1
|
Kim WS, Kim TM, Cho SG, Jarque I, Iskierka-Jażdżewska E, Poon LM, Prince HM, Zhang H, Cao J, Zhang M, Tessoulin B, Oh SY, Lim F, Carpio C, Tan TD, Ayyappan S, Gutierrez A, Cai J, Ufkin M, Shariff S, Brouwer-Visser J, Chaudhry A, Mohamed H, Ambati S, Walewski J. Odronextamab monotherapy in patients with relapsed/refractory diffuse large B cell lymphoma: primary efficacy and safety analysis in phase 2 ELM-2 trial. NATURE CANCER 2025; 6:528-539. [PMID: 40097657 PMCID: PMC12003196 DOI: 10.1038/s43018-025-00921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/23/2025] [Indexed: 03/19/2025]
Abstract
The phase 2, multicohort, ongoing ELM-2 study evaluates odronextamab, a CD20×CD3 bispecific antibody, in patients with relapsed/refractory (R/R) B cell non-Hodgkin lymphoma after ≥2 lines of therapy. Here primary analysis of the diffuse large B cell lymphoma (DLBCL) cohort is reported. Patients received intravenous odronextamab in 21-day cycles until progression or unacceptable toxicity, with cycle 1 step-up dosing to mitigate cytokine release syndrome (CRS) risk. The primary endpoint was objective response rate (ORR). Secondary endpoints included complete response (CR) rate, duration of response, progression-free survival (PFS) and overall survival. A total of 127 patients were enrolled. At the 29.9-month efficacy follow-up, the ORR was 52.0% and CR rate was 31.5%. Median durations of response and CR were 10.2 and 17.9 months, respectively. Undetectable minimal residual disease at cycle 4 day 15 was associated with PFS benefit. With a step-up of 0.7 to 4 to 20 mg (n = 60), CRS was the most common treatment-emergent adverse event (53.3% (grade ≥3, 1.7%)). No immune effector cell-associated neurotoxicity syndrome was reported. Infections were reported in 82/127 (64.6%) patients (grade ≥3, 38.6%; coronavirus disease 2019, 18.1% (grade ≥3, 12.6%)). In conclusion, odronextamab showed encouraging efficacy in heavily pretreated R/R DLBCL and generally manageable safety with supportive care. Clinical trial registration: NCT03888105 .
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Female
- Male
- Middle Aged
- Aged
- Adult
- Aged, 80 and over
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Antibodies, Bispecific/adverse effects
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/administration & dosage
- Treatment Outcome
- Progression-Free Survival
- Young Adult
Collapse
Affiliation(s)
- Won Seog Kim
- Sungkyunkwan University School of Medicine, Samsung Medical Center Division of Hematology-Oncology, Seoul, South Korea.
| | - Tae Min Kim
- Seoul National University Hospital, Seoul National University Cancer Research Institute, Seoul, South Korea
| | - Seok-Goo Cho
- Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Isidro Jarque
- Hematology Department, Hospital Universitari i Politècnic La Fe, Centro de Investigación Biomédica en Red de Cáncer-CIBERONC, Valencia, Spain
| | | | - Li Mei Poon
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore, Singapore
| | - H Miles Prince
- Epworth HealthCare and University of Melbourne, Melbourne, Victoria, Australia
| | - Huilai Zhang
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Junning Cao
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Mingzhi Zhang
- Department of Oncology, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Benoît Tessoulin
- Hematology Department, Nantes University Hospital, Nantes, France
| | | | | | - Cecilia Carpio
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Tran-Der Tan
- Hematology and Medical Oncology, Koo Foundation Sun Yat Sen Cancer Center, Taipei City, Taiwan
| | | | - Antonio Gutierrez
- Department of Hematology, Hospital Universitario Son Espases, IdISBa, Palma, Spain
| | - Jingxian Cai
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | | | | | | | | | - Jan Walewski
- Narodowy Instytut Onkologii im. Marii Skłodowskiej-Curie Państwowy Instytut Badawczy, Warszawa, Poland
| |
Collapse
|
2
|
Okon A, Yang J, Giancola JB, Molina OJ, Sayers J, Cheah KM, Li Y, Strieter ER, Raines RT. Facile Access to Branched Multispecific Proteins. Bioconjug Chem 2024; 35:954-962. [PMID: 38879814 PMCID: PMC11254548 DOI: 10.1021/acs.bioconjchem.4c00162] [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] [Indexed: 07/18/2024]
Abstract
Approaches that leverage orthogonal chemical reactions to generate protein-protein conjugates have expanded access to bespoke chimeras. Although the literature is replete with examples of the semisynthesis of bispecific proteins, few methods exist for the semisynthesis of protein conjugates of higher complexity (i.e., greater than two-protein fusions). The recent emergence of trispecific cell engagers for immune cell redirection therapies necessitates the development of chemical methods for the construction of trispecific proteins that would otherwise be inaccessible via natural protein synthesis. Here, we demonstrate that 3-bromo-5-methylene pyrrolone (3Br-5MP) can be used to effect the facile chemical synthesis of trispecific peptides and proteins with exquisite control over the addition of each monomer. The multimeric complexes maintain epitope functionality both in human cells and upon immobilization. We anticipate that facile access to trispecific proteins using this 3Br-5MP will have broad utility in basic science research and will quicken the pace of research to establish novel, multimeric immune cell redirection therapies.
Collapse
Affiliation(s)
- Aniekan Okon
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jinyi Yang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - JoLynn B. Giancola
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Oscar J. Molina
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jessica Sayers
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Keith M. Cheah
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Yanfeng Li
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Eric R. Strieter
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Ronald T. Raines
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Fowler NH, Chavez JC, Riedell PA. Moving T-Cell Therapies into the Standard of Care for Patients with Relapsed or Refractory Follicular Lymphoma: A Review. Target Oncol 2024; 19:495-510. [PMID: 38896212 PMCID: PMC11271334 DOI: 10.1007/s11523-024-01070-z] [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] [Accepted: 05/06/2024] [Indexed: 06/21/2024]
Abstract
Patients with follicular lymphoma, an indolent form of non-Hodgkin lymphoma, typically experience multiple relapses over their disease course. Periods of remission become progressively shorter with worse clinical outcomes after each subsequent line of therapy. Currently, no clear standard of care/preferred treatment approach exists for patients with relapsed or refractory follicular lymphoma. As novel agents continue to emerge for treatment in the third-line setting, guidance is needed for selecting the most appropriate therapy for each patient. Several classes of targeted therapeutic agents, including monoclonal antibodies, phosphoinositide 3-kinase inhibitors, enhancer of zeste homolog 2 inhibitors, chimeric antigen receptor (CAR) T-cell therapies, and bispecific antibodies, have been approved by regulatory authorities based on clinical benefit in patients with relapsed or refractory follicular lymphoma. Additionally, antibody-drug conjugates and other immunocellular therapies are being evaluated in this setting. Effective integration of CAR-T cell therapy into the treatment paradigm after two or more prior therapies requires appropriate patient selection based on transformation status following a rebiopsy; a risk evaluation based on age, fitness, and remission length; and eligibility for CAR-T cell therapy. Consideration of important logistical factors (e.g., proximity to the treatment center and caregiver support during key periods of CAR-T cell therapy) is also critical. Overall, an individualized treatment plan that considers patient-related factors (e.g., age, disease status, tumor burden, comorbidities) and prior treatment types is recommended for patients with relapsed or refractory follicular lymphoma. Future analyses of real-world data and a better understanding of mechanisms of relapse are needed to further refine patient selection and identify optimal sequencing of therapies in this setting.
Collapse
Affiliation(s)
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Peter A Riedell
- David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, USA
| |
Collapse
|
4
|
Gong N, Han X, Xue L, Billingsley MM, Huang X, El-Mayta R, Qin J, Sheppard NC, June CH, Mitchell MJ. Small-molecule-mediated control of the anti-tumour activity and off-tumour toxicity of a supramolecular bispecific T cell engager. Nat Biomed Eng 2024; 8:513-528. [PMID: 38378820 DOI: 10.1038/s41551-023-01147-6] [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: 01/22/2023] [Accepted: 10/24/2023] [Indexed: 02/22/2024]
Abstract
The broader clinical use of bispecific T cell engagers for inducing anti-tumour toxicity is hindered by their on-target off-tumour toxicity and the associated neurotoxicity and cytokine-release syndrome. Here we show that the off-tumour toxicity of a supramolecular bispecific T cell engager binding to the T cell co-receptor CD3 and to the human epidermal growth factor receptor 2 on breast tumour cells can be halted by disengaging the T cells from the tumour cells via the infusion of the small-molecule drug amantadine, which disassembles the supramolecular aggregate. In mice bearing human epidermal growth factor receptor 2-expressing tumours and with a human immune system, high intravenous doses of such a 'switchable T cell nanoengager' elicited strong tumour-specific adaptive immune responses that prevented tumour relapse, while the infusion of amantadine restricted off-tumour toxicity, cytokine-release syndrome and neurotoxicity. Supramolecular chemistry may be further leveraged to control the anti-tumour activity and off-tumour toxicity of bispecific antibodies.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Xisha Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingya Qin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Neil C Sheppard
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Lin T, Liu D, Guan Z, Zhao X, Li S, Wang X, Hou R, Zheng J, Cao J, Shi M. CRISPR screens in mechanism and target discovery for AML. Heliyon 2024; 10:e29382. [PMID: 38660246 PMCID: PMC11040068 DOI: 10.1016/j.heliyon.2024.e29382] [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: 07/05/2023] [Revised: 03/20/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024] Open
Abstract
CRISPR-based screens have discovered novel functional genes involving in diverse tumor biology and elucidated the mechanisms of the cancer pathological states. Recently, with its randomness and unbiasedness, CRISPR screens have been used to discover effector genes with previously unknown roles for AML. Those novel targets are related to AML survival resembled cellular pathways mediating epigenetics, synthetic lethality, transcriptional regulation, mitochondrial and energy metabolism. Other genes that are crucial for pharmaceutical targeting and drug resistance have also been identified. With the rapid development of novel strategies, such as barcodes and multiplexed mosaic CRISPR perturbation, more potential therapeutic targets and mechanism in AML will be discovered. In this review, we present an overview of recent progresses in the development of CRISPR-based screens for the mechanism and target identification in AML and discuss the challenges and possible solutions in this rapidly growing field.
