11/26/2022] Open
Abstract
Tafasitamab is a humanized monoclonal antibody that binds to the CD19 antigen, which is expressed in tumor cells from patients with diffuse large B-cell lymphoma (DLBCL). On June 24, 2021, a positive opinion for a conditional marketing authorization was issued by the European Medicines Agency (EMA)’s Committee for Medicinal Products for Human Use (CHMP) for tafasitamab, in combination with lenalidomide, for the treatment of adult patients with relapsed or refractory DLBCL who are ineligible for autologous stem cell transplantation. Tafasitamab was evaluated in the phase 2 single-arm, multicenter, open-label L-MIND clinical trial. The primary endpoint of this trial was objective response rate (ORR). The best ORR, achieved at any time during the study, was 56.8% (95% confidence interval: 45.3%–67.8%), and the median duration of response was 34.6 months (95% confidence interval: 26.1–not reached). The most frequently reported adverse events by system organ class were infections and infestations (72.8%; grade ≥3: 29.6%), blood and lymphatic system disorders (65.4%; grade ≥3: 56.8%), gastrointestinal disorders (64.2%; grade ≥3: 2.5%), and general disorders and administration site conditions (58.0%; grade ≥3: 8.6%). The aim of this article is to summarize the scientific review of the application which led to the positive opinion by the CHMP.
Collapse
|
45
|
Klisovic RB, Leung WH, Brugger W, Dirnberger‐Hertweck M, Winderlich M, Ambarkhane SV, Jabbour EJ. A phase 2a, single-arm, open-label study of tafasitamab, a humanized, Fc-modified, anti-CD19 antibody, in patients with relapsed/refractory B-precursor cell acute lymphoblastic leukemia. Cancer 2021; 127:4190-4197. [PMID: 34343354 PMCID: PMC9292493 DOI: 10.1002/cncr.33796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND B-precursor cell acute lymphoblastic leukemia (B-ALL) in adults is an aggressive and challenging condition, and patients with relapsed/refractory (R/R) disease after allogeneic stem cell transplantation (SCT), or noncandidates for SCT, have a particularly poor prognosis. The authors investigated the activity of the Fc-modified anti-CD19 antibody tafasitamab in adults with R/R B-ALL (NCT01685021). METHODS Adults with R/R B-ALL received single-agent tafasitamab 12 mg/kg weekly for up to four 28-day cycles. Patients with complete remission (with or without neutrophil/platelet recovery; complete remission [CR] or complete remission with incomplete count recovery [CRi]) after cycles 2, 3, or 4 could continue tafasitamab every 2 weeks for up to 3 further months. The primary end point was overall response rate (ORR). RESULTS Twenty-two patients were treated (median, 2 prior lines of therapy; range, 1-8). Six patients completed 2 cycles, and 2 of these patients responded for an ORR of 9%; 16 patients (73%) progressed before their first response assessment. Responses lasted 8 and 4 weeks in the 2 patients with CR and minimal residual disease (MRD)-negative CRi, respectively. Tafasitamab produced rapid B-cell/blast depletion in 21 of 22 patients within 1 to 2 weeks of first administration. Tafasitamab was well tolerated, with the most frequent adverse events being infusion-related reactions (59.1%) and fatigue (40.9%). Grade 3 to 4 febrile neutropenia (22.7%) was the most common hematologic adverse event. CONCLUSIONS Tafasitamab monotherapy was associated with clinical activity in a subset of patients with R/R B-ALL, including short-lasting CR and MRD-negative CRi. Given its favorable tolerability profile, further development of tafasitamab in chemoimmunotherapy combinations and MRD settings should be explored.
