201
|
Dai Y, Jia L, Wang L, Sun H, Ji Y, Wang C, Song L, Liang S, Chen D, Feng Y, Bai X, Zhang D, Arai F, Chen H, Feng L. Magnetically Actuated Cell-Robot System: Precise Control, Manipulation, and Multimode Conversion. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105414. [PMID: 35233944 DOI: 10.1002/smll.202105414] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/04/2021] [Indexed: 06/14/2023]
Abstract
Border-nearing microrobots with self-propelling and navigating capabilities have promising applications in micromanipulation and bioengineering, because they can stimulate the surrounding fluid flow for object transportation. However, ensuring the biosafety of microrobots is a concurrent challenge in bioengineering applications. Here, macrophage template-based microrobots (cell robots) that can be controlled individually or in chain-like swarms are proposed, which can transport various objects. The cell robots are constructed using the phagocytic ability of macrophages to load nanomagnetic particles while maintaining their viability. The robots exhibit high position control accuracy and generate a flow field that can be used to transport microspheres and sperm when exposed to an external magnetic field near a wall. The cell robots can also form chain-like swarms to transport a large object (more than 100 times the volume). This new insight into the manipulation of macrophage-based cell robots provides a new concept by converting other biological cells into microrobots for micromanipulation in biomedical applications.
Collapse
Affiliation(s)
- Yuguo Dai
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
- Department of Mechanical Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Lina Jia
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Luyao Wang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Hongyan Sun
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Yiming Ji
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Chutian Wang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Li Song
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Shuzhang Liang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Dixiao Chen
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Yanmin Feng
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Xue Bai
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Deyuan Zhang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | - Fumihito Arai
- Department of Mechanical Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Huawei Chen
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | - Lin Feng
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| |
Collapse
|
202
|
Borna S, Lee E, Sato Y, Bacchetta R. Towards gene therapy for IPEX syndrome. Eur J Immunol 2022; 52:705-716. [PMID: 35355253 PMCID: PMC9322407 DOI: 10.1002/eji.202149210] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/29/2022] [Accepted: 03/29/2022] [Indexed: 11/30/2022]
Abstract
Immune dysregulation polyendocrinopathy enteropathy X linked (IPEX) syndrome is an uncurable disease of the immune system, with immune dysregulation that is caused by mutations in FOXP3. Current treatment options, such as pharmacological immune suppression and allogeneic hematopoietic stem cell transplantation, have been beneficial but present limitations, and their life‐long consequences are ill‐defined. Other similar blood monogenic diseases have been successfully treated using gene transfer in autologous patient cells, thus providing an effective and less invasive therapeutic. Development of gene therapy for patients with IPEX is particularly challenging because successful strategies must restore the complex expression profile of the transcription factor FOXP3, ensuring it is tightly regulated and its cell subset‐specific roles are maintained. This review summarizes current efforts toward achieving gene therapy to treat immune dysregulation in IPEX patients.
Collapse
Affiliation(s)
- Simon Borna
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Esmond Lee
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Yohei Sato
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rosa Bacchetta
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
203
|
Abedi MH, Yao MS, Mittelstein DR, Bar-Zion A, Swift MB, Lee-Gosselin A, Barturen-Larrea P, Buss MT, Shapiro MG. Ultrasound-controllable engineered bacteria for cancer immunotherapy. Nat Commun 2022; 13:1585. [PMID: 35332124 PMCID: PMC8948203 DOI: 10.1038/s41467-022-29065-2] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 02/16/2022] [Indexed: 12/25/2022] Open
Abstract
Rapid advances in synthetic biology are driving the development of genetically engineered microbes as therapeutic agents for a multitude of human diseases, including cancer. The immunosuppressive microenvironment of solid tumors, in particular, creates a favorable niche for systemically administered bacteria to engraft and release therapeutic payloads. However, such payloads can be harmful if released outside the tumor in healthy tissues where the bacteria also engraft in smaller numbers. To address this limitation, we engineer therapeutic bacteria to be controlled by focused ultrasound, a form of energy that can be applied noninvasively to specific anatomical sites such as solid tumors. This control is provided by a temperature-actuated genetic state switch that produces lasting therapeutic output in response to briefly applied focused ultrasound hyperthermia. Using a combination of rational design and high-throughput screening we optimize the switching circuits of engineered cells and connect their activity to the release of immune checkpoint inhibitors. In a clinically relevant cancer model, ultrasound-activated therapeutic microbes successfully turn on in situ and induce a marked suppression of tumor growth. This technology provides a critical tool for the spatiotemporal targeting of potent bacterial therapeutics in a variety of biological and clinical scenarios.
Collapse
Affiliation(s)
- Mohamad H Abedi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Department of Biochemistry, Institute for Protein Design and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Michael S Yao
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - David R Mittelstein
- Division of Engineering and Applied Sciences, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Avinoam Bar-Zion
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Margaret B Swift
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Audrey Lee-Gosselin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Pierina Barturen-Larrea
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Marjorie T Buss
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
204
|
Zhao X, Wan X, Huang T, Yao S, Wang S, Ding Y, Zhao Y, Li Z, Li L. Acidity-responsive nanocages as robust reactive oxygen species generators with butterfly effects for maximizing oxidative damage and enhancing cancer therapy. J Colloid Interface Sci 2022; 618:270-282. [PMID: 35339963 DOI: 10.1016/j.jcis.2022.03.084] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/12/2022]
Abstract
Recently, with the rational design of transition metal-containing nanoagents, chemodynamic therapy (CDT) has been developed and considered a promising method for cancer therapy through Fenton and Fenton-like reaction-induced hydroxyl radical (·OH) generation and cellular oxidative damage. However, it is still a great challenge to realize high reactive oxygen species (ROS) generation and therapeutic efficiency under the strict conditions of the tumor microenvironment (TME). Herein, we design and fabricate a TME-responsive core-shell nanocage composed of a CaCO3 nanolayer and a heterogeneous CoP core (CaCO3@CoP, CCP) with the synergy of CDT and calcium overload to maximize oxidative damage and enhance cancer therapy. The CaCO3 nanoshell is sensitive to pH and can be rapidly degraded upon endocytosis, leading to intracellular Ca2+ accumulation, which further triggers the production of mitochondrial ROS. Subsequently, the CoP hollow nanocage with fully exposed Co active sites has high Fenton-like reactive activity to produce ·OH and induce mitochondrial damage. Mitochondrial damage and ROS elevation, in turn, can modulate Ca2+ dynamics and augment calcium overload. The reciprocal interaction and loop feedback between calcium overload and photoenhanced ROS generation via photothermal therapy (PTT) can further trigger the immunogenic cell death (ICD) process to activate the maturation of dendritic cells (DCs), activation of cytotoxic and helper T cells, and excretion of proinflammatory cytokines to enhance antitumor immunity in vivo. With the butterfly effect, CCP finally brings forth a greatly enhanced cancer therapeutic outcome in murine models.