Collapse
Affiliation(s)
- Tian Lin
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Dan Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Zhangchun Guan
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Xuan Zhao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Sijin Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Xu Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Rui Hou
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- College of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Jiang Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| |
Collapse
|
6
|
Wang L, Leach V, Muthusamy N, Byrd J, Long M. A CD3 humanized mouse model unmasked unique features of T-cell responses to bispecific antibody treatment. Blood Adv 2024; 8:470-481. [PMID: 37871327 PMCID: PMC10837186 DOI: 10.1182/bloodadvances.2023010971] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/04/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
ABSTRACT T-cell bispecific antibodies (T-BsAbs) such as blinatumomab hold great promise for cancer immunotherapy. A better understanding of the in vivo immune response induced by T-BsAbs is crucial to improving their efficacy and safety profile. However, such efforts are hindered by the limitations of current preclinical models. To address this, we developed a syngeneic murine model with humanized CD3 and target antigen (CD20). This model enables the development of disseminated leukemia with a high tumor burden, which mirrors clinical findings in human patients with relapsed/refractory acute lymphoblastic leukemia. Treatment of this model with T-BsAbs results in cytokine release syndrome, with cytokine profiles and levels reflecting observations made in human patients. This model also faithfully recapitulates the dynamics of T-cell activation seen in human patients, including the temporary disappearance of T cells from the bloodstream. During this phase, T cells are sequestered in secondary lymphoid organs and undergo activation. Clinical correlative studies that rely primarily on peripheral blood samples are likely to overlook this critical activation stage, leading to a substantial underestimation of the extent of T-cell activation. Furthermore, we demonstrate that surface expression of the T-BsAb target antigen by leukemia cells triggers a swift immune response, promoting their own rejection. Humanizing the target antigen in the recipient mice is crucial to facilitate tolerance induction and successful establishment of high tumor burden. Our findings underscore the importance of meticulously optimized syngeneic murine models for investigating T-BsAb-induced immune responses and for translational research aimed at improving efficacy and safety.
Collapse
Affiliation(s)
- Lingling Wang
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Vincent Leach
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - John Byrd
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Meixiao Long
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| |
Collapse
|
7
|
Weddell J. Mechanistically modeling peripheral cytokine dynamics following bispecific dosing in solid tumors. CPT Pharmacometrics Syst Pharmacol 2023; 12:1726-1737. [PMID: 36710368 PMCID: PMC10681545 DOI: 10.1002/psp4.12928] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
Bispecific antibodies exhibit proven clinical benefit, and many bispecifics are currently in clinical development for oncology. Cytokine release syndrome (CRS) is a common clinical adverse effect observed following CD3-based bispecific dosing. However, the pathophysiology of CRS is not fully understood, and no computational model mechanistically describing clinical cytokine dynamics following bispecific dosing in solid tumors exists. Here, a quantitative systems pharmacology (QSP) model describing peripheral clinical cytokine dynamics following bispecific dosing in solid tumors is presented. Using tebentafusp as a case study, a CD3-bispecific approved for uveal melanoma, the model successfully captures the dynamics of five cytokines. The QSP model was shown to predict observed phenomena, such as cytokine maximum concentration suppression using step-up dosing regimens and the importance of on-target off-tumor binding toward CRS and toxicity. Furthermore, the QSP model provides rationale for these biological phenomena based on dynamics of immune cell activation and desensitization in tumors and healthy tissues. Overall, the QSP model structure presented here serves as a basis to infer cytokine dynamics for other CD3-based bispecifics or tumor types by altering model parameters to capture the scenario of interest, supporting applications including dose selection, candidate nomination, and disease area selection.
Collapse
Affiliation(s)
- Jared Weddell
- Clinical Pharmacology and Exploratory DevelopmentAstellas Pharma Global Development Inc.NorthbrookIllinoisUSA
| |
Collapse
|
8
|
Balendran S, Tam C, Ku M. T-Cell Engaging Antibodies in Diffuse Large B Cell Lymphoma-An Update. J Clin Med 2023; 12:6737. [PMID: 37959202 PMCID: PMC10647650 DOI: 10.3390/jcm12216737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Novel cellular immunotherapies such as T-cell engaging antibodies (TCEAbs) are changing the landscape of treatment for diffuse large B cell lymphoma (DLBCL), especially in the relapsed/refractory (R/R) setting. TCEAbs harness the power of the host immune system to induce killing of tumor cells by binding to both the tumor antigen and the T-cell receptor. Since the approval of blinatumomab for R/R acute lymphoblastic leukemia, there has been significant development in novel TCEAbs. Many of these novel TCEAbs have shown promising effectiveness in R/R DLBCL, with favorable response rates including complete remissions, even in heavily pretreated patients. There are unique therapy-related toxicities with TCEAbs, namely cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity (ICANS), and it is important to both recognize and manage these side effects appropriately. This review examines the development and mechanism of action of these TCEAbs, and the available published data from clinical trials. Their role in the treatment of DLBCL, the management of therapy-related adverse events, and the mechanisms of resistance will also be discussed.
Collapse
Affiliation(s)
| | | | - Matthew Ku
- St. Vincent’s Hospital, Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
9
|
Khosla AA, Jatwani K, Singh R, Reddy A, Jaiyesimi I, Desai A. Bispecific Antibodies in Lung Cancer: A State-of-the-Art Review. Pharmaceuticals (Basel) 2023; 16:1461. [PMID: 37895932 PMCID: PMC10609957 DOI: 10.3390/ph16101461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/25/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Bispecific antibodies have emerged as a promising class of therapeutics in the field of oncology, offering an innovative approach to target cancer cells while sparing healthy tissues. These antibodies are designed to bind two different antigens, enabling them to bridge immune cells with cancer cells, resulting in enhanced tumor cell killing and improved treatment responses. This review article summarizes the current landscape of bispecific antibodies in lung cancer, including their mechanisms of action, clinical development, and potential applications in other solid tumor malignancies. Additionally, the challenges and opportunities associated with their use in the clinic are discussed, along with future directions for research and development in this exciting area of cancer immunotherapy.
Collapse
Affiliation(s)
- Atulya Aman Khosla
- Division of Internal Medicine, William Beaumont University Hospital, Royal Oak, MI 48073, USA;
| | - Karan Jatwani
- Division of Hematology-Oncology, Roswell Park Cancer Center, Buffalo, NY 14203, USA
| | - Rohit Singh
- Division of Hematology-Oncology, University of Vermont, Burlington, VT 05405, USA
| | - Aswanth Reddy
- Division of Hematology-Oncology, Mercy Clinic, Fort Smith, AR 72903, USA
| | - Ishmael Jaiyesimi
- Division of Hematology-Oncology, William Beaumont University Hospital, Royal Oak, MI 48073, USA
| | - Aakash Desai
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
10
|
Chen CJJ, Choi MY, Heyman BM. Targeted Therapy in Follicular Lymphoma: Towards a Chemotherapy-Free Approach. Cancers (Basel) 2023; 15:4483. [PMID: 37760453 PMCID: PMC10526830 DOI: 10.3390/cancers15184483] [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: 06/12/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The treatment of follicular lymphoma (FL) has previously centered on chemoimmunotherapy, which can be disadvantageous due to patient intolerance, cumulative toxicities, and disease refractoriness. Targeted therapies can produce deep responses and improve progression-free and overall survival with more tolerable adverse event profiles. METHODS We summarize the current literature and key clinical trials regarding targeted therapies in follicular lymphoma both in the front-line and in the relapsed-refractory setting. RESULTS Targeted therapies studied in FL include immune modulators, anti-CD20 antibodies, Bruton's tyrosine kinase (BTK) inhibitors, enhancers of zeste homolog 2 (EZH2) inhibitors, phosphoinositide 3-kinase (PI3K) inhibitors, and B-cell lymphoma 2 (BCL-2) inhibitors. Chimeric antigen receptor (CAR-T) therapy and bispecific T-cell engager (BiTE) therapies also show promise in monotherapy and in combination with targeted therapies. These therapies exhibit high overall response rates and substantial progression-free survival and overall survival, even in high-risk patients or patients previously refractory to chemotherapy or rituximab. Adverse events vary substantially but are generally manageable and compare favorably to the cumulative toxicities of chemotherapy. CONCLUSION Targeted therapies represent a paradigm shift in the treatment of FL. Further studies are needed to directly compare these targeted therapies and their combinations, as well as to investigate biomarkers predictive of response.
Collapse
Affiliation(s)
- Chung-Jiah J. Chen
- Department of Medicine, Division of Hematology-Oncology, UC San Diego Health, La Jolla, CA 92037, USA; (C.-J.J.C.); (M.Y.C.)
| | - Michael Y. Choi
- Department of Medicine, Division of Hematology-Oncology, UC San Diego Health, La Jolla, CA 92037, USA; (C.-J.J.C.); (M.Y.C.)
| | - Benjamin M. Heyman
- Department of Medicine, Division of Regenerative Medicine, UC San Diego Health, La Jolla, CA 92037, USA
| |
Collapse
|
11
|
Noori M, Yazdanpanah N, Rezaei N. Safety and efficacy of T-cell-redirecting bispecific antibodies for patients with multiple myeloma: a systematic review and meta-analysis. Cancer Cell Int 2023; 23:193. [PMID: 37670301 PMCID: PMC10478206 DOI: 10.1186/s12935-023-03045-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/28/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND In recent years, several bispecific antibodies (BsAbs) have been introduced that revolutionized the treatment approach for patients with multiple myeloma (MM). In the present study, we sought for conducting a systematic review and meta-analysis with the aim of evaluating the safety and efficacy of BsAbs in MM patients. METHODS PubMed, Scopus, Web of Science, and Embase databases were systematically searched on June 10, 2022. Two steps of title/abstract and full-text screening were performed for selecting the relevant articles. The primary endpoint was considered to evaluate the safety of BsAbs by examining the rate of hematologic and non-hematologic adverse effects (AEs). The secondary outcome was set at the efficacy of BsAbs through pooling objective response rate (ORR), (stringent) complete response (sCR/CR), very good partial response (VGPR), and partial response (PR). RESULTS Eleven publications with a total of nine evaluable BsAbs were included for qualitative and quantitative data synthesis. Hematologic AEs were more common among patients than non-hematologic events, with the most frequent events being anemia (41.4%), neutropenia (36.4%), and thrombocytopenia (26.3%). The most common non-hematological AE was infection, which occurred in 39.9% of patients, followed by dysgeusia (28.3%), fatigue (26.5%), and diarrhea (25.8%). Besides, 8.1% of patients experienced immune effector cell-associated neurotoxicity syndrome and neurotoxicity occurred in 5.1% of them. Moreover, 59.8% of patients experienced cytokine release syndrome. The pooled rate of deaths attributable to BsAbs was estimated at 0.1%. In terms of efficacy measures, the ORR was achieved in 62.6% of MM patients, and the pooled rates of sCR/CR, VGPR, and PR were 22.7%, 23.0%, and 12.1%, respectively. CONCLUSIONS In an era with several emerging promising treatments for MM, BsAbs have achieved a high ORR and tolerable AEs in heavily pretreated patients. However, there is still room for developing BsAbs with a lower rate of AEs and capable of bypassing tumor evasion mechanisms.