Collapse
Affiliation(s)
- Rebecca B. Klisovic
- Department of Hematology and Medical OncologyEmory School of MedicineAtlantaGeorgia
| | - Wing H. Leung
- Department of Paediatrics and Adolescent MedicineThe University of Hong KongPokfulamHong Kong
| | - Wolfram Brugger
- MorphoSys AGPlaneggGermany
- Present address:
Autolus TherapeuticsLondonUK
| | | | | | | | | |
Collapse
|
46
|
Zinzani PL, Rodgers T, Marino D, Frezzato M, Barbui AM, Castellino C, Meli E, Fowler NH, Salles G, Feinberg B, Kurukulasuriya NC, Tillmanns S, Parche S, Dey D, Fingerle-Rowson G, Ambarkhane S, Winderlich M, Nowakowski GS. RE-MIND: Comparing Tafasitamab + Lenalidomide (L-MIND) with a Real-world Lenalidomide Monotherapy Cohort in Relapsed or Refractory Diffuse Large B-cell Lymphoma. Clin Cancer Res 2021; 27:6124-6134. [PMID: 34433649 PMCID: PMC9414300 DOI: 10.1158/1078-0432.ccr-21-1471] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/03/2021] [Accepted: 08/19/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE Tafasitamab, an Fc-modified, humanized, anti-CD19 monoclonal antibody, in combination with lenalidomide, demonstrated efficacy in transplant-ineligible patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL), in the single-arm, phase II L-MIND study (NCT02399085). RE-MIND, a retrospective observational study, generated a historic control for L-MIND to delineate the contribution of tafasitamab to the efficacy of the combination. PATIENTS AND METHODS Data were retrospectively collected from patients with R/R DLBCL treated with lenalidomide monotherapy for comparison with tafasitamab + lenalidomide-treated patients (L-MIND). Key eligibility criteria were aligned with L-MIND. Estimated propensity score-based Nearest Neighbor 1:1 Matching methodology balanced the cohorts for nine prespecified prognostic baseline covariates. The primary endpoint was investigator-assessed best overall response rate (ORR). Secondary endpoints included complete response (CR) rate, progression-free survival (PFS), and overall survival (OS). RESULTS Data from 490 patients going through lenalidomide monotherapy were collected; 140 qualified for matching with the L-MIND cohort. The primary analysis included 76 patients from each cohort who received a lenalidomide starting dose of 25 mg/day. Cohort baseline covariates were comparable. A significantly better ORR of 67.1% (95% confidence interval, 55.4-77.5) was observed for the combination therapy versus 34.2% (23.7-46.0) for lenalidomide monotherapy [odds ratio, 3.89 (1.90-8.14); P < 0.0001]. Higher CR rates were achieved with combination therapy compared with lenalidomide monotherapy [39.5% (28.4-51.4) vs. 13.2% (6.5-22.9)]. Survival endpoints favored combination therapy. Lenalidomide monotherapy outcomes were similar to previously published data. CONCLUSIONS RE-MIND enabled the estimation of the additional treatment effect achieved by combining tafasitamab with lenalidomide in patients with R/R DLBCL.
Collapse
Affiliation(s)
- Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Thomas Rodgers
- University of Rochester Medical Center, Rochester, New York
| | - Dario Marino
- Oncology 1 Unit, Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | | | | | - Erika Meli
- Dipartimento Ematologia ed Oncologia, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Nathan H. Fowler
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gilles Salles
- Hématologie, Hospices Civils de Lyon and Université de Lyon, Lyon, France
| | | | | | | | | | | | | | | | | | - Grzegorz S. Nowakowski
- Division of Hematology, Mayo Clinic, Rochester, Minnesota.,Corresponding Author: Grzegorz S. Nowakowski, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Phone: 507-405-0312, E-mail:
| |
Collapse
|
47
|
Zinzani PL, Minotti G. Anti-CD19 monoclonal antibodies for the treatment of relapsed or refractory B-cell malignancies: a narrative review with focus on diffuse large B-cell lymphoma. J Cancer Res Clin Oncol 2021; 148:177-190. [PMID: 34741682 PMCID: PMC8752543 DOI: 10.1007/s00432-021-03833-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE CD19 is a cell surface protein that is found on both healthy and malignant B cells. Accordingly, it has become an important target for novel treatments for non-Hodgkin lymphomas and B-cell leukaemia. Three anti-CD19 monoclonal antibodies with distinct mechanisms of action have been developed for the treatment of B-cell malignancies. METHODS We reviewed the preclinical and clinical data on the development of the newly approved anti-CD19 monoclonal antibodies blinatumomab, tafasitamab and loncastuximab tesirine, and consider their place in the treatment of relapsed or refractory B-cell malignancies. RESULTS Blinatumomab is a bispecific T-cell engager that binds to both CD19 on B cells and CD3 on T cells, facilitating antibody-dependent cytotoxicity. Blinatumomab significantly prolongs overall survival in patients with relapsed or refractory B-cell acute lymphoblastic leukaemia, although cytokine release syndrome and severe neurotoxicity may necessitate discontinuation. Tafasitamab, which has modified anti-CD19 Fab and Fc regions, has significantly enhanced affinity for both CD19 and effector cell receptors compared with unmodified anti-CD19. In L-MIND, tafasitamab plus lenalidomide provided an overall response rate (ORR) of 57.5% in patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) in patients non-transplant eligible. Loncastuximab tesirine is an antibody-drug conjugate that has been studied as monotherapy and in combination with ibrutinib in 3L + relapsed or refractory DLBCL. The ORR was 48.3% in a phase II trial of loncastuximab tesirine. The optimal place of anti-CD19 monoclonal antibodies in therapy has yet to be determined, but the prospect of improved outcomes for at least some patients with treatment-resistant B-cell malignancies appears likely, particularly in those with limited therapeutic options and poor prognosis.