Collapse
Affiliation(s)
- Xingru Zhao
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xingyi Wan
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Tian Huang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; Center on Nanoenergy Research, School of physical Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Shuncheng Yao
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shaobo Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; Center on Nanoenergy Research, School of physical Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Yiming Ding
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; Center on Nanoenergy Research, School of physical Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Yunchao Zhao
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; Center on Nanoenergy Research, School of physical Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, PR China; Center on Nanoenergy Research, School of physical Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Linlin Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, PR China; School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing 100049, PR China; Center on Nanoenergy Research, School of physical Science and Technology, Guangxi University, Nanning 530004, PR China.
| |
Collapse
|
205
|
Hong DS, Butler MO, Pachynski RK, Sullivan R, Kebriaei P, Boross-Harmer S, Ghobadi A, Frigault MJ, Dumbrava EE, Sauer A, Brophy F, Navenot JM, Fayngerts S, Wolchinsky Z, Broad R, Batrakou DG, Wang R, Solis LM, Duose DY, Sanderson JP, Gerry AB, Marks D, Bai J, Norry E, Fracasso PM. Phase 1 Clinical Trial Evaluating the Safety and Anti-Tumor Activity of ADP-A2M10 SPEAR T-Cells in Patients With MAGE-A10+ Head and Neck, Melanoma, or Urothelial Tumors. Front Oncol 2022; 12:818679. [PMID: 35372008 PMCID: PMC8972123 DOI: 10.3389/fonc.2022.818679] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/18/2022] [Indexed: 02/02/2023] Open
Abstract
Background ADP-A2M10 specific peptide enhanced affinity receptor (SPEAR) T-cells are genetically engineered autologous T-cells that express a high-affinity melanoma-associated antigen (MAGE)-A10-specific T-cell receptor (TCR) targeting MAGE-A10-positive tumors in the context of human leukocyte antigen (HLA)-A*02. ADP-0022-004 is a phase 1, dose-escalation trial to evaluate the safety and anti-tumor activity of ADP-A2M10 in three malignancies (https://clinicaltrials.gov: NCT02989064). Methods Eligible patients were HLA-A*02 positive with advanced head and neck squamous cell carcinoma (HNSCC), melanoma, or urothelial carcinoma (UC) expressing MAGE-A10. Patients underwent apheresis; T-cells were isolated, transduced with a lentiviral vector containing the MAGE-A10 TCR, and expanded. Patients underwent lymphodepletion with fludarabine and cyclophosphamide prior to receiving ADP-A2M10. ADP-A2M10 was administered in two dose groups receiving 0.1×109 and >1.2 to 6×109 transduced cells, respectively, and an expansion group receiving 1.2 to 15×109 transduced cells. Results Ten patients (eight male and two female) with HNSCC (four), melanoma (three), and UC (three) were treated. Three patients were treated in each of the two dose groups, and four patients were treated in the expansion group. The most frequently reported adverse events grade ≥3 were leukopenia (10), lymphopenia (10), neutropenia (10), anemia (nine), and thrombocytopenia (five). Two patients reported cytokine release syndrome (one each with grade 1 and grade 3), with resolution. Best response included stable disease in four patients, progressive disease in five patients, and not evaluable in one patient. ADP-A2M10 cells were detectable in peripheral blood from patients in each dose group and the expansion group and in tumor tissues from patients in the higher dose group and the expansion group. Peak persistence was greater in patients from the higher dose group and the expansion group compared with the lower dose group. Conclusions ADP-A2M10 has shown an acceptable safety profile with no evidence of toxicity related to off-target binding or alloreactivity in these malignancies. Persistence of ADP-A2M10 in the peripheral blood and trafficking of ADP-A2M10 into the tumor was demonstrated. Because MAGE-A10 expression frequently overlaps with MAGE-A4 expression in tumors and responses were observed in the MAGE-A4 trial (NCT03132922), this clinical program closed, and trials with SPEAR T-cells targeting the MAGE-A4 antigen are ongoing.
Collapse
Affiliation(s)
- David S. Hong
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: David S. Hong,
| | - Marcus O. Butler
- Princess Margaret Hospital Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Russell K. Pachynski
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Ryan Sullivan
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Partow Kebriaei
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sarah Boross-Harmer
- Princess Margaret Hospital Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Armin Ghobadi
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Matthew J. Frigault
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Ecaterina E. Dumbrava
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amy Sauer
- Adaptimmune LLC, Philadelphia, PA, United States
| | | | | | | | | | - Robyn Broad
- Adaptimmune Limited, Abingdon, United Kingdom
| | | | - Ruoxi Wang
- Adaptimmune Limited, Abingdon, United Kingdom
| | - Luisa M. Solis
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dzifa Yawa Duose
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | - Diane Marks
- Adaptimmune LLC, Philadelphia, PA, United States
| | - Jane Bai
- Adaptimmune LLC, Philadelphia, PA, United States
| | - Elliot Norry
- Adaptimmune LLC, Philadelphia, PA, United States
| | | |
Collapse
|
206
|
Preclinical Evaluation of CAR T Cell Function: In Vitro and In Vivo Models. Int J Mol Sci 2022; 23:ijms23063154. [PMID: 35328572 PMCID: PMC8955360 DOI: 10.3390/ijms23063154] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 01/12/2023] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR) T cells is a rapidly emerging modality that engineers T cells to redirect tumor-specific cytotoxicity. CAR T cells have been well characterized for their efficacy against B cell malignancies, and rigorously studied in other types of tumors. Preclinical evaluation of CAR T cell function, including direct tumor killing, cytokine production, and memory responses, is crucial to the development and optimization of CAR T cell therapies. Such comprehensive examinations are usually performed in different types of models. Model establishment should focus on key challenges in the clinical setting and the capability to generate reliable data to indicate CAR T cell therapeutic potency in the clinic. Further, modeling the interaction between CAR T cells and tumor microenvironment provides additional insight for the future endeavors to enhance efficacy, especially against solid tumors. This review will summarize both in vitro and in vivo models for CAR T cell functional evaluation, including how they have evolved with the needs of CAR T cell research, the information they can provide for preclinical assessment of CAR T cell products, and recent technology advances to test CAR T cells in more clinically relevant models.
Collapse
|
207
|
Shafer P, Kelly LM, Hoyos V. Cancer Therapy With TCR-Engineered T Cells: Current Strategies, Challenges, and Prospects. Front Immunol 2022; 13:835762. [PMID: 35309357 PMCID: PMC8928448 DOI: 10.3389/fimmu.2022.835762] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/10/2022] [Indexed: 12/23/2022] Open
Abstract
To redirect T cells against tumor cells, T cells can be engineered ex vivo to express cancer-antigen specific T cell receptors (TCRs), generating products known as TCR-engineered T cells (TCR T). Unlike chimeric antigen receptors (CARs), TCRs recognize HLA-presented peptides derived from proteins of all cellular compartments. The use of TCR T cells for adoptive cellular therapies (ACT) has gained increased attention, especially as efforts to treat solid cancers with ACTs have intensified. In this review, we describe the differing mechanisms of T cell antigen recognition and signal transduction mediated through CARs and TCRs. We describe the classes of cancer antigens recognized by current TCR T therapies and discuss both classical and emerging pre-clinical strategies for antigen-specific TCR discovery, enhancement, and validation. Finally, we review the current landscape of clinical trials for TCR T therapy and discuss what these current results indicate for the development of future engineered TCR approaches.