Collapse
Affiliation(s)
- Maryam Noori
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Student Research Committee, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Farhangnia P, Ghomi SM, Akbarpour M, Delbandi AA. Bispecific antibodies targeting CTLA-4: game-changer troopers in cancer immunotherapy. Front Immunol 2023; 14:1155778. [PMID: 37441075 PMCID: PMC10333476 DOI: 10.3389/fimmu.2023.1155778] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
Antibody-based cancer immunotherapy has become a powerful asset in the arsenal against malignancies. In this regard, bispecific antibodies (BsAbs) are a ground-breaking novel approach in the therapy of cancers. Recently, BsAbs have represented a significant advancement in improving clinical outcomes. BsAbs are designed to target two different antigens specifically. Over a hundred various BsAb forms currently exist, and more are constantly being manufactured. An antagonistic regulator of T cell activation is cytotoxic T lymphocyte-associated protein 4 (CTLA-4) or CD152, a second counter-receptor for the B7 family of co-stimulatory molecules was introduced in 1996 by Professor James P. Allison and colleagues. Contrary to the explosive success of dual immune checkpoint blockade for treating cancers, a major hurdle still yet persist is that immune-related adverse events (irAEs) observed by combining immune checkpoint inhibitors (ICIs) or monoclonal antibodies such as ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1). A promising strategy to overcome this hurdle is using BsAbs. This article will summarize BsAbs targeting CTLA-4, their applications in cancer immunotherapy, and relevant clinical trial advances. We will also discuss the pre-clinical rationale for using these BsAbs, and provide the current landscape of the field.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Shamim Mollazadeh Ghomi
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahzad Akbarpour
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Advanced Cellular Therapeutics Facility (ACTF), Hematopoietic Cellular Therapy Program, Section of Hematology & Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, United States
| | - Ali-Akbar Delbandi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Gambles MT, Yang J, Kopeček J. Multi-targeted immunotherapeutics to treat B cell malignancies. J Control Release 2023; 358:232-258. [PMID: 37121515 PMCID: PMC10330463 DOI: 10.1016/j.jconrel.2023.04.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
The concept of multi-targeted immunotherapeutic systems has propelled the field of cancer immunotherapy into an exciting new era. Multi-effector molecules can be designed to engage with, and alter, the patient's immune system in a plethora of ways. The outcomes can vary from effector cell recruitment and activation upon recognition of a cancer cell, to a multipronged immune checkpoint blockade strategy disallowing evasion of the cancer cells by immune cells, or to direct cancer cell death upon engaging multiple cell surface receptors simultaneously. Here, we review the field of multi-specific immunotherapeutics implemented to treat B cell malignancies. The mechanistically diverse strategies are outlined and discussed; common B cell receptor antigen targeting strategies are outlined and summarized; and the challenges of the field are presented along with optimistic insights for the future.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
14
|
Leveraging Natural Killer Cell Innate Immunity against Hematologic Malignancies: From Stem Cell Transplant to Adoptive Transfer and Beyond. Int J Mol Sci 2022; 24:ijms24010204. [PMID: 36613644 PMCID: PMC9820370 DOI: 10.3390/ijms24010204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Numerous recent advancements in T-cell based immunotherapies have revolutionized the treatment of hematologic malignancies. In the race towards the first approved allogeneic cellular therapy product, there is growing interest in utilizing natural killer (NK) cells as a platform for off-the-shelf cellular therapies due to their scalable manufacturing potential, potent anti-tumor efficacy, and superior safety profile. Allogeneic NK cell therapies are now being actively explored in the setting of hematopoietic stem cell transplantation and adoptive transfer. Increasingly sophisticated gene editing techniques have permitted the engineering of chimeric antigen receptors, ectopic cytokine expression, and tumor recognition signals to improve the overall cytotoxicity of NK cell therapies. Furthermore, the enhancement of antibody-dependent cellular cytotoxicity has been achieved through the use of NK cell engagers and combination regimens with monoclonal antibodies that act synergistically with CD16-expressing NK cells. Finally, a greater understanding of NK cell biology and the mechanisms of resistance have allowed the preclinical development of NK checkpoint blockade and methods to modulate the tumor microenvironment, which have been evaluated in early phase trials. This review will discuss the recent clinical advancements in NK cell therapies in hematologic malignancies as well as promising avenues of future research.
Collapse
|
15
|
Welty NE, Gill SI. Cancer Immunotherapy Beyond Checkpoint Blockade: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022; 4:563-578. [PMID: 36636439 PMCID: PMC9830230 DOI: 10.1016/j.jaccao.2022.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 12/24/2022] Open
Abstract
Avoidance of immune destruction is recognized as one of the hallmarks of cancer development. Although first predicted as a potential antitumor treatment modality more than 50 years ago, the widespread clinical use of cancer immunotherapies has only recently become a reality. Cancer immunotherapy works by reactivation of a stalled pre-existing immune response or by eliciting a de novo immune response, and its toolkit comprises antibodies, vaccines, cytokines, and cell-based therapies. The treatment paradigm in some malignancies has completely changed over the past 10 to 15 years. Massive efforts in preclinical development have led to a surge of clinical trials testing innovative therapeutic approaches as monotherapy and, increasingly, in combination. Here we provide an overview of approved and emerging antitumor immune therapies, focusing on the rich landscape of therapeutic approaches beyond those that block the canonical PD-1/PD-L1 and CTLA-4 axes and placing them in the context of the latest understanding of tumor immunology.
Collapse
Key Words
- BiTE, bispecific T cell engager
- CAR, chimeric antigen receptor
- CRS, cytokine-release syndrome
- FDA, U.S. Food and Drug Administration
- HLA, human leukocyte antigen
- ICI, immune checkpoint inhibitor
- IL, interleukin
- NK, natural killer
- NSCLC, non–small cell lung cancer
- TIL, tumor-infiltrating lymphocyte
- alloHCT, allogeneic hematopoietic stem cell transplantation
- cancer
- immune therapy
- immunotherapy
- innovation
- mAb, monoclonal antibody
- treatment
Collapse
Affiliation(s)
- Nathan E. Welty
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Saar I. Gill
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Address for correspondence: Dr Saar I. Gill, Smilow Center for Translational Research, Room 8-101, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
16
|
Liu D, Qi X, Wei X, Zhao L, Wang X, Li S, Wang Z, Shi L, Xu J, Hong M, Liu Z, Zhao L, Wang X, Zhang B, Zhang Y, Wang F, Cao YJ. A Novel Her2/VEGFR2/CD3 trispecific antibody with an optimal structural design showed improved T-cell-redirecting antitumor efficacy. Am J Cancer Res 2022; 12:7788-7803. [PMID: 36451856 PMCID: PMC9706591 DOI: 10.7150/thno.75037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Rationale: T-cell-redirecting bispecific antibodies (bsAbs) and trispecific antibodies (tsAbs) designed to recognize different epitopes or antigens have emerged as promising cancer therapies. Current approaches are all designed to include another antibody specific to the site of the primary antibody, and the molecular structures are generally established. However, the dimensions of target molecule and epitope location play a key role in the efficiency of the immunological synapse (IS) formation and subsequent T-cell-redirecting activities, therefore the connection flexibility of these antibodies determines the geometries of different formats of these molecules and will have a major impact on the efficacy. Methods: We describe a novel recombination strategy using various linker designs to site-specifically fuse anti-Her2 (2Rs15) or anti-VEGFR2 (3VGR19) nanobodies to different positions of the anti-CD3 antibody fragment (Fab, SP34). Based on the comparison among the various antigen-specific bsAbs, we could determine the desired fusion site of each nanobody to SP34, and further ensure the optimal structure of tsAbs with synergistic dual-antigen enhanced T-cell-redirecting activities. Results: This approach allows precise control of the formation of IS between Her2- and/or VEGFR2-expressing cancer cells and T cells, to obtain the optimal structure of the Her2/VEGFR2/CD3 tsAb without the need to map antibody-binding epitopes. Optimization of Her2/VEGFR2/CD3 tsAb results in enhanced T-cell-redirecting in vitro and in vivo antitumor efficacy compared with the corresponding bsAbs alone or in combination, and the potency to overcome tumor relapse due to antigen escape or resistance to Herceptin and Cyramza therapy. Conclusion: The novel design strategy for developing tsAbs using a site-specific recombination approach represents a promising platform for immuno-oncology and in applications other than cancer therapy.
Collapse
Affiliation(s)
- Dong Liu
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Xuexiu Qi
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Xiaoyi Wei
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Lijun Zhao
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Xuechun Wang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Shuhong Li
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Zhidong Wang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Licai Shi
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Jiean Xu
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Mei Hong
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Zhong Liu
- Lunan Pharmaceutical Group Co., Ltd, Feixian County, Shandong, 273400, China
| | - Lili Zhao
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Feixian County, Shandong, 273400, China.,✉ Corresponding authors: ;
| | - Xiankun Wang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China.,Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Bo Zhang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China.,Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Yuhan Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Beijing Translational Center for Biopharmaceuticals Institute of Biophysics, Chinese Academy of Sciences Beijing 100101, China
| | - Feng Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Beijing Translational Center for Biopharmaceuticals Institute of Biophysics, Chinese Academy of Sciences Beijing 100101, China
| | - Yu J. Cao
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China.,Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.,✉ Corresponding authors: ;
| |
Collapse
|
17
|
Kang J, Sun T, Zhang Y. Immunotherapeutic progress and application of bispecific antibody in cancer. Front Immunol 2022; 13:1020003. [PMID: 36341333 PMCID: PMC9630604 DOI: 10.3389/fimmu.2022.1020003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/05/2022] [Indexed: 08/19/2023] Open
Abstract
Bispecific antibodies (bsAbs) are artificial antibodies with two distinct antigen-binding sites that can bind to different antigens or different epitopes on the same antigen. Based on a variety of technology platforms currently developed, bsAbs can exhibit different formats and mechanisms of action. The upgrading of antibody technology has promoted the development of bsAbs, which has been effectively used in the treatment of tumors. So far, 7 bsAbs have been approved for marketing in the world, and more than 200 bsAbs are in clinical and preclinical research stages. Here, we summarize the development process of bsAbs, application in tumor treatment and look forward to the challenges in future development.