Collapse
Affiliation(s)
- Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Via Massarenti 9, 40138, Bologna, Italy. .,Department of Specialist, Diagnostic and Experimental Medicine, University of Bologna, Bologna, Italy.
| | - Giorgio Minotti
- Department of Medicine, Center for Integrated Research and Unit of Drug Science, University Campus Bio-Medico, Rome, Italy
| |
Collapse
|
48
|
Tilch MK, Robak T, Ghiggi C, Wuff E, Herold S, Theobald M, Hess G. Safety of the Anti-CD19 antibody Tafasitamab in Long Term Responders from A Phase II Trial for Relapsed Lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:270-275. [PMID: 34776401 DOI: 10.1016/j.clml.2021.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/22/2021] [Accepted: 10/09/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Information about the long-term tolerability of tafasitamab is still limited. METHODS 5 of 92 patients treated within a phase IIa study of single-agent tafasitamab in relapsed or refractory B NHL were followed for up to five years or longer for long-term tolerability. RESULTS Treatment was very well tolerated in an outpatient setting with no hospitalizations needed and mild and tolerable adverse events that occurred mostly within the first two years of treatment. CONCLUSIONS Given the excellent tolerability and efficacy of tafasitamab this agent can be used to induce remission in relapsed or refractory lymphoma either alone or in combination with chemotherapy.
Collapse
Affiliation(s)
- Marie-Kristin Tilch
- Department of Hematology, Oncology and Pneumology & University Cancer Center, University Medical Center of the Johannes Gutenberg-University, Mainz, Rhineland-Palatinate, Germany
| | - Tadeusz Robak
- Department of Hematology, Medical University of Lodz, Lodz, Poland
| | - Chiara Ghiggi
- Division of Hematology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Elke Wuff
- Department of Hematology, Oncology and Pneumology & University Cancer Center, University Medical Center of the Johannes Gutenberg-University, Mainz, Rhineland-Palatinate, Germany
| | - Stephanie Herold
- Department of Hematology, Oncology and Pneumology & University Cancer Center, University Medical Center of the Johannes Gutenberg-University, Mainz, Rhineland-Palatinate, Germany
| | - Matthias Theobald
- Department of Hematology, Oncology and Pneumology & University Cancer Center, University Medical Center of the Johannes Gutenberg-University, Mainz, Rhineland-Palatinate, Germany
| | - Georg Hess
- Department of Hematology, Oncology and Pneumology & University Cancer Center, University Medical Center of the Johannes Gutenberg-University, Mainz, Rhineland-Palatinate, Germany.