Collapse
Affiliation(s)
- Paul Shafer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Program in Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Lauren M. Kelly
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Program in Cancer & Cell Biology, Baylor College of Medicine, Houston, TX, United States
| | - Valentina Hoyos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
208
|
Emerging CAR T Cell Strategies for the Treatment of AML. Cancers (Basel) 2022; 14:cancers14051241. [PMID: 35267549 PMCID: PMC8909045 DOI: 10.3390/cancers14051241] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Chimeric antigen receptors (CARs) targeting CD19 have emerged as a new treatment for hematological malignancies. As a “living therapy”, CARs can precisely target and eliminate tumors while proliferating inside the patient’s body. Various preclinical and clinical studies are ongoing to identify potential CAR-T cell targets for acute myeloid leukemia (AML). We shed light on the continuing efforts of CAR development to overcome tumor escape, exhaustion, and toxicities. Furthermore, we summarize the recent progress of a range of putative targets exploring this unmet need to treat AML. Lastly, we discuss the advances in preclinical models that built the foundation for ongoing clinical trials. Abstract Engineered T cells expressing chimeric antigen receptors (CARs) on their cell surface can redirect antigen specificity. This ability makes CARs one of the most promising cancer therapeutic agents. CAR-T cells for treating patients with B cell hematological malignancies have shown impressive results. Clinical manifestation has yielded several trials, so far five CAR-T cell therapies have received US Food and Drug Administration (FDA) approval. However, emerging clinical data and recent findings have identified some immune-related toxicities due to CAR-T cell therapy. Given the outcome and utilization of the same proof of concept, further investigation in other hematological malignancies, such as leukemias, is warranted. This review discusses the previous findings from the pre-clinical and human experience with CAR-T cell therapy. Additionally, we describe recent developments of novel targets for adoptive immunotherapy. Here we present some of the early findings from the pre-clinical studies of CAR-T cell modification through advances in genetic engineering, gene editing, cellular programming, and formats of synthetic biology, along with the ongoing efforts to restore the function of exhausted CAR-T cells through epigenetic remodeling. We aim to shed light on the new targets focusing on acute myeloid leukemia (AML).
Collapse
|
209
|
Leveraging biomaterials for enhancing T cell immunotherapy. J Control Release 2022; 344:272-288. [PMID: 35217099 DOI: 10.1016/j.jconrel.2022.02.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022]
Abstract
The dynamic roles of T cells in the immune system to recognize and destroy the infected or mutated cells render T cell therapy a prospective treatment for a variety of diseases including cancer, autoimmune diseases, and allograft rejection. However, the clinical applications of T cell therapy remain unsatisfactory due to the tedious manufacturing process, off-target cytotoxicity, poor cell persistence, and associated adverse effects. To this end, various biomaterials have been introduced to enhance T cell therapy by facilitating proliferation, enhancing local enrichment, prolonging retention, and alleviating side effects. This review highlights the design strategies of biomaterials developed for T cell expansion, enrichment, and delivery as well as their corresponding therapeutic effects. The prospects of biomaterials for enhancing T cell immunotherapy are also discussed in this review.
Collapse
|
210
|
Xue Y, Che J, Ji X, Li Y, Xie J, Chen X. Recent advances in biomaterial-boosted adoptive cell therapy. Chem Soc Rev 2022; 51:1766-1794. [PMID: 35170589 DOI: 10.1039/d1cs00786f] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adoptive immunotherapies based on the transfer of functional immune cells hold great promise in treating a wide range of malignant diseases, especially cancers, autoimmune diseases, and infectious diseases. However, manufacturing issues and biological barriers lead to the insufficient population of target-selective effector cells at diseased sites after adoptive transfer, hindering effective clinical translation. The convergence of immunology, cellular biology, and materials science lays a foundation for developing biomaterial-based engineering platforms to overcome these challenges. Biomaterials can be rationally designed to improve ex vivo immune cell expansion, expedite functional engineering, facilitate protective delivery of immune cells in situ, and navigate the infused cells in vivo. Herein, this review presents a comprehensive summary of the latest progress in biomaterial-based strategies to enhance the efficacy of adoptive cell therapy, focusing on function-specific biomaterial design, and also discusses the challenges and prospects of this field.
Collapse
Affiliation(s)
- Yonger Xue
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu 210009, China. .,Center for BioDelivery Sciences, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai, 200240, P. R. China.,Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki 210-0821, Japan
| | - Junyi Che
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xuemei Ji
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yunuo Li
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu 210009, China.
| | - Jinbing Xie
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu 210009, China. .,Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki 210-0821, Japan.,State Key Laboratory of Bioelectronics, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu 210009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore. .,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
211
|
Fåne A, Myhre MR, Inderberg EM, Wälchli S. In vivo experimental mouse model to test CD19CAR T cells generated with different methods. Methods Cell Biol 2022; 167:149-161. [PMID: 35152992 DOI: 10.1016/bs.mcb.2021.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Pre-clinical evaluation of CAR T cells includes procedures testing T-cell efficacy and safety in as close to real world conditions as possible. An important step in efficacy testing is the in vivo study, most often using immunodeficient murine models into which both the poison and the cure are injected; namely a human cancer cell line and genetically modified human T cells. The capacity of the T cells to control the cancer progression will provide information about the CAR T-cell efficacy. We here provide a protocol to test CAR T cells in vivo using the validated anti-B-cell marker CD19CAR and an aggressive lymphoma model.
Collapse
Affiliation(s)
- Anne Fåne
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Marit Renée Myhre
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sebastien Wälchli
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
212
|
Li Y, Che J, Chang L, Guo M, Bao X, Mu D, Sun X, Zhang X, Lu W, Xie J. CD47- and Integrin α4/β1-Comodified-Macrophage-Membrane-Coated Nanoparticles Enable Delivery of Colchicine to Atherosclerotic Plaque. Adv Healthc Mater 2022; 11:e2101788. [PMID: 34786845 DOI: 10.1002/adhm.202101788] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/26/2021] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease and the major pathological factor of most cardiovascular diseases, leading to ≈1/3 of deaths worldwide. Improving local delivery of anti-inflammatory drugs to the site of atherosclerosis has significant promise to prevent the development of atherosclerotic plaque clinically. Here, a modified-macrophage-membrane-coated nanoparticle drug delivery able to transport colchicine to the atherosclerotic site is reported. This hybrid system efficiently targets endothelial cells under an inflammatory environment while escaping the endocytosis of macrophages. Furthermore, the anti-inflammatory effect of the modified-macrophage-membrane-coated nanoparticles on foam cells is studied. In vivo, the migration of the modified-macrophage-membrane-coated nanoparticles to atherosclerotic lesions is confirmed in a vulnerable atherosclerotic plaque mouse model. Intravenous injections of the hybrid system successfully reduce the lipid plaque load and improve the plaque stability. This strategy provides a potential therapeutic system for the targeted delivery of anti-inflammatory drugs to the atherosclerotic site for the treatment of atherosclerosis in cardiovascular diseases.
Collapse
Affiliation(s)
- Yuyu Li
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Junyi Che
- Institute of Translational Medicine Department of Science and Technology Drum Tower Hospital Medical School of Nanjing University Nanjing 211800 China
| | - Lei Chang
- Department of Cardiology Affiliated Nanjing Drum Tower Hospital of Nanjing Medical University Nanjing 211800 China
| | - Meng Guo
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Xue Bao
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Dan Mu
- Department of Radiology Drum Tower Hospital Medical School of Nanjing University Nanjing 211800 China
| | - Xuan Sun
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Xin Zhang
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Wen Lu
- Department of Cardiology Xu Zhou Central Hospital Xu Zhou 221009 China
| | - Jun Xie
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| |
Collapse
|
213
|
Cell-based immunotherapies in gynecologic cancers. Curr Opin Obstet Gynecol 2022; 34:10-14. [PMID: 34967809 DOI: 10.1097/gco.0000000000000760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review provides an update on recent developments in cell-based immunotherapy in gynecologic cancers. RECENT FINDINGS Chimeric antigen receptor (CAR) technology has made significant progress allowing now for not only expressing CARs on T-cells, but also on other immune effector cells, such as natural killer cells and macrophages. Cell-based vaccines have started to show promising results in clinical trials. SUMMARY Cell-based immunotherapies in gynecologic cancers continue to evolve with promising clinical efficacy in select patients.