Collapse
Affiliation(s)
- Jingyue Kang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tonglin Sun
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Zhang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Dakhel S, Galbiati A, Migliorini F, Comacchio C, Oehler S, Prati L, Scheuermann J, Cazzamalli S, Neri D, Bassi G, Favalli N. Isolation of a Natural Killer Group 2D Small-Molecule Ligand from DNA-Encoded Chemical Libraries. ChemMedChem 2022; 17:e202200350. [PMID: 35929380 DOI: 10.1002/cmdc.202200350] [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: 06/30/2022] [Revised: 08/04/2022] [Indexed: 11/11/2022]
Abstract
Natural Killer Group 2D (NKG2D) is a homo-dimeric transmembrane protein which is typically expressed on the surface of natural killer (NK) cells, natural killer T (NKT) cells, gamma delta T (γδT) cells, activated CD8 positive T-cells and activated macrophages. Bispecific molecules, capable of bridging NKG2D with a target protein expressed on the surface of tumor cells, may be used to redirect the cytotoxic activity of NK-cells towards antigen-positive malignanT-cells. In this work, we report the discovery of a novel NKG2D small molecule binder [K D = (410±60) nM], isolated from a DNA-Encoded Chemical Library (DEL). The discovery of small organic NKG2D ligands may facilitate the generation of fully synthetic bispecific adaptors, which may serve as an alternative to bispecific antibody products and which may benefit from better tumor targeting properties.
Collapse
Affiliation(s)
| | | | | | | | | | - Luca Prati
- Philogen SpA, R&D (Philochem), SWITZERLAND
| | - Jörg Scheuermann
- ETH Zürich: Eidgenossische Technische Hochschule Zurich, chemistry and applied biosciences, SWITZERLAND
| | | | | | | | - Nicholas Favalli
- Philogen SpA, R&D (Philochem), Libernstrasse 3, 8112, Otelfingen, SWITZERLAND
| |
Collapse
|
19
|
Translational Modeling Predicts Efficacious Therapeutic Dosing Range of Teclistamab for Multiple Myeloma. Target Oncol 2022; 17:433-439. [PMID: 35749004 PMCID: PMC9345835 DOI: 10.1007/s11523-022-00893-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 10/25/2022]
Abstract
BACKGROUND Teclistamab (JNJ-64007957), a B-cell maturation antigen × CD3 bispecific antibody, displayed potent T-cell-mediated cytotoxicity of multiple myeloma cells in preclinical studies. OBJECTIVE A first-in-human, Phase I, dose escalation study (MajesTEC-1) is evaluating teclistamab in patients with relapsed/refractory multiple myeloma. PATIENTS AND METHODS To estimate the efficacious therapeutic dosing range of teclistamab, pharmacokinetic (PK) data following the first cycle doses in the low-dose cohorts in the Phase I study were modeled using a 2-compartment model and simulated to predict the doses that would have average and trough serum teclistamab concentrations in the expected therapeutic range (between EC50 and EC90 values from an ex vivo cytotoxicity assay). RESULTS The doses predicted to have average serum concentrations between the EC50 and EC90 range were validated. In addition, simulations showed that weekly intravenous and subcutaneous doses of 0.70 mg/kg and 0.72 mg/kg, respectively, resulted in mean trough levels comparable to the maximum EC90. The most active doses in the Phase I study were weekly intravenous doses of 0.27 and 0.72 mg/kg and weekly subcutaneous doses of 0.72 and 1.5 mg/kg, with the weekly 1.5 mg/kg subcutaneous doses selected as the recommended Phase II dose (RP2D). With active doses, exposure was maintained above the mean EC90. All patients who responded to the RP2D of teclistamab had exposure above the maximum EC90 in both serum and bone marrow on cycle 3, Day 1 of treatment. CONCLUSIONS Our findings show that PK simulations of early clinical data together with ex vivo cytotoxicity estimates can inform the identification of a bispecific antibody's therapeutic range. CLINICAL TRIAL REGISTRATION NCT03145181, date of registration: May 9, 2017.
Collapse
|
20
|
Ordóñez-Reyes C, Garcia-Robledo JE, Chamorro DF, Mosquera A, Sussmann L, Ruiz-Patiño A, Arrieta O, Zatarain-Barrón L, Rojas L, Russo A, de Miguel-Perez D, Rolfo C, Cardona AF. Bispecific Antibodies in Cancer Immunotherapy: A Novel Response to an Old Question. Pharmaceutics 2022; 14:pharmaceutics14061243. [PMID: 35745815 PMCID: PMC9229626 DOI: 10.3390/pharmaceutics14061243] [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: 04/24/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 01/14/2023] Open
Abstract
Immunotherapy has redefined the treatment of cancer patients and it is constantly generating new advances and approaches. Among the multiple options of immunotherapy, bispecific antibodies (bsAbs) represent a novel thoughtful approach. These drugs integrate the action of the immune system in a strategy to redirect the activation of innate and adaptive immunity toward specific antigens and specific tumor locations. Here we discussed some basic aspects of the design and function of bsAbs, their main challenges and the state-of-the-art of these molecules in the treatment of hematological and solid malignancies and future perspectives.
Collapse
Affiliation(s)
- Camila Ordóñez-Reyes
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Juan Esteban Garcia-Robledo
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ 85054, USA
| | - Diego F. Chamorro
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Andrés Mosquera
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
| | - Liliana Sussmann
- Department of Neurology, Fundación Universitaria de Ciencias de la Salud, Bogotá 111221, Colombia;
| | - Alejandro Ruiz-Patiño
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Oscar Arrieta
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), Mexico City 14080, Mexico; (O.A.); (L.Z.-B.)
| | - Lucia Zatarain-Barrón
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), Mexico City 14080, Mexico; (O.A.); (L.Z.-B.)
| | - Leonardo Rojas
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
| | | | - Diego de Miguel-Perez
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.d.M.-P.); (C.R.)
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.d.M.-P.); (C.R.)
| | - Andrés F. Cardona
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
- Direction of Research, Science and Education, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC), Bogotá 110131, Colombia
- Correspondence: ; Tel.: +57-(1)-6190052; Fax: +57-(1)-6190053
| |
Collapse
|
21
|
Bannerji R, Arnason JE, Advani RH, Brown JR, Allan JN, Ansell SM, Barnes JA, O'Brien SM, Chávez JC, Duell J, Rosenwald A, Crombie JL, Ufkin M, Li J, Zhu M, Ambati SR, Chaudhry A, Lowy I, Topp MS. Odronextamab, a human CD20×CD3 bispecific antibody in patients with CD20-positive B-cell malignancies (ELM-1): results from the relapsed or refractory non-Hodgkin lymphoma cohort in a single-arm, multicentre, phase 1 trial. Lancet Haematol 2022; 9:e327-e339. [PMID: 35366963 PMCID: PMC10681157 DOI: 10.1016/s2352-3026(22)00072-2] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Odronextamab is a hinge-stabilised, fully human IgG4-based CD20 × CD3 bispecific antibody that binds CD3 on T cells and CD20 on B cells. We aimed to evaluate the safety and antitumour activity of odronextamab in patients with relapsed or refractory B-cell non-Hodgkin lymphoma. METHODS This single-arm, multicentre, phase 1, dose-escalation and dose-expansion (ELM-1) trial was conducted at ten academic sites across the USA and Germany. Patients aged 18 years or older with CD20-positive relapsed or refractory B-cell malignancies who previously received CD20-directed antibody therapy and who had at least one measurable lesion, and an ECOG performance status of 0 or 1 were included. Patients received intravenous odronextamab, according to a step-up dosing schedule in cycle 1, followed by treatment once per week at target doses ranging from 0·1 mg to 320 mg during cycles 2-4 (each cycle was 21 days). After cycle 4, maintenance treatment occurred every 2 weeks until disease progression or unacceptable toxicity. The primary endpoint of safety was assessed by the incidence of adverse events and dose-limiting toxicities to determine the maximum tolerated dose or phase 2 dose of odronextamab, or both. Preliminary antitumour activity, as measured by objective response rate, was a secondary endpoint. This study is registered with ClinicalTrials.gov, NCT02290951. FINDINGS From Feb 4, 2015, to Sept 25, 2021, 145 heavily pretreated patients (median of 3 (IQR 2-5] previous therapies) were enrolled (94 to the dose-escalation and 51 to the dose-expansion part of the study). The median age of patients was 67·0 years (IQR 57·0-73·0); 101 (70%) were male and 44 (30%) were female; most participants were White (119 [82%]) and not Hispanic or Latino (132 [91%]). 42 (29%) patients received previous CAR T therapy and 119 (82%) were refractory to the last line of therapy. Median duration of follow-up was 4·2 months (IQR 1·5-11·5). During dose escalation, odronextamab was administered up to the maximum dose of 320 mg once per week and no dose-limiting toxicities were observed. The recommended dose for expansion in patients with follicular lymphoma grade 1-3a was 80 mg and was 160 mg for patients with diffuse large B-cell lymphoma. Cytokine release syndrome and neurological treatment-emergent adverse events were predominantly low grade and did not result in treatment discontinuation. The most common grade 3 or worse treatment-emergent adverse events were anaemia (36 [25%]), lymphopenia (28 [19%]), hypophosphataemia (27 [19%]), neutropenia (27 [19%]), and thrombocytopenia (20 [14%]). Serious treatment-emergent adverse events occurred in 89 (61%) of 145 patients; the most frequent were cytokine release syndrome (41 [28%]), pyrexia (11 [8%]), pneumonia (nine [6%]), and infusion-related reaction (six [4%]). Four deaths were considered related to treatment (gastric perforation in a patient with gastric involvement by lymphoma, lung infection, pneumonia, and tumour-lysis syndrome). Objective response rate was 51% (95% CI 42-59; 72 of 142). In patients with follicular lymphoma who received odronextamab doses of 5 mg or higher, the objective response rate was 91% (95% CI 75-98; 29 of 32) and the complete response rate was 72% (95% CI 53-86; 23 of 32). In patients with diffuse large B-cell lymphoma without previous CAR T-cell therapy who received doses of 80 mg or higher, the objective response rate was 53% (eight of 15) and all responses were complete responses. In patients with diffuse large B-cell lymphoma who had previous CAR T-cell therapy and received doses of 80 mg or higher, the objective response rate was 33% (ten of 30) and complete response rate was 27% (eight of 30). INTERPRETATION Odronextamab monotherapy showed a manageable safety profile and encouraging preliminary activity, including durable responses in heavily pretreated patients with B-cell non-Hodgkin lymphoma, supporting further clinical investigation in phase 2 and 3 trials. FUNDING Regeneron Pharmaceuticals.