| |
Collapse
|
49
|
Liu Z, Xu W, Chen Z, Fu W, Zhan W, Gao Y, Zhou J, Zhou Y, Wu J, Wang Q, Zhang X, Hao A, Wu W, Zhang Q, Li Y, Fan K, Chen R, Jiang Q, Mayer CT, Schoofs T, Xie Y, Jiang S, Wen Y, Yuan Z, Wang K, Lu L, Sun L, Wang Q. An ultrapotent pan-β-coronavirus lineage B (β-CoV-B) neutralizing antibody locks the receptor-binding domain in closed conformation by targeting its conserved epitope. Protein Cell 2021; 13:655-675. [PMID: 34554412 PMCID: PMC8458794 DOI: 10.1007/s13238-021-00871-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/02/2021] [Indexed: 01/09/2023] Open
Abstract
New threats posed by the emerging circulating variants of SARS-CoV-2 highlight the need to find conserved neutralizing epitopes for therapeutic antibodies and efficient vaccine design. Here, we identified a receptor-binding domain (RBD)-binding antibody, XG014, which potently neutralizes β-coronavirus lineage B (β-CoV-B), including SARS-CoV-2, its circulating variants, SARS-CoV and bat SARSr-CoV WIV1. Interestingly, antibody family members competing with XG014 binding show reduced levels of cross-reactivity and induce antibody-dependent SARS-CoV-2 spike (S) protein-mediated cell-cell fusion, suggesting a unique mode of recognition by XG014. Structural analyses reveal that XG014 recognizes a conserved epitope outside the ACE2 binding site and completely locks RBD in the non-functional "down" conformation, while its family member XG005 directly competes with ACE2 binding and position the RBD "up". Single administration of XG014 is effective in protection against and therapy of SARS-CoV-2 infection in vivo. Our findings suggest the potential to develop XG014 as pan-β-CoV-B therapeutics and the importance of the XG014 conserved antigenic epitope for designing broadly protective vaccines against β-CoV-B and newly emerging SARS-CoV-2 variants of concern.
Collapse
Affiliation(s)
- Zezhong Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wei Xu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhenguo Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wangjun Fu
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wuqiang Zhan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yidan Gao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jie Zhou
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yunjiao Zhou
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jianbo Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qian Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiang Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Aihua Hao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wei Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qianqian Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Kaiyue Fan
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ruihong Chen
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiaochu Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Christian T Mayer
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yumei Wen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Kang Wang
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Lei Sun
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Qiao Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences; Shanghai Institute of Infectious Disease and Biosecurity; the Fifth People's Hospital of Shanghai; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology); Institutes of Biomedical Sciences; Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
50
|
Staber PB, Jurczak W, Greil R, Vucinic V, Middeke JM, Montillo M, Munir T, Neumeister P, Schetelig J, Stilgenbauer S, Striebel F, Dirnberger-Hertweck M, Weirather J, Brugger W, Kelemen P, Wendtner CM, Woyach JA. Tafasitamab combined with idelalisib or venetoclax in patients with CLL previously treated with a BTK inhibitor. Leuk Lymphoma 2021; 62:3440-3451. [PMID: 34414843 DOI: 10.1080/10428194.2021.1964020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Patients with relapsed/refractory chronic lymphocytic leukemia (R/R CLL) whose treatment failed with a Bruton's tyrosine kinase inhibitor have poor outcomes. We investigated tafasitamab plus idelalisib (cohort A) or venetoclax (cohort B) in this patient population in a phase II study (NCT02639910). In total, 24 patients were enrolled (cohort A: n = 11, median time on study, 7.4 months; cohort B: n = 13, median time on study, 15.6 months). The most common treatment-emergent adverse event (TEAE) in cohort A was anemia (63.6%) and in cohort B was infusion-related reaction (53.8%). The most common severe TEAE was neutropenia (cohort A: 45.5%; cohort B: 46.2%). The best overall response rate was 90.9% (cohort A) and 76.9% (cohort B). Undetectable minimal residual disease in peripheral blood was achieved in 2/8 patients (cohort A) and 6/7 patients (cohort B). Overall, these results suggest that anti-CD19 antibody-based combinations may be important in the treatment of patients with CLL.
Collapse
Affiliation(s)
- Philipp Bernhard Staber
- Clinical Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Wojciech Jurczak
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, Poland
| | - Richard Greil
- Laboratory for Immunological and Molecular Cancer Research, Salzburg Cancer Research Institute, Salzburg, Austria
| | - Vladan Vucinic
- Department of Hematology and Internal Oncology, University Hospital Leipzig AöR, Leipzig, Germany
| | - Jan Moritz Middeke
- Medical Clinic 1, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Marco Montillo
- Hematology Department, Niguarda Cancer Center, Niguarda Hospital, Milan, Italy
| | - Talha Munir
- Clinical Hematology, The Leeds Teaching Hospital, Leeds, UK
| | - Peter Neumeister
- Clinical Department of Hematology, Medical University of Graz, Graz, Austria
| | - Johannes Schetelig
- Medical Clinic 1, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | | | | | | | | | | | | | - Clemens-Martin Wendtner
- Department of Hematology, Oncology, Immunology, Palliative Medicine, Infectious Diseases, and Tropical Medicine, Munich Clinic Schwabing, Munich, Germany
| | - Jennifer Ann Woyach
- Division of Hematology, Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|