Collapse
|
214
|
Blumenschein GR, Devarakonda S, Johnson M, Moreno V, Gainor J, Edelman MJ, Heymach JV, Govindan R, Bachier C, Doger de Spéville B, Frigault MJ, Olszanski AJ, Lam VK, Hyland N, Navenot JM, Fayngerts S, Wolchinsky Z, Broad R, Batrakou D, Pentony MM, Sanderson JP, Gerry A, Marks D, Bai J, Holdich T, Norry E, Fracasso PM. Phase I clinical trial evaluating the safety and efficacy of ADP-A2M10 SPEAR T cells in patients with MAGE-A10 + advanced non-small cell lung cancer. J Immunother Cancer 2022; 10:jitc-2021-003581. [PMID: 35086946 PMCID: PMC8796260 DOI: 10.1136/jitc-2021-003581] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2021] [Indexed: 02/06/2023] Open
Abstract
Background ADP-A2M10 specific peptide enhanced affinity receptor (SPEAR) T cells (ADP-A2M10) are genetically engineered autologous T cells that express a high-affinity melanoma-associated antigen A10 (MAGE-A10)-specific T-cell receptor (TCR) targeting MAGE-A10+ tumors in the context of human leukocyte antigen (HLA)-A*02. ADP-0022-003 was a phase I dose-escalation trial that aimed to evaluate the safety and antitumor activity of ADP-A2M10 in non-small cell lung cancer (NSCLC) (NCT02592577). Methods Eligible patients were HLA-A*02 positive with advanced NSCLC expressing MAGE-A10. Patients underwent apheresis; T cells were isolated, transduced with a lentiviral vector containing the TCR targeting MAGE-A10, and expanded. Patients underwent lymphodepletion with varying doses/schedules of fludarabine and cyclophosphamide prior to receiving ADP-A2M10. ADP-A2M10 were administered at 0.08–0.12×109 (dose group 1), 0.5–1.2×109 (dose group 2), and 1.2–15×109 (dose group 3/expansion) transduced cells. Results Eleven patients (male, n=6; female, n=5) with NSCLC (adenocarcinoma, n=8; squamous cell carcinoma, n=3) were treated. Five, three, and three patients received cells in dose group 1, dose group 2, and dose group 3/expansion, respectively. The most frequently reported grade ≥3 adverse events were lymphopenia (n=11), leukopenia (n=10), neutropenia (n=8), anemia (n=6), thrombocytopenia (n=5), and hyponatremia (n=5). Three patients presented with cytokine release syndrome (grades 1, 2, and 4, respectively). One patient received the highest dose of lymphodepletion (fludarabine 30 mg/m2 on days –5 to –2 and cyclophosphamide 1800 mg/m2 on days −5 to −4) prior to a second infusion of ADP-A2M10 and had a partial response, subsequently complicated by aplastic anemia and death. Responses included: partial response (after second infusion; one patient), stable disease (four patients), clinical or radiographic progressive disease (five patients), and not evaluable (one patient). ADP-A2M10 were detectable in peripheral blood and in tumor tissue. Peak persistence was higher in patients who received higher doses of ADP-A2M10. Conclusions ADP-A2M10 demonstrated an acceptable safety profile and no evidence of toxicity related to off-target binding or alloreactivity. There was persistence of ADP-A2M10 in peripheral blood as well as ADP-A2M10 trafficking into the tumor. Given the discovery that MAGE-A10 and MAGE-A4 expression frequently overlap, this clinical program closed as trials with SPEAR T cells targeting MAGE-A4 are ongoing.
Collapse
Affiliation(s)
- George R Blumenschein
- Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Melissa Johnson
- Lung Cancer Research and Drug Development, Sarah Cannon Research Institute at Tennessee Oncology, Nashville, Tennessee, USA
| | - Victor Moreno
- START Madrid-FJD, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Justin Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Martin J Edelman
- Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - John V Heymach
- Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ramaswamy Govindan
- Medical Oncology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Carlos Bachier
- Hematology, Sarah Cannon Center for Blood Cancer at TriStar Centennial, Nashville, Tennessee, USA
| | | | - Matthew J Frigault
- Bone Marrow Transplant & Cellular Therapy, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony J Olszanski
- Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Vincent K Lam
- Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | - Robyn Broad
- Adaptimmune, Milton Park, Abingdon, Oxfordshire, UK
| | | | | | | | - Andrew Gerry
- Adaptimmune, Milton Park, Abingdon, Oxfordshire, UK
| | - Diane Marks
- Adaptimmune, Philadelphia, Pennsylvania, USA
| | - Jane Bai
- Adaptimmune, Philadelphia, Pennsylvania, USA
| | - Tom Holdich
- Adaptimmune, Milton Park, Abingdon, Oxfordshire, UK
| | | | | |
Collapse
|
215
|
Kyte JA. Strategies for Improving the Efficacy of CAR T Cells in Solid Cancers. Cancers (Basel) 2022; 14:cancers14030571. [PMID: 35158839 PMCID: PMC8833730 DOI: 10.3390/cancers14030571] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cell therapy with genetically retargeted T cells shows strong clinical efficacy against leukaemia and lymphoma. To make this therapy efficient against solid cancers, a series of hurdles must be addressed. This includes the need to enable the T cells to survive long term in patients and to overcome immunosuppressive mechanisms in the tumour. Further, it is essential to prevent tumour cells from escaping by losing the protein that is recognised by the infused cells. The present article provides an overview of the key strategies that are currently being investigated to overcome these hurdles. A series of approaches have been described in preclinical models, but these remain untested in patients. The further progress of the field will depend on evaluating more strategies in a proper clinical setting. Abstract Therapy with T cells equipped with chimeric antigen receptors (CARs) shows strong efficacy against leukaemia and lymphoma, but not yet against solid cancers. This has been attributed to insufficient T cell persistence, tumour heterogeneity and an immunosuppressive tumour microenvironment. The present article provides an overview of key strategies that are currently investigated to overcome these hurdles. Basic aspects of CAR design are revisited, relevant for tuning the stimulatory signal to the requirements of solid tumours. Novel approaches for enhancing T cell persistence are highlighted, based on epigenetic or post-translational modifications. Further, the article describes CAR T strategies that are being developed for overcoming tumour heterogeneity and the escape of cancer stem cells, as well as for countering prevalent mechanisms of immune suppression in solid cancers. In general, personalised medicine is faced with a lack of drugs matching the patient’s profile. The advances and flexibility of modern gene engineering may allow for the filling of some of these gaps with tailored CAR T approaches addressing mechanisms identified as important in the individual patient. At this point, however, CAR T cell therapy remains unproved in solid cancers. The further progress of the field will depend on bringing novel strategies into clinical evaluation, while maintaining safety.
Collapse
Affiliation(s)
- Jon Amund Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Mail Box 4950 Nydalen, 0424 Oslo, Norway;
- Department of Clinical Cancer Research, Oslo University Hospital, Mail Box 4950 Nydalen, 0424 Oslo, Norway
| |
Collapse
|
216
|
Development and clinical translation of ex vivo gene therapy. Comput Struct Biotechnol J 2022; 20:2986-3003. [PMID: 35782737 PMCID: PMC9218169 DOI: 10.1016/j.csbj.2022.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/27/2022] Open
Abstract
Retroviral gene therapy has emerged as a promising therapeutic modality for multiple inherited and acquired human diseases. The capability of delivering curative treatment or mediating therapeutic benefits for a long-term period following a single application fundamentally distinguishes this medical intervention from traditional medicine and various lentiviral/γ-retroviral vector-mediated gene therapy products have been approved for clinical use. Continued advances in retroviral vector engineering, genomic editing, synthetic biology and immunology will broaden the medical applications of gene therapy and improve the efficacy and safety of the treatments based on genetic correction and alteration. This review will summarize the advent and clinical translation of ex vivo gene therapy, with the focus on the milestones during the exploitation of genetically engineered hematopoietic stem cells (HSCs) tackling a variety of pathological conditions which led to marketing approval. Finally, current statue and future prospects of gene editing as an alternative therapeutic approach are also discussed.