Collapse
Affiliation(s)
- Rajat Bannerji
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| | - Jon E Arnason
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | | | | | | | | | - Johannes Duell
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | | | | | | | - Jingjin Li
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Min Zhu
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | - Israel Lowy
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Max S Topp
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| |
Collapse
|
22
|
Tavarozzi R, Manzato E. The Role of Bispecific Antibodies in Non-Hodgkin's Lymphoma: From Structure to Prospective Clinical Use. Antibodies (Basel) 2022; 11:16. [PMID: 35225874 PMCID: PMC8883977 DOI: 10.3390/antib11010016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022] Open
Abstract
Bispecific antibodies (bsAbs) are molecules that simultaneously bind two different antigens (Ags). bsAbs represent a very active field in tumor immunotherapy with more than one hundred molecules currently being tested. More specifically, they have elicited a great interest in the setting of non-Hodgkin's lymphoma (NHLs), where they could represent a viable option for more fragile patients or those resistant to other conventional therapies. This review aims to give a brief overview of the different available bsAb formats and their mechanisms of action, pinpointing the differences between IgG-like and non-IgG-like classes and will then focus on those in advanced clinical development for NHLs.
Collapse
Affiliation(s)
- Rita Tavarozzi
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
- SCDU of Hematology, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Enrica Manzato
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, 56127 Pisa, Italy;
| |
Collapse
|
23
|
Thoreau F, Chudasama V. Enabling the next steps in cancer immunotherapy: from antibody-based bispecifics to multispecifics, with an evolving role for bioconjugation chemistry. RSC Chem Biol 2022; 3:140-169. [PMID: 35360884 PMCID: PMC8826860 DOI: 10.1039/d1cb00082a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 10/22/2021] [Indexed: 12/02/2022] Open
Abstract
In the past two decades, immunotherapy has established itself as one of the leading strategies for cancer treatment, as illustrated by the exponentially growing number of related clinical trials. This trend was, in part, prompted by the clinical success of both immune checkpoint modulation and immune cell engagement, to restore and/or stimulate the patient's immune system's ability to fight the disease. These strategies were sustained by progress in bispecific antibody production. However, despite the decisive progress made in the treatment of cancer, toxicity and resistance are still observed in some cases. In this review, we initially provide an overview of the monoclonal and bispecific antibodies developed with the objective of restoring immune system functions to treat cancer (cancer immunotherapy), through immune checkpoint modulation, immune cell engagement or a combination of both. Their production, design strategy and impact on the clinical trial landscape are also addressed. In the second part, the concept of multispecific antibody formats, notably MuTICEMs (Multispecific Targeted Immune Cell Engagers & Modulators), as a possible answer to current immunotherapy limitations is investigated. We believe it could be the next step to take for cancer immunotherapy research and expose why bioconjugation chemistry might play a key role in these future developments.
Collapse
Affiliation(s)
- Fabien Thoreau
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
24
|
Lim J, Sari-Ak D, Bagga T. Siglecs as Therapeutic Targets in Cancer. BIOLOGY 2021; 10:1178. [PMID: 34827170 PMCID: PMC8615218 DOI: 10.3390/biology10111178] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
Abstract
Hypersialylation is a common post-translational modification of protein and lipids found on cancer cell surfaces, which participate in cell-cell interactions and in the regulation of immune responses. Sialic acids are a family of nine-carbon α-keto acids found at the outermost ends of glycans attached to cell surfaces. Given their locations on cell surfaces, tumor cells aberrantly overexpress sialic acids, which are recognized by Siglec receptors found on immune cells to mediate broad immunomodulatory signaling. Enhanced sialylation exposed on cancer cell surfaces is exemplified as "self-associated molecular pattern" (SAMP), which tricks Siglec receptors found on leukocytes to greatly down-regulate immune responsiveness, leading to tumor growth. In this review, we focused on all 15 human Siglecs (including Siglec XII), many of which still remain understudied. We also highlighted strategies that disrupt the course of Siglec-sialic acid interactions, such as antibody-based therapies and sialic acid mimetics leading to tumor cell depletion. Herein, we introduced the central roles of Siglecs in mediating pro-tumor immunity and discussed strategies that target these receptors, which could benefit improved cancer immunotherapy.
Collapse
Affiliation(s)
- Jackwee Lim
- Singapore Immunology Network, A*STAR, 8a Biomedical Grove, Singapore 138648, Singapore;
| | - Duygu Sari-Ak
- Department of Medical Biology, School of Medicine, University of Health Sciences, Istanbul 34668, Turkey;
| | - Tanaya Bagga
- Singapore Immunology Network, A*STAR, 8a Biomedical Grove, Singapore 138648, Singapore;
| |
Collapse
|
25
|
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: 13] [Impact Index Per Article: 3.3] [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
|
26
|
Kambhampati S, Song JY, Herrera AF, Chan WC. Barriers to achieving a cure in lymphoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:965-983. [PMID: 35582375 PMCID: PMC8992454 DOI: 10.20517/cdr.2021.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/08/2021] [Accepted: 10/26/2021] [Indexed: 11/23/2022]
Abstract
Lymphoma is a diverse disease with a variety of different subtypes, each characterized by unique pathophysiology, tumor microenvironment, and underlying signaling pathways leading to oncogenesis. With our increasing understanding of the molecular biology of lymphoma, there have been a number of novel targeted therapies and immunotherapy approaches that have been developed for the treatment of this complex disease. Despite rapid progress in the field, however, many patients still relapse largely due to the development of drug resistance to these therapies. A better understanding of the mechanisms underlying resistance is needed to develop more novel treatment strategies that circumvent these mechanisms and design better treatment algorithms that personalize therapies to patients and sequence these therapies in the most optimal manner. This review focuses on the recent advances in therapies in lymphoma, including targeted therapies, monoclonal antibodies, antibody-drug conjugates, cellular therapy, bispecific antibodies, and checkpoint inhibitors. We discuss the genetic and cellular principles of drug resistance that span across all the therapies, as well as some of the unique mechanisms of resistance that are specific to these individual classes of therapies and the strategies that have been developed to address these modes of resistance.
Collapse
Affiliation(s)
- Swetha Kambhampati
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Joo Y. Song
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Alex F. Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Wing C. Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
27
|
Kaleka G, Schiller G. Immunotherapy for Acute Myeloid Leukemia: Allogeneic hematopoietic cell transplantation is here to stay. Leuk Res 2021; 112:106732. [PMID: 34864447 DOI: 10.1016/j.leukres.2021.106732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 01/20/2023]
Abstract
Acute Myeloid Leukemia (AML) represents 1 % of all new cancer diagnosis made annually in the US and has a five-year survival of 30 %. Traditional treatment includes aggressive induction therapy followed by consolidation therapy that may include a hematopoietic stem cell transplant (HSCT). Thus far, HSCT remains the only potentially curative therapy for many patients with AML owing to the graft-versus-leukemia effect elicited by this treatment. The use of novel therapies, specifically immunotherapy, in the treatment of AML has been limited by the lack of appropriate target antigens, therapy associated toxicities and variable success with treatment. Antigenic variability on leukemia cells and the sharing of antigens by malignant and non-malignant cells makes the identification of appropriate antigens problematic. While studies with immunotherapeutic agents are underway, prior investigations have demonstrated a mixed response with some studies prematurely discontinued due to associated toxicities. This review presents a discussion of the envisioned role of immunotherapy in the treatment of AML in the setting of mixed therapeutic success and potentially lethal toxicities.
Collapse
Affiliation(s)
- Guneet Kaleka
- UCLA-Olive View Medical Center, Department of Medicine, Room 2B-182, 14445 Olive View Drive, Sylmar, CA, 91342, United States.
| | - Gary Schiller
- Department of Medicine, Hematology & Oncology at UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
28
|
Abstract
Advances in understanding the ways in which the immune system fails to control tumor growth or prevent autoimmunity have led to the development of powerful therapeutic strategies to treat these diseases. In contrast to conventional therapies that have a broadly suppressive effect, immunotherapies are more akin to targeted therapies because they are mechanistically driven and are typically developed with the goal of "drugging" a specific underlying pathway or phenotype. This means that their effects and toxicities are, at least in theory, more straightforward to anticipate. The development of functionalized antibodies, genetically engineered T cells, and immune checkpoint inhibitors continues to accelerate, illuminating new biology and bringing new treatment to patients. In the following sections, we provide an overview of immunotherapeutic concepts, highlight recent advances in the field of immunotherapies, and discuss controversies and future directions, particularly as these pertain to hematologic oncology or blood-related diseases. We conclude by illustrating how original research published in this journal fits into and contributes to the overall framework of advances in immunotherapy.
Collapse
Affiliation(s)
- Stefanie Lesch
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA; and
- Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Saar Gill
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, PA; and
- Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
29
|
Abramson HN. Immunotherapy of Multiple Myeloma: Promise and Challenges. Immunotargets Ther 2021; 10:343-371. [PMID: 34527606 PMCID: PMC8437262 DOI: 10.2147/itt.s306103] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022] Open
Abstract
Whereas the treatment of MM was dependent solely on alkylating agents and corticosteroids during the prior three decades, the landscape of therapeutic measures to treat the disease began to expand enormously early in the current century. The introduction of new classes of small-molecule drugs, such as proteasome blockers (bortezomib and carfilzomib), immunomodulators (lenalidomide and pomalidomide), nuclear export inhibitors (selinexor), and histone deacetylase blockers (panobinostat), as well as the application of autologous stem cell transplantation (ASCT), resulted in a seismic shift in how the disease is treated. The picture changed dramatically once again starting with the 2015 FDA approval of two monoclonal antibodies (mAbs) - the anti-CD38 daratumumab and the anti-SLAMF7 elotuzumab. Daratumumab, in particular, has had a great impact on MM therapy and today is often included in various regimens to treat the disease, both in newly diagnosed cases and in the relapse/refractory setting. Recently, other immunotherapies have been added to the arsenal of drugs available to fight this malignancy. These include isatuximab (also anti-CD38) and, in the past year, the antibody-drug conjugate (ADC) belantamab mafodotin and the chimeric antigen receptor (CAR) T-cell product idecabtagene vicleucel (ide-cel). While the accumulated benefits of these newer agents have resulted in a doubling of the disease's five-year survival rate to more than 5 years and improved quality of life, the disease remains incurable. Almost without exception patients experience relapse and/or become refractory to the drugs used, making the search for innovative therapies all the more essential. This review covers the current scope of anti-myeloma immunotherapeutic agents, both those in clinical use and on the horizon, including naked mAbs, ADCs, bi- and multi-targeted mAbs, and CAR T-cells. Emphasis is placed on the benefits of each along with the challenges that need to be overcome if MM is to be considered curable in the future.
Collapse
Affiliation(s)
- Hanley N Abramson
- Wayne State University, Department of Pharmaceutical Sciences, Detroit, MI, 48201, USA
| |
Collapse
|
30
|
Namoglu EC, Hughes ME, Nasta SD. Targeted immunotherapies to consider for B Cell non-hodgkin lymphoma. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1967142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Esin C. Namoglu
- Lymphoma Program, Hematology/Oncology Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Mitchell E. Hughes
- Lymphoma Program, Hematology/Oncology Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sunita D. Nasta
- Lymphoma Program, Hematology/Oncology Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
31
|
Lim SM, Pyo KH, Soo RA, Cho BC. The promise of bispecific antibodies: Clinical applications and challenges. Cancer Treat Rev 2021; 99:102240. [PMID: 34119803 DOI: 10.1016/j.ctrv.2021.102240] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023]
Abstract
The development of cancer therapies using monoclonal antibodies has been successful during the last 30 years. Recently much progress was achieved with technologies involving bispecific and multi-specific antibodies. Bispecific antibodies (BsAbs) are antibodies that bind two distinct epitopes, and a large number of potential clinical applications of BsAbs have been described. Here we review mechanism of action, clinical development and future challenges of BsAbs which could be a serve as a valuable arsenal in cancer patients.
Collapse
Affiliation(s)
- Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyoung-Ho Pyo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ross A Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore.
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
32
|
Epcoritamab induces potent anti-tumor activity against malignant B-cells from patients with DLBCL, FL and MCL, irrespective of prior CD20 monoclonal antibody treatment. Blood Cancer J 2021; 11:38. [PMID: 33602901 PMCID: PMC7892878 DOI: 10.1038/s41408-021-00430-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/30/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
Epcoritamab (DuoBody-CD3xCD20, GEN3013) is a novel bispecific IgG1 antibody redirecting T-cells toward CD20+ tumor cells. Here, we assessed the preclinical efficacy of epcoritamab against primary tumor cells present in the lymph node biopsies from newly diagnosed (ND) and relapsed/refractory (RR) B-NHL patients. In the presence of T-cells from a healthy donor, epcoritamab demonstrated potent activity against primary tumor cells, irrespective of prior treatments, including CD20 mAbs. Median lysis of 65, 74, and 84% were achieved in diffuse large B-cell lymphoma (n = 16), follicular lymphoma (n = 15), and mantle cell lymphoma (n = 8), respectively. Furthermore, in this allogeneic setting, we discovered that the capacity of B-cell tumors to activate T-cells was heterogeneous and showed an inverse association with their surface expression levels of the immune checkpoint molecule Herpesvirus Entry Mediator (HVEM). In the autologous setting, when lymph node (LN)-residing T-cells were the only source of effector cells, the epcoritamab-dependent cytotoxicity strongly correlated with local effector cell-to-target cell ratios. Further analyses revealed that LN-residing-derived or peripheral blood-derived T-cells of B-NHL patients, as well as heathy donor T-cells equally mediated epcoritamab-dependent cytotoxicity. These results show the promise of epcoritamab for treatment of newly-diagnosed or relapsed/refractory B-NHL patients, including those who became refractory to previous CD20-directed therapies.
Collapse
|
33
|
McKean WB, Moser JC, Rimm D, Hu-Lieskovan S. Biomarkers in Precision Cancer Immunotherapy: Promise and Challenges. Am Soc Clin Oncol Educ Book 2021; 40:e275-e291. [PMID: 32453632 DOI: 10.1200/edbk_280571] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The rapid expansion of modern cancer immunotherapeutics has led to a dramatic improvement in patient survival and sustained remission for otherwise refractory malignancies. However, a significant limitation behind these current treatment modalities is an irregularity in clinical response, which is especially pronounced among checkpoint inhibition. This unpredictability leads to significant side effects, financial costs, and health care burden, with unsatisfactory clinical benefit in the majority of treated patients. Additionally, although ongoing studies and trials investigate the use of multiple biomarkers predictive of patient response or harm, none of these are comprehensive in predicting potential benefit. This unmet need for validated biomarkers is largely secondary to a prohibitive complexity within tumor parenchyma and microenvironment, dynamic clonal and proteomic changes to therapy, heterogenous host immune defects, and varied standardization among sample preparation and reporting. Herein, we discuss current advantages of predictive biomarkers, as well as limitations in their clinical use and application. We also review future directions, ideal characteristics, and trial design needed for proper precision immuno-oncology and biomarker development.
Collapse
Affiliation(s)
- William B McKean
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | | - David Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | | |
Collapse
|
34
|
Khaldoyanidi S, Nagorsen D, Stein A, Ossenkoppele G, Subklewe M. Immune Biology of Acute Myeloid Leukemia: Implications for Immunotherapy. J Clin Oncol 2021; 39:419-432. [PMID: 33434043 PMCID: PMC8078464 DOI: 10.1200/jco.20.00475] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
| | | | - Anthony Stein
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Gerrit Ossenkoppele
- Amsterdam University Medical Center, Location VU University Medical Center, Amsterdam, the Netherlands
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
35
|
Current Immunotherapy Approaches in Non-Hodgkin Lymphomas. Vaccines (Basel) 2020; 8:vaccines8040708. [PMID: 33260966 PMCID: PMC7768428 DOI: 10.3390/vaccines8040708] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
Non-Hodgkin lymphomas (NHLs) are lymphoid malignancies of B- or T-cell origin. Despite great advances in treatment options and significant improvement of survival parameters, a large part of NHL patients either present with a chemotherapy-refractory disease or experience lymphoma relapse. Chemotherapy-based salvage therapy of relapsed/refractory NHL is, however, capable of re-inducing long-term remissions only in a minority of patients. Immunotherapy-based approaches, including bispecific antibodies, immune checkpoint inhibitors and genetically engineered T-cells carrying chimeric antigen receptors, single-agent or in combination with therapeutic monoclonal antibodies, immunomodulatory agents, chemotherapy or targeted agents demonstrated unprecedented clinical activity in heavily-pretreated patients with NHL, including chemotherapy-refractory cases with complex karyotype changes and other adverse prognostic factors. In this review, we recapitulate currently used immunotherapy modalities in NHL and discuss future perspectives of combinatorial immunotherapy strategies, including patient-tailored approaches.
Collapse
|
36
|
Collins M, Awwad S, Ibeanu N, Khaw PT, Guiliano D, Brocchini S, Khalili H. Dual-acting therapeutic proteins for intraocular use. Drug Discov Today 2020; 26:44-55. [PMID: 33137484 DOI: 10.1016/j.drudis.2020.10.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/22/2020] [Accepted: 10/26/2020] [Indexed: 12/25/2022]
Abstract
Intravitreally injected antibody-based medicines have revolutionised the treatment of retinal disease. Bispecific and dual-functional antibodies and therapeutic proteins have the potential to further increase the efficacy of intraocular medicines.
Collapse
Affiliation(s)
- Matthew Collins
- School of Health, Sport and Bioscience, University of East London, London, E15 4LZ, UK; School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Sahar Awwad
- School of Pharmacy, University College London, London, WC1N 1AX, UK; National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, EC1V 9EL, UK
| | - Nkiru Ibeanu
- School of Pharmacy, University College London, London, WC1N 1AX, UK; National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, EC1V 9EL, UK
| | - Peng T Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, EC1V 9EL, UK
| | - David Guiliano
- School of Health, Sport and Bioscience, University of East London, London, E15 4LZ, UK
| | - Steve Brocchini
- School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Hanieh Khalili
- School of Health, Sport and Bioscience, University of East London, London, E15 4LZ, UK; School of Pharmacy, University College London, London, WC1N 1AX, UK.
| |
Collapse
|
37
|
Huarte E, O'Connor RS, Peel MT, Nunez-Cruz S, Leferovich J, Juvekar A, Yang YO, Truong L, Huang T, Naim A, Milone MC, Smith PA. Itacitinib (INCB039110), a JAK1 Inhibitor, Reduces Cytokines Associated with Cytokine Release Syndrome Induced by CAR T-cell Therapy. Clin Cancer Res 2020; 26:6299-6309. [PMID: 32998963 DOI: 10.1158/1078-0432.ccr-20-1739] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/14/2020] [Accepted: 09/23/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE T cells engineered to express a chimeric antigen receptor (CAR) are a promising cancer immunotherapy. Such targeted therapies have shown long-term relapse-free survival in patients with B-cell leukemia and lymphoma. However, cytokine release syndrome (CRS) represents a serious, potentially life-threatening side effect often associated with CAR T-cell therapy. CRS manifests as a rapid (hyper)immune reaction driven by excessive inflammatory cytokine release, including IFNγ and IL6. EXPERIMENTAL DESIGN Many cytokines implicated in CRS are known to signal through the JAK-STAT pathway. Here we study the effect of blocking JAK pathway signaling on CAR T-cell proliferation, antitumor activity, and cytokine levels in in vitro and in vivo models. RESULTS We report that itacitinib, a potent, selective JAK1 inhibitor, was able to significantly and dose-dependently reduce levels of multiple cytokines implicated in CRS in several in vitro and in vivo models. Importantly, we also report that at clinically relevant doses that mimic human JAK1 pharmacologic inhibition, itacitinib did not significantly inhibit proliferation or antitumor killing capacity of three different human CAR T-cell constructs (GD2, EGFR, and CD19). Finally, in an in vivo model, antitumor activity of CD19-CAR T cells adoptively transferred into CD19+ tumor-bearing immunodeficient animals was unabated by oral itacitinib treatment. CONCLUSIONS Together, these data suggest that itacitinib has potential as a prophylactic agent for the prevention of CAR T cell-induced CRS, and a phase II clinical trial of itacitinib for prevention of CRS induced by CAR T-cell therapy has been initiated (NCT04071366).