Collapse
|
217
|
Carlberg C, Velleuer E. Cancer Immunology. Mol Immunol 2022. [DOI: 10.1007/978-3-031-04025-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
218
|
Lin ECY, Chen SW, Chen LK, Lin TA, Wu YX, Juan CC, Chang YI. Glucosamine Interferes With Myelopoiesis and Enhances the Immunosuppressive Activity of Myeloid-Derived Suppressor Cells. Front Nutr 2021; 8:762363. [PMID: 34901113 PMCID: PMC8660085 DOI: 10.3389/fnut.2021.762363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/19/2021] [Indexed: 12/16/2022] Open
Abstract
Glucosamine (GlcN) is the most widely consumed dietary supplement and exhibits anti-inflammatory effects. However, the influence of GlcN on immune cell generation and function is largely unclear. In this study, GlcN was delivered into mice to examine its biological function in hematopoiesis. We found that GlcN promoted the production of immature myeloid cells, known as myeloid-derived suppressor cells (MDSCs), both in vivo and in vitro. Additionally, GlcN upregulated the expression of glucose transporter 1 in hematopoietic stem and progenitor cells (HSPCs), influenced HSPC functions, and downregulated key genes involved in myelopoiesis. Furthermore, GlcN increased the expression of arginase 1 and inducible nitric oxide synthase to produce high levels of reactive oxygen species, which was regulated by the STAT3 and ERK1/2 pathways, to increase the immunosuppressive ability of MDSCs. We revealed a novel role for GlcN in myelopoiesis and MDSC activity involving a potential link between GlcN and immune system, as well as the new therapeutic benefit.
Collapse
Affiliation(s)
- Eric Chang-Yi Lin
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Shuoh-Wen Chen
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Luen-Kui Chen
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Ting-An Lin
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan.,Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Yu-Xuan Wu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Chi-Chang Juan
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Yuan-I Chang
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| |
Collapse
|
219
|
Kouakanou L, Peters C, Brown CE, Kabelitz D, Wang LD. Vitamin C, From Supplement to Treatment: A Re-Emerging Adjunct for Cancer Immunotherapy? Front Immunol 2021; 12:765906. [PMID: 34899716 PMCID: PMC8663797 DOI: 10.3389/fimmu.2021.765906] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Vitamin C (VitC), in addition to its role as a general antioxidant, has long been considered to possess direct anti-cancer activity at high doses. VitC acts through oxidant and epigenetic mechanisms, which at high doses can exert direct killing of tumor cells in vitro and delay tumor growth in vivo. Recently, it has also been shown that pharmacologic-dose VitC can contribute to control of tumors by modulating the immune system, and studies have been done interrogating the role of physiologic-dose VitC on novel adoptive cellular therapies (ACTs). In this review, we discuss the effects of VitC on anti-tumor immune cells, as well as the mechanisms underlying those effects. We address important unanswered questions concerning both VitC and ACTs, and outline challenges and opportunities facing the use of VitC in the clinical setting as an adjunct to immune-based anti-cancer therapies.
Collapse
Affiliation(s)
- Léonce Kouakanou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Christian Peters
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christine E Brown
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Leo D Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States.,Department of Pediatrics, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
220
|
Cell therapy products are revolutionizing multiple therapeutic areas; to maintain this pace of innovation, manufacturing solutions must adapt. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
221
|
Turk OM, Woodall RC, Gutova M, Brown CE, Rockne RC, Munson JM. Delivery strategies for cell-based therapies in the brain: overcoming multiple barriers. Drug Deliv Transl Res 2021; 11:2448-2467. [PMID: 34718958 PMCID: PMC8987295 DOI: 10.1007/s13346-021-01079-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Cell-based therapies to the brain are promising for the treatment of multiple brain disorders including neurodegeneration and cancers. In order to access the brain parenchyma, there are multiple physiological barriers that must be overcome depending on the route of delivery. Specifically, the blood-brain barrier has been a major difficulty in drug delivery for decades, and it still presents a challenge for the delivery of therapeutic cells. Other barriers, including the blood-cerebrospinal fluid barrier and lymphatic-brain barrier, are less explored, but may offer specific challenges or opportunities for therapeutic delivery. Here we discuss the barriers to the brain and the strategies currently in place to deliver cell-based therapies, including engineered T cells, dendritic cells, and stem cells, to treat diseases. With a particular focus on cancers, we also highlight the current ongoing clinical trials that use cell-based therapies to treat disease, many of which show promise at treating some of the deadliest illnesses.
Collapse
Affiliation(s)
- Olivia M Turk
- Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | - Ryan C Woodall
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, City of Hope, Duarte, CA, USA
| | - Margarita Gutova
- Department of Stem Cell Biology and Regenerative Medicine, City of Hope, Duarte, CA, USA
| | - Christine E Brown
- Departments of Hematology & Hematopoietic Cell Transplantation and Immuno-Oncology, City of Hope, Duarte, CA, USA
| | - Russell C Rockne
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, City of Hope, Duarte, CA, USA
| | - Jennifer M Munson
- Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, VA, USA.
| |
Collapse
|
222
|
Hewitt MM, Trainor N, Ostrout N, Abraham E. Cell therapy manufacturing: process analytic technologies needed to achieve flexible, feedback-driven automation. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
223
|
The Cancer Surfaceome Atlas integrates genomic, functional and drug response data to identify actionable targets. NATURE CANCER 2021; 2:1406-1422. [PMID: 35121907 PMCID: PMC9940627 DOI: 10.1038/s43018-021-00282-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023]
Abstract
Cell-surface proteins (SPs) are a rich source of immune and targeted therapies. By systematically integrating single-cell and bulk genomics, functional studies and target actionability, in the present study we comprehensively identify and annotate genes encoding SPs (GESPs) pan-cancer. We characterize GESP expression patterns, recurrent genomic alterations, essentiality, receptor-ligand interactions and therapeutic potential. We also find that mRNA expression of GESPs is cancer-type specific and positively correlates with protein expression, and that certain GESP subgroups function as common or specific essential genes for tumor cell growth. We also predict receptor-ligand interactions substantially deregulated in cancer and, using systems biology approaches, we identify cancer-specific GESPs with therapeutic potential. We have made this resource available through the Cancer Surfaceome Atlas ( http://fcgportal.org/TCSA ) within the Functional Cancer Genome data portal.
Collapse
|
224
|
The Challenges and Opportunities in the Development of MicroRNA Therapeutics: A Multidisciplinary Viewpoint. Cells 2021; 10:cells10113097. [PMID: 34831320 PMCID: PMC8619171 DOI: 10.3390/cells10113097] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/22/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023] Open
Abstract
microRNAs (miRs) are emerging as attractive therapeutic targets because of their small size, specific targetability, and critical role in disease pathogenesis. However, <20 miR targeting molecules have entered clinical trials, and none progressed to phase III. The difficulties in miR target identification, the moderate efficacy of miR inhibitors, cell type-specific delivery, and adverse outcomes have impeded the development of miR therapeutics. These hurdles are rooted in the functional complexity of miR's role in disease and sequence complementarity-dependent/-independent effects in nontarget tissues. The advances in understanding miR's role in disease, the development of efficient miR inhibitors, and innovative delivery approaches have helped resolve some of these hurdles. In this review, we provide a multidisciplinary viewpoint on the challenges and opportunities in the development of miR therapeutics.
Collapse
|
225
|
Murty T, Mackall CL. Gene editing to enhance the efficacy of cancer cell therapies. Mol Ther 2021; 29:3153-3162. [PMID: 34673274 PMCID: PMC8571170 DOI: 10.1016/j.ymthe.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/18/2022] Open
Abstract
Adoptive T cell therapies have shown impressive signals of activity, but their clinical impact could be enhanced by technologies to increase T cell potency and diminish the cost and labor involved in manufacturing these products. Gene editing platforms are under study in this arena to (1) enhance immune cell potency by knocking out molecules that inhibit immune responses; (2) deliver genetic payloads into precise genomic locations and thereby enhance safety and/or improve the gene expression profile by leveraging physiologic promoters, enhancers, and repressors; and (3) enable off-the-shelf therapies by preventing alloreactivity and immune rejection. This review discusses gene editing approaches that have been the best studied in the context of human T cells and adoptive T cell therapies, summarizing their current status and near-term potential for translation.
Collapse
Affiliation(s)
- Tara Murty
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA; Program in Biophysics, Stanford University, Stanford, CA, USA; Medical Scientist Training Program, Stanford University, Stanford, CA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
226
|
Kalinin RS, Ukrainskaya VM, Chumakov SP, Moysenovich AM, Tereshchuk VM, Volkov DV, Pershin DS, Maksimov EG, Zhang H, Maschan MA, Rubtsov YP, Stepanov AV. Engineered Removal of PD-1 From the Surface of CD19 CAR-T Cells Results in Increased Activation and Diminished Survival. Front Mol Biosci 2021; 8:745286. [PMID: 34722633 PMCID: PMC8548718 DOI: 10.3389/fmolb.2021.745286] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
CAR-T cell therapy is the most advanced way to treat therapy resistant hematologic cancers, in particular B cell lymphomas and leukemias, with high efficiency. Donor T cells equipped ex vivo with chimeric receptor recognize target tumor cells and kill them using lytic granules. CAR-T cells that recognize CD19 marker of B cells (CD19 CAR-T) are considered the gold standard of CAR-T therapy and are approved by FDA. But in some cases, CD19 CAR-T cell therapy fails due to immune suppressive microenvironment. It is shown that tumor cells upregulate expression of PD-L1 surface molecule that binds and increases level and signal provided by PD-1 receptor on the surface of therapeutic CAR-T cells. Induction of this negative signaling results in functional impairment of cytotoxic program in CAR-T cells. Multiple attempts were made to block PD-1 signaling by reducing binding or surface level of PD-1 in CAR-T cells by various means. In this study we co-expressed CD19-CAR with PD-1-specific VHH domain of anti-PD-1 nanobody to block PD-1/PD-L1 signaling in CD19 CAR-T cells. Unexpectedly, despite increased activation of CAR-T cells with low level of PD-1, these T cells had reduced survival and diminished cytotoxicity. Functional impairment caused by disrupted PD-1 signaling was accompanied by faster maturation and upregulation of exhaustion marker TIGIT in CAR-T cells. We conclude that PD-1 in addition to its direct negative effect on CAR-induced signaling is required for attenuation of strong stimulation leading to cell death and functional exhaustion. These observations suggest that PD-1 downregulation should not be considered as the way to improve the quality of therapeutic CAR-T cells.
Collapse
Affiliation(s)
- R S Kalinin
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - V M Ukrainskaya
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - S P Chumakov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - A M Moysenovich
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - V M Tereshchuk
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - D V Volkov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - D S Pershin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - E G Maksimov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - H Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, China
| | - M A Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Y P Rubtsov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - A V Stepanov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia.,Department of Chemistry, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
227
|
Ma L, Sakamoto Y, Ando K, Fujita H, Takahashi A, Takeshima T, Otsuka H, Ebner DK, Kakimi K, Imai T, Shimokawa T. Th balance related host genetic background affects the therapeutic effects of combining carbon-ion radiotherapy with dendritic cell immunotherapy. Int J Radiat Oncol Biol Phys 2021; 112:780-789. [PMID: 34740767 DOI: 10.1016/j.ijrobp.2021.10.141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE The goal of this study is to clarify the underlying mechanisms of metastasis suppression by CiDC (carbon-ion radiotherapy (CIRT) combined with immature dendritic cell (iDC) immunotherapy), which was previously shown to significantly suppress pulmonary metastasis in a NR-S1-bearing C3H/He mouse model. METHODS AND MATERIALS Mouse carcinoma cell lines (LLC, LM8, Colon-26 and Colon-26MGS) were grafted into the right hind paw of syngeneic mice (C57BL/6J, C3H/He and BALB/c). Seven days later, the tumors on the mice were locally irradiated with carbon-ions (290 MeV/n, 6 cm SOBP, 1 or 2 Gy). At 1.5 days after irradiation, bone marrow-derived immature dendritic cells were administrated intravenously into a subset of the mice. The number of lung metastases was evaluated within three weeks after irradiation. In vitro cultured cancer cells were irradiated with carbon-ion (290 MeV/n, mono-energy, LET approximately 70 ∼ 80 keV/µm), and then co-cultured with iDCs for three days to determine the DC maturation. RESULTS CiDC effectively repressed distant lung metastases in cancer cell (LLC and LM8)-bearing C57BL/6J and C3H/He mouse models. However, Colon-26 and Colon-26MGS-bearing BALB/c models did not show enhancement of metastasis suppression by combination treatment. This was further evaluated by comparing LM8-bearing C3H/He and LLC-bearing C57BL/6J models with a Colon-26-bearing BALB/c model. In vitro co-culture assays demonstrated that all irradiated cell lines were able to activate C3H/He or C57BL/6J-derived iDCs into mature DCs, but not BALB/c-derived iDCs. CONCLUSION The genetic background of the host may have a strong impact on the potency of combination therapy. Future animal and clinical testing should evaluate host genetic factors when evaluating treatment efficacy.
Collapse
Affiliation(s)
- Liqiu Ma
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; Gunma University Heavy Ion Medical Center, Gunma, Japan; China Institute of Atomic Energy, Beijing 102413, China
| | - Yoshimitsu Sakamoto
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ken Ando
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; Department of Radiation Oncology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hidetoshi Fujita
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | | | - Tsuguhide Takeshima
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hiromi Otsuka
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Daniel K Ebner
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo, Tokyo, Japan
| | - Takashi Imai
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takashi Shimokawa
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| |
Collapse
|
228
|
Wu J, Chen Z, Wickström SL, Gao J, He X, Jing X, Wu J, Du Q, Yang M, Chen Y, Zhang D, Yin X, Guo Z, Jensen L, Yang Y, Tao W, Lundqvist A, Kiessling R, Cao Y. Interleukin-33 is a Novel Immunosuppressor that Protects Cancer Cells from TIL Killing by a Macrophage-Mediated Shedding Mechanism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101029. [PMID: 34486239 PMCID: PMC8564439 DOI: 10.1002/advs.202101029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/22/2021] [Indexed: 02/05/2023]
Abstract
Recognition of specific antigens expressed in cancer cells is the initial process of cytolytic T cell-mediated cancer killing. However, this process can be affected by other non-cancerous cellular components in the tumor microenvironment. Here, it is shown that interleukin-33 (IL-33)-activated macrophages protect melanoma cells from tumor-infiltrating lymphocyte-mediated killing. Mechanistically, IL-33 markedly upregulates metalloprotease 9 (MMP-9) expression in macrophages, which acts as a sheddase to trim NKG2D, an activating receptor expressed on the surface of natural killer (NK) cells, CD8+ T cells, subsets of CD4+ T cells, iNKT cells, and γδ T cells. Further, MMP-9 also cleaves the MHC class I molecule, cell surface antigen-presenting complex molecules, expressed in melanoma cells. Consequently, IL-33-induced macrophage MMP-9 robustly mitigates the tumor killing-effect by T cells. Genetic and pharmacological loss-of-function of MMP-9 sheddase restore T cell-mediated cancer killing. Together, these data provide compelling in vitro and in vivo evidence showing novel mechanisms underlying the IL-33-macrophage-MMP-9 axis-mediated immune tolerance against cancer cells. Targeting each of these signaling components, including IL-33 and MMP-9 provides a new therapeutic paradigm for improving anticancer efficacy by immune therapy.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Histocompatibility Antigens Class I/metabolism
- Humans
- Immunity/drug effects
- Interleukin-33/pharmacology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/metabolism
- Lymphocytes, Tumor-Infiltrating/cytology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Matrix Metalloproteinase 9/chemistry
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinase 9/metabolism
- Melanoma/immunology
- Melanoma/therapy
- Mice
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Neoplasms/immunology
- Neoplasms/therapy
- RNA Interference
- RNA, Small Interfering/metabolism
- Up-Regulation/drug effects
- Zebrafish
Collapse
Affiliation(s)
- Jing Wu
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 65Sweden
- Department of PharmacyThe Second Hospital of Shandong UniversityJinanShandong250000China
| | - Ziqing Chen
- Department of Oncology and PathologyKarolinska InstituteStockholm171 77Sweden
| | - Stina L. Wickström
- Department of Oncology and PathologyKarolinska InstituteStockholm171 77Sweden
| | - Juan Gao
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 65Sweden
| | - Xingkang He
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 65Sweden
- Institute of GastroenterologyZhejiang UniversityHangzhou310016China
| | - Xu Jing
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 65Sweden
| | - Jieyu Wu
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 65Sweden
| | - Qiqiao Du
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 65Sweden
| | - Muyi Yang
- Department of Oncology and PathologyKarolinska InstituteStockholm171 77Sweden
| | - Yi Chen
- Department of Oncology and PathologyKarolinska InstituteStockholm171 77Sweden
| | - Dingding Zhang
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 65Sweden
- School of MedicineSichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu611731China
| | - Xin Yin
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 65Sweden
| | - Ziheng Guo
- Department of Pancreatic SurgeryWest China HospitalSichuan UniversityChengduSichuan610045China
| | - Lasse Jensen
- Division of Cardiovascular MedicineDepartment of Medical and Health SciencesLinkoping UniversitySweden
| | - Yunlong Yang
- Department of Cellular and Genetic MedicineSchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Wei Tao
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Andreas Lundqvist
- Department of Oncology and PathologyKarolinska InstituteStockholm171 77Sweden
| | - Rolf Kiessling
- Department of Oncology and PathologyKarolinska InstituteStockholm171 77Sweden
- Karolinska University HospitalSolnaStockholm171 64Sweden
| | - Yihai Cao
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstituteStockholm171 65Sweden
| |
Collapse
|
229
|
Liu C, Qi T, Milner JJ, Lu Y, Cao Y. Speed and Location Both Matter: Antigen Stimulus Dynamics Controls CAR-T Cell Response. Front Immunol 2021; 12:748768. [PMID: 34691062 PMCID: PMC8531752 DOI: 10.3389/fimmu.2021.748768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 12/18/2022] Open
Abstract
Despite the success in B-cell malignancies, chimeric antigen receptor (CAR)-T cell therapies have not yet demonstrated consistent efficacy across all patients and tumor types, particularly against solid tumors. Higher rates of T cell exhaustion are associated with inferior clinical outcomes following CAR-T cell therapy, which is prevalent in solid tumors. T cell exhaustion may originate from persistent and chronic antigen stimulation by tumor cells that resist and/or evade T cell-mediated killing. We exploited CAR-T exhaustion with a classic negative feedback model (incoherent feedforward loop, IFFL) to investigate the balance between CAR-T cell activation and exhaustion under different antigen presentation dynamics. Built upon the experimental and clinical data, we hypothesize that the speed and anatomical location of antigenic stimulation are both crucial to CAR-T cell response. Chronic antigenic stimulation as well as the harsh tumor microenvironment present multiple barriers to CAR-T cell efficacy in solid tumors. Many therapeutic strategies are individually insufficient to improve of CAR-T responses against solid tumors, as they clear but one of the many barriers CAR-T cells face in solid tumors. A combination strategy targeting multiple barriers holds promise to improve CAR-T therapy in solid tumors.
Collapse
Affiliation(s)
- Can Liu
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Timothy Qi
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - J. Justin Milner
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yong Lu
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
230
|
Jiang H, Fu D, Bidgoli A, Paczesny S. T Cell Subsets in Graft Versus Host Disease and Graft Versus Tumor. Front Immunol 2021; 12:761448. [PMID: 34675938 PMCID: PMC8525316 DOI: 10.3389/fimmu.2021.761448] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/13/2021] [Indexed: 01/04/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is an essential therapeutic modality for patients with hematological malignancies and other blood disorders. Unfortunately, acute graft-versus-host disease (aGVHD) remains a major source of morbidity and mortality following allo-HCT, which limits its use in a broader spectrum of patients. Chronic graft-versus-host disease (cGVHD) also remains the most common long-term complication of allo-HCT, occurring in reportedly 30-70% of patients surviving more than 100 days. Chronic GVHD is also the leading cause of non-relapse mortality (NRM) occurring more than 2 years after HCT for malignant disease. Graft versus tumor (GVT) is a major component of the overall beneficial effects of allogeneic HCT in the treatment of hematological malignancies. Better understanding of GVHD pathogenesis is important to identify new therapeutic targets for GVHD prevention and therapy. Emerging data suggest opposing roles for different T cell subsets, e.g., IFN-γ producing CD4+ and CD8+ T cells (Th1 and Tc1), IL-4 producing T cells (Th2 and Tc2), IL-17 producing T cells (Th17 and Tc17), IL-9 producing T cells (Th9 and Tc9), IL-22 producing T cells (Th22), T follicular helper cells (Tfh), regulatory T-cells (Treg) and tissue resident memory T cells (Trm) in GVHD and GVT etiology. In this review, we first summarize the general description of the cytokine signals that promote the differentiation of T cell subsets and the roles of these T cell subsets in the pathogenesis of GVHD. Next, we extensively explore preclinical findings of T cell subsets in both GVHD/GVT animal models and humans. Finally, we address recent findings about the roles of T-cell subsets in clinical GVHD and current strategies to modulate T-cell differentiation for treating and preventing GVHD in patients. Further exploring and outlining the immune biology of T-cell differentiation in GVHD that will provide more therapeutic options for maintaining success of allo-HCT.
Collapse
Affiliation(s)
| | | | | | - Sophie Paczesny
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
231
|
Liang Y, Gong H, Li Y, Lu Y, Wu X, Zhang X, Ding D, Tang X, Tang Q. Aggregation-Induced Emission-Based Vaccine Improves Potential Antitumor Immunotherapy. J Biomed Nanotechnol 2021; 17:2053-2061. [PMID: 34706805 DOI: 10.1166/jbn.2021.3174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Recently, immunomodulation based on biomaterials has held great promise for preventing and treating cancer. Tumor vaccination can be considered as one of promising immunotherapies, compared with the vaccines for infectious disease, it still stays in its infant. Herein, we designed a near-infrared-emitting AIEgens (named TPE-Ph-DCM) based vaccine as an adjuvant in enhancing immune response. AIE-based photodynamic vaccine exhibited efficiently enhancement of the DC?s antigen prestation and elicited antigen-specific cytotoxic T lymphocyte functionality, and significantly inhibited B16-OVA tumor growth prophylactically and therapeutically in mice model. This study is expected to provide a scientific basis for developing effective and safe tumor vaccines.
Collapse
Affiliation(s)
- Yong Liang
- Central and Clinical Laboratory, The Affiliated Huaian Hospital of Xuzhou Medical University and Huaian Second Hospital, Huaian, 223002, China
| | - Huanle Gong
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Yan Li
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yinghao Lu
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Xiaoqian Wu
- Central and Clinical Laboratory, The Affiliated Huaian Hospital of Xuzhou Medical University and Huaian Second Hospital, Huaian, 223002, China
| | - Xiaoyan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Tianjin, 300071, China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Tianjin, 300071, China
| | - Xiaolong Tang
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of the Ministry of Education, Anhui University of Science & Technology, Huainan, 232001, China
| | - Qiyun Tang
- Department of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
232
|
Cui X, Liu R, Duan L, Cao D, Zhang Q, Zhang A. CAR-T therapy: Prospects in targeting cancer stem cells. J Cell Mol Med 2021; 25:9891-9904. [PMID: 34585512 PMCID: PMC8572776 DOI: 10.1111/jcmm.16939] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/20/2021] [Accepted: 09/01/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs), a group of tumour cells with stem cell characteristics, have the ability of self-renewal, multi-lineage differentiation and tumour formation. Since CSCs are resistant to conventional radiotherapy and chemotherapy, their existence may be one of the root causes of cancer treatment failure and tumour progression. The elimination of CSCs may be effective for eventual tumour eradication. Because of the good therapeutic effects without major histocompatibility complex (MHC) restriction and the unique characteristics of CSCs, chimeric antigen receptor T-cell (CAR-T) therapy is expected to be an important method to eliminate CSCs. In this review, we have discussed the feasibility of CSCs-targeted CAR-T therapy for cancer treatment, summarized current research and clinical trials of targeting CSCs with CAR-T cells and forecasted the challenges and future direction from the perspectives of toxicity, persistence and potency, trafficking, infiltration, immunosuppressive tumour microenvironment, and tumour heterogeneity.
Collapse
Affiliation(s)
- Xiaoyue Cui
- Basic Laboratory, Suining Central Hospital, Suining, China
| | - Rui Liu
- Department of Breast and Thyroid Surgery, Suining Central Hospital, Suining, China
| | - Lian Duan
- Basic Laboratory, Suining Central Hospital, Suining, China
| | - Dan Cao
- Basic Laboratory, Suining Central Hospital, Suining, China.,Key Laboratory of Metabolic Diseases, Suining Central Hospital, Suining, China
| | - Qiaoling Zhang
- Basic Laboratory, Suining Central Hospital, Suining, China.,Key Laboratory of Metabolic Diseases, Suining Central Hospital, Suining, China
| | - Aijie Zhang
- Basic Laboratory, Suining Central Hospital, Suining, China
| |
Collapse
|
233
|
Hu M, Rogers NM, Li J, Zhang GY, Wang YM, Shaw K, O'Connell PJ, Alexander SI. Antigen Specific Regulatory T Cells in Kidney Transplantation and Other Tolerance Settings. Front Immunol 2021; 12:717594. [PMID: 34512640 PMCID: PMC8428972 DOI: 10.3389/fimmu.2021.717594] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
Kidney transplantation is the most common solid organ transplant and the best current therapy for end-stage kidney failure. However, with standard immunosuppression, most transplants develop chronic dysfunction or fail, much of which is due to chronic immune injury. Tregs are a subset of T cells involved in limiting immune activation and preventing autoimmune disease. These cells offer the potential to provide tolerance or to allow reduction in immunosuppression in kidney transplants. The importance of Tregs in kidney transplantation has been shown in a number of seminal mouse and animal studies, including those with T cell receptors (TCRs) transgenic Tregs (TCR-Tregs) or Chimeric Antigen Receptor (CAR) Tregs (CAR-Tregs) showing that specificity increases the potency of Treg function. Here we outline the animal and human studies and clinical trials directed at using Tregs in kidney transplantation and other tolerance settings and the various modifications to enhance allo-specific Treg function in vivo and in vitro.
Collapse
Affiliation(s)
- Min Hu
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Geoff Y Zhang
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Karli Shaw
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Philip J O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Stephen I Alexander
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW, Australia
| |
Collapse
|
234
|
Daly S, O’Sullivan A, MacLoughlin R. Cellular Immunotherapy and the Lung. Vaccines (Basel) 2021; 9:1018. [PMID: 34579255 PMCID: PMC8473388 DOI: 10.3390/vaccines9091018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
The new era of cellular immunotherapies has provided state-of-the-art and efficient strategies for the prevention and treatment of cancer and infectious diseases. Cellular immunotherapies are at the forefront of innovative medical care, including adoptive T cell therapies, cancer vaccines, NK cell therapies, and immune checkpoint inhibitors. The focus of this review is on cellular immunotherapies and their application in the lung, as respiratory diseases remain one of the main causes of death worldwide. The ongoing global pandemic has shed a new light on respiratory viruses, with a key area of concern being how to combat and control their infections. The focus of cellular immunotherapies has largely been on treating cancer and has had major successes in the past few years. However, recent preclinical and clinical studies using these immunotherapies for respiratory viral infections demonstrate promising potential. Therefore, in this review we explore the use of multiple cellular immunotherapies in treating viral respiratory infections, along with investigating several routes of administration with an emphasis on inhaled immunotherapies.
Collapse
Affiliation(s)
- Sorcha Daly
- College of Medicine, Nursing & Health Sciences, National University of Ireland, H91 TK33 Galway, Ireland;
| | - Andrew O’Sullivan
- Research and Development, Science and Emerging Technologies, Aerogen Limited, Galway Business Park, H91 HE94 Galway, Ireland;
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen Limited, Galway Business Park, H91 HE94 Galway, Ireland;
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| |
Collapse
|
235
|
Sun Y, Li F, Sonnemann H, Jackson KR, Talukder AH, Katailiha AS, Lizee G. Evolution of CD8 + T Cell Receptor (TCR) Engineered Therapies for the Treatment of Cancer. Cells 2021; 10:cells10092379. [PMID: 34572028 PMCID: PMC8469972 DOI: 10.3390/cells10092379] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/30/2022] Open
Abstract
Engineered T cell receptor T (TCR-T) cell therapy has facilitated the generation of increasingly reliable tumor antigen-specific adaptable cellular products for the treatment of human cancer. TCR-T cell therapies were initially focused on targeting shared tumor-associated peptide targets, including melanoma differentiation and cancer-testis antigens. With recent technological developments, it has become feasible to target neoantigens derived from tumor somatic mutations, which represents a highly personalized therapy, since most neoantigens are patient-specific and are rarely shared between patients. TCR-T therapies have been tested for clinical efficacy in treating solid tumors in many preclinical studies and clinical trials all over the world. However, the efficacy of TCR-T therapy for the treatment of solid tumors has been limited by a number of factors, including low TCR avidity, off-target toxicities, and target antigen loss leading to tumor escape. In this review, we discuss the process of deriving tumor antigen-specific TCRs, including the identification of appropriate tumor antigen targets, expansion of antigen-specific T cells, and TCR cloning and validation, including techniques and tools for TCR-T cell vector construction and expression. We highlight the achievements of recent clinical trials of engineered TCR-T cell therapies and discuss the current challenges and potential solutions for improving their safety and efficacy, insights that may help guide future TCR-T studies in cancer.
Collapse
Affiliation(s)
- Yimo Sun
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Fenge Li
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Heather