Collapse
Affiliation(s)
| | - Roddy S O'Connor
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Selene Nunez-Cruz
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Leferovich
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Yan-Ou Yang
- Incyte Research Institute, Wilmington, Delaware
| | - Lisa Truong
- Incyte Research Institute, Wilmington, Delaware
| | | | - Ahmad Naim
- Incyte Corporation, Wilmington, Delaware
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | |
Collapse
|
38
|
Hofmann T, Krah S, Sellmann C, Zielonka S, Doerner A. Greatest Hits-Innovative Technologies for High Throughput Identification of Bispecific Antibodies. Int J Mol Sci 2020; 21:E6551. [PMID: 32911608 PMCID: PMC7554978 DOI: 10.3390/ijms21186551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
Recent years have shown a tremendous increase and diversification in antibody-based therapeutics with advances in production techniques and formats. The plethora of currently investigated bi- to multi-specific antibody architectures can be harnessed to elicit a broad variety of specific modes of actions in oncology and immunology, spanning from enhanced selectivity to effector cell recruitment, all of which cannot be addressed by monospecific antibodies. Despite continuously growing efforts and methodologies, the identification of an optimal bispecific antibody as the best possible combination of two parental monospecific binders, however, remains challenging, due to tedious cloning and production, often resulting in undesired extended development times and increased expenses. Although automated high throughput screening approaches have matured for pharmaceutical small molecule development, it was only recently that protein bioconjugation technologies have been developed for the facile generation of bispecific antibodies in a 'plug and play' manner. In this review, we provide an overview of the most relevant methodologies for bispecific screening purposes-the DuoBody concept, paired light chain single cell production approaches, Sortase A and Transglutaminase, the SpyTag/SpyCatcher system, and inteins-and elaborate on the benefits as well as drawbacks of the different technologies.
Collapse
Affiliation(s)
- Tim Hofmann
- Advanced Cell Culture Technologies, Merck Life Sciences KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany;
| | - Simon Krah
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany; (S.K.); (C.S.); (S.Z.)
| | - Carolin Sellmann
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany; (S.K.); (C.S.); (S.Z.)
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany; (S.K.); (C.S.); (S.Z.)
| | - Achim Doerner
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany; (S.K.); (C.S.); (S.Z.)
| |
Collapse
|
39
|
Morcos PN, Li J, Hosseini I, Li CC. Quantitative Clinical Pharmacology of T-Cell Engaging Bispecifics: Current Perspectives and Opportunities. Clin Transl Sci 2020; 14:75-85. [PMID: 32882099 PMCID: PMC7877841 DOI: 10.1111/cts.12877] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
T-cell directing/engaging bispecifics (TDBs) enable a powerful mode of action by activating T-cells through the creation of artificial immune synapses. Their pharmacological response involves the dynamic inter-relationships among T-cells, tumor cells, and TDBs. This results in complex and challenging issues in understanding pharmacokinetics, tissue distribution, target engagement, and exposure-response relationship. Dosing strategy plays a crucial role in determining the therapeutic window of TDBs because of the desire to maximize therapeutic efficacy in the context of known mechanism-related adverse events, such as cytokine release syndrome and neurological adverse events. Such adverse events are commonly reported as the most prominent events during the initial treatment cycles and dissipate over time. Therefore, the kinetic characterization of the inter-relationships between exposure/target engagement and safety/efficacy outcomes is crucial in designing the optimal dosing regimen to maximize the benefit/risk of TDB agents. In this review, we discuss the key clinical pharmacological considerations in drug discovery and development for TDBs and provide a summary of TDBs currently in clinical development. We also propose forward-looking perspectives and opportunities to derive insights through quantitative clinical pharmacology approaches.
Collapse
Affiliation(s)
- Peter N Morcos
- Pharmaceutical Sciences
- Pharma Research and Early Development (pRED), Roche Innovation Center, New York, New York, USA
| | - Junyi Li
- Department of Clinical Pharmacology, Genentech, Roche, South San Francisco, California, USA
| | - Iraj Hosseini
- Preclinical and Translational Pharmacokinetics, Genentech, Roche, South San Francisco, California, USA
| | - Chi-Chung Li
- Department of Clinical Pharmacology, Genentech, Roche, South San Francisco, California, USA
| |
Collapse
|
40
|
Guo G, Han J, Wang Y, Li Y. A potential downstream platform approach for WuXiBody-based IgG-like bispecific antibodies. Protein Expr Purif 2020; 173:105647. [DOI: 10.1016/j.pep.2020.105647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
|
41
|
Betts A, van der Graaf PH. Mechanistic Quantitative Pharmacology Strategies for the Early Clinical Development of Bispecific Antibodies in Oncology. Clin Pharmacol Ther 2020; 108:528-541. [PMID: 32579234 PMCID: PMC7484986 DOI: 10.1002/cpt.1961] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023]
Abstract
Bispecific antibodies (bsAbs) have become an integral component of the therapeutic research strategy to treat cancer. In addition to clinically validated immune cell re‐targeting, bsAbs are being designed for tumor targeting and as dual immune modulators. Explorative preclinical and emerging clinical data indicate potential for enhanced efficacy and reduced systemic toxicity. However, bsAbs are a complex modality with challenges to overcome in early clinical trials, including selection of relevant starting doses using a minimal anticipated biological effect level approach, and predicting efficacious dose despite nonintuitive dose response relationships. Multiple factors can contribute to variability in the clinic, including differences in functional affinity due to avidity, receptor expression, effector to target cell ratio, and presence of soluble target. Mechanistic modeling approaches are a powerful integrative tool to understand the complexities and aid in clinical translation, trial design, and prediction of regimens and strategies to reduce dose limiting toxicities of bsAbs. In this tutorial, the use of mechanistic modeling to impact decision making for bsAbs is presented and illustrated using case study examples.
Collapse
Affiliation(s)
- Alison Betts
- Applied Biomath, Concord, Massachusetts, USA.,Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Piet H van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands.,Certara, Canterbury, UK
| |
Collapse
|
42
|
Giuliani M, Poggi A. Checkpoint Inhibitors and Engineered Cells: New Weapons for Natural Killer Cell Arsenal Against Hematological Malignancies. Cells 2020; 9:1578. [PMID: 32610578 PMCID: PMC7407972 DOI: 10.3390/cells9071578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells represent one of the first lines of defense against malignant cells. NK cell activation and recognition are regulated by a balance between activating and inhibitory receptors, whose specific ligands can be upregulated on tumor cells surface and tumor microenvironment (TME). Hematological malignancies set up an extensive network of suppressive factors with the purpose to induce NK cell dysfunction and impaired immune-surveillance ability. Over the years, several strategies have been developed to enhance NK cells-mediated anti-tumor killing, while other approaches have arisen to restore the NK cell recognition impaired by tumor cells and other cellular components of the TME. In this review, we summarize and discuss the strategies applied in hematological malignanciesto block the immune check-points and trigger NK cells anti-tumor effects through engineered chimeric antigen receptors.
Collapse
Affiliation(s)
- Massimo Giuliani
- Department of Oncology, Luxembourg Institute of Health, Luxembourg City L-1526, Luxembourg
| | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino IST, 16132 Genoa, Italy;
| |
Collapse
|
43
|
Ribera JM, Genescà E, Ribera J. Bispecific T-cell engaging antibodies in B-cell precursor acute lymphoblastic leukemias: focus on blinatumomab. Ther Adv Hematol 2020; 11:2040620720919632. [PMID: 32523659 PMCID: PMC7236391 DOI: 10.1177/2040620720919632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/05/2020] [Indexed: 11/28/2022] Open
Abstract
Bispecific T-cell engaging antibodies are constructs engineered to bind to two different antigens, one to a tumor-specific target and the other to CD3-positive T cells or natural killer (NK) cells. Blinatumomab engages CD19 and CD3, performing effective serial lysis. The clinical development program in acute lymphoblastic leukemia (ALL) includes clinical trials in relapsed or refractory (R/R) patients and in B-cell precursor (BCP) ALL patients with measurable residual disease. Several trials are currently being conducted in de novo BCP-ALL, either in induction, consolidation, or before or after hematopoietic stem cell transplant. Combination with other targeted therapies or with other immunotherapeutic approaches are also underway. Several strategies are aimed to optimize the use of blinatumomab either by overcoming the mechanisms of resistance (e.g. inhibition of PD-1/PD-L1) or by improvements in the route of application, among others.
Collapse
Affiliation(s)
- Jose-Maria Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias I Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, C/ Canyet, s/n, Badalona, 08916, Spain
| | - Eulalia Genescà
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Jordi Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| |
Collapse
|
44
|
Current Challenges in Providing Good Leukapheresis Products for Manufacturing of CAR-T Cells for Patients with Relapsed/Refractory NHL or ALL. Cells 2020; 9:cells9051225. [PMID: 32429189 PMCID: PMC7290830 DOI: 10.3390/cells9051225] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/09/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
Background: T lymphocyte collection through leukapheresis is an essential step for chimeric antigen receptor T (CAR-T) cell therapy. Timing of apheresis is challenging in heavily pretreated patients who suffer from rapid progressive disease and receive T cell impairing medication. Methods: A total of 75 unstimulated leukaphereses were analyzed including 45 aphereses in patients and 30 in healthy donors. Thereof, 41 adult patients with Non-Hodgkin’s lymphoma (85%) or acute lymphoblastic leukemia (15%) underwent leukapheresis for CAR-T cell production. Results: Sufficient lymphocytes were harvested from all patients even from those with low peripheral lymphocyte counts of 0.18/nL. Only four patients required a second leukapheresis session. Leukapheresis products contained a median of 98 × 108 (9 - 341 × 108) total nucleated cells (TNC) with 38 × 108 (4 - 232 × 108) CD3+ T cells. Leukapheresis products from healthy donors as well as from patients in complete remission were characterized by high TNC and CD3+ T lymphocyte counts. CAR-T cell products could be manufactured for all but one patient. Conclusions: Sufficient yield of lymphocytes for CAR-T cell production is feasible also for patients with low peripheral blood counts. Up to 12–15 L blood volume should be processed in patients with absolute lymphocyte counts ≤ 1.0/nL.
Collapse
|
45
|
Einsele H, Borghaei H, Orlowski RZ, Subklewe M, Roboz GJ, Zugmaier G, Kufer P, Iskander K, Kantarjian HM. The BiTE (bispecific T-cell engager) platform: Development and future potential of a targeted immuno-oncology therapy across tumor types. Cancer 2020; 126:3192-3201. [PMID: 32401342 DOI: 10.1002/cncr.32909] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/03/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022]
Abstract
Immuno-oncology therapies engage the immune system to treat cancer. BiTE (bispecific T-cell engager) technology is a targeted immuno-oncology platform that connects patients' own T cells to malignant cells. The modular nature of BiTE technology facilitates the generation of molecules against tumor-specific antigens, allowing off-the-shelf immuno-oncotherapy. Blinatumomab was the first approved canonical BiTE molecule and targets CD19 surface antigens on B cells, making blinatumomab largely independent of genetic alterations or intracellular escape mechanisms. Additional BiTE molecules in development target other hematologic malignancies (eg, multiple myeloma, acute myeloid leukemia, and B-cell non-Hodgkin lymphoma) and solid tumors (eg, prostate cancer, glioblastoma, gastric cancer, and small-cell lung cancer). BiTE molecules with an extended half-life relative to the canonical BiTE molecules are also being developed. Advances in immuno-oncology made with BiTE technology could substantially improve the treatment of hematologic and solid tumors and offer enhanced activity in combination with other treatments.
Collapse
Affiliation(s)
- Hermann Einsele
- Department of Internal Medicine II, Universität Würzburg, Würzburg, Germany
| | - Hossein Borghaei
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Gail J Roboz
- Weill Cornell Medicine, Division of Hematology and Oncology, The New York Presbyterian Hospital, New York, New York
| | | | - Peter Kufer
- Amgen Research (Munich) GmbH, Munich, Germany
| | | | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
46
|
Lejeune M, Köse MC, Duray E, Einsele H, Beguin Y, Caers J. Bispecific, T-Cell-Recruiting Antibodies in B-Cell Malignancies. Front Immunol 2020; 11:762. [PMID: 32457743 PMCID: PMC7221185 DOI: 10.3389/fimmu.2020.00762] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/03/2020] [Indexed: 12/12/2022] Open
Abstract
Bispecific antibodies (BsAbs) are designed to recognize and bind to two different antigens or epitopes. In the last few decades, BsAbs have been developed within the context of cancer therapies and in particular for the treatment of hematologic B-cell malignancies. To date, more than one hundred different BsAb formats exist, including bispecific T-cell engagers (BiTEs), and new constructs are constantly emerging. Advances in protein engineering have enabled the creation of BsAbs with specific mechanisms of action and clinical applications. Moreover, a better understanding of resistance and evasion mechanisms, as well as advances in the protein engineering and in immunology, will help generating a greater variety of BsAbs to treat various cancer types. This review focuses on T-cell-engaging BsAbs and more precisely on the various BsAb formats currently being studied in the context of B-cell malignancies, on ongoing clinical trials and on the clinical concerns to be taken into account in the development of new BsAbs.
Collapse
Affiliation(s)
- Margaux Lejeune
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Murat Cem Köse
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Elodie Duray
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Hermann Einsele
- Department of Internal Medicine II, University of Würzburg, Würzburg, Germany
| | - Yves Beguin
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium.,Department of Hematology, CHU de Liège, Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium.,Department of Hematology, CHU de Liège, Liège, Belgium
| |
Collapse
|
47
|
Caraccio C, Krishna S, Phillips DJ, Schürch CM. Bispecific Antibodies for Multiple Myeloma: A Review of Targets, Drugs, Clinical Trials, and Future Directions. Front Immunol 2020; 11:501. [PMID: 32391000 PMCID: PMC7193016 DOI: 10.3389/fimmu.2020.00501] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/04/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell malignancy and the second most common hematological neoplasm in adults, comprising 1.8% of all cancers. With an annual incidence of ~30,770 cases in the United States, MM has a high mortality rate, leading to 12,770 deaths per year. MM is a genetically complex, highly heterogeneous malignancy, with significant inter- and intra-patient clonal variability. Recent years have witnessed dramatic improvements in the diagnostics, classification, and treatment of MM. However, patients with high-risk disease have not yet benefited from therapeutic advances. High-risk patients are often primary refractory to treatment or relapse early, ultimately resulting in progression toward aggressive end-stage MM, with associated extramedullary disease or plasma cell leukemia. Therefore, novel treatment modalities are needed to improve the outcomes of these patients. Bispecific antibodies (BsAbs) are immunotherapeutics that simultaneously target and thereby redirect effector immune cells to tumor cells. BsAbs have shown high efficacy in B cell malignancies, including refractory/relapsed acute lymphoblastic leukemia. Various BsAbs targeting MM-specific antigens such as B cell maturation antigen (BCMA), CD38, and CD138 are currently in pre-clinical and clinical development, with promising results. In this review, we outline these advances, focusing on BsAb drugs, their targets, and their potential to improve survival, especially for high-risk MM patients. In combination with current treatment strategies, BsAbs may pave the way toward a cure for MM.
Collapse
|
48
|
Song W, Das M, Chen X. Nanotherapeutics for Immuno-Oncology: A Crossroad for New Paradigms. Trends Cancer 2020; 6:288-298. [PMID: 32209444 PMCID: PMC7101275 DOI: 10.1016/j.trecan.2020.01.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 02/06/2023]
Abstract
With the rapid increase in the use of nanotechnology and immunotherapy for cancer management in the recent past, there are great implications for using nanotechnology in immuno-oncology. However, to deliver clinical success, the scientific and clinical rationale must be critically evaluated when applying nanotechnology to immuno-oncology challenges. This opinion article distinguishes between designing nanotherapeutics for immunotherapy and the past focus on the placement of chemotherapy agents in nanoparticles. We believe the integration of nanotechnology with cancer immunotherapy for nano-immunotherapeutics provides unique opportunities for both fields, paving the way for entirely new therapeutic paradigms. As a particular focus in our article, we envision the necessities and challenges of nanotechnology in the development of in situ cancer vaccines, immune checkpoint inhibitors, adoptive cell transfer, and bispecific antibody therapy.
Collapse
Affiliation(s)
- Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, Jilin, 130022, China.
| | - Manisit Das
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, Jilin, 130022, China
| |
Collapse
|
49
|
Genome-scale CRISPR activation screen uncovers tumor-intrinsic modulators of CD3 bispecific antibody efficacy. Sci Rep 2019; 9:20068. [PMID: 31882897 PMCID: PMC6934601 DOI: 10.1038/s41598-019-56670-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
Bispecific antibodies (bsAb) that bridge tumor cells and CD3-positive effector T cells are being developed against many tumor cell targets. While tumor cell factors other than target expression level appear to play a role in determining the efficacy of CD3 bsAb, the identity of such factors remains largely unknown. Using a co-culture system of primary human T cells and B lymphoma cell lines, we demonstrate a range of sensitivities to CD20xCD3 bsAb that is independent of CD20 surface expression. To identify genes that modulate tumor cell sensitivity to CD3 bsAb, we employed a genome-scale CRISPR activation screen in a CD20xCD3-sensitive human B lymphoma cell line. Among the most highly enriched sgRNAs were those targeting genes with predicted effects on cell-cell adhesion, including sialophorin (SPN). Increased expression of SPN impeded tumor cell clustering with T cells, thereby limiting CD3 bsAb-mediated tumor cell lysis. This inhibitory effect of SPN appeared to be dependent on sialylated core 2 O-glycosylation of the protein. While SPN is not endogenously expressed in the majority of B cell lymphomas, it is highly expressed in acute myeloid leukemia. CRISPR-mediated SPN knockout in AML cell lines facilitated T cell-tumor cell clustering and enhanced CD3 bsAb-mediated AML cell lysis. In sum, our data establish that the cell cross-linking mechanism of CD3 bsAb is susceptible to subversion by anti-adhesive molecules expressed on the tumor cell surface. Further evaluation of anti-adhesive pathways may provide novel biomarkers of clinical response and enable the development of effective combination regimens for this promising therapeutic class.
Collapse
|
50
|
Nixon AB, Schalper KA, Jacobs I, Potluri S, Wang IM, Fleener C. Peripheral immune-based biomarkers in cancer immunotherapy: can we realize their predictive potential? J Immunother Cancer 2019; 7:325. [PMID: 31775882 PMCID: PMC6880594 DOI: 10.1186/s40425-019-0799-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/30/2019] [Indexed: 12/14/2022] Open
Abstract
The immunologic landscape of the host and tumor play key roles in determining how patients will benefit from immunotherapy, and a better understanding of these factors could help inform how well a tumor responds to treatment. Recent advances in immunotherapy and in our understanding of the immune system have revolutionized the treatment landscape for many advanced cancers. Notably, the use of immune checkpoint inhibitors has demonstrated durable responses in various malignancies. However, the response to such treatments is variable and currently unpredictable, the availability of predictive biomarkers is limited, and a substantial proportion of patients do not respond to immune checkpoint therapy. Identification and investigation of potential biomarkers that may predict sensitivity to immunotherapy is an area of active research. It is envisaged that a deeper understanding of immunity will aid in harnessing the full potential of immunotherapy, and allow appropriate patients to receive the most appropriate treatments. In addition to the identification of new biomarkers, the platforms and assays required to accurately and reproducibly measure biomarkers play a key role in ensuring consistency of measurement both within and between patients. In this review we discuss the current knowledge in the area of peripheral immune-based biomarkers, drawing information from the results of recent clinical studies of a number of different immunotherapy modalities in the treatment of cancer, including checkpoint inhibitors, bispecific antibodies, chimeric antigen receptor T cells, and anti-cancer vaccines. We also discuss the various technologies and approaches used in detecting and measuring circulatory biomarkers and the ongoing need for harmonization.
Collapse
Affiliation(s)
- Andrew B Nixon
- Duke University School of Medicine, Department of Medicine/Medical Oncology, 133 Jones Building, Research Drive, Durham, NC, 27710, USA.
| | - Kurt A Schalper
- Yale School of Medicine, Translational Immuno-Oncology Laboratory, Yale Cancer Center, 333 Cedar St. FMP117, New Haven, CT, 06520-8023, USA
| | - Ira Jacobs
- Pfizer Inc, Early Oncology Development and Clinical Research, 219 East 42nd St, New York, NY, 10017-5755, USA
| | - Shobha Potluri
- Pfizer Inc., Computational Biology, 230 E Grand Ave, South San Francisco, CA, 94080, USA
| | - I-Ming Wang
- Pfizer Inc., 10777 Science Center Dr., San Diego, CA, 92121, USA
| | - Catherine Fleener
- Pfizer Inc., Translational Oncology, La Jolla, CA, USA.,Present Address: Translational Science at Samumed, LLC, La Jolla, CA, USA
| |
Collapse
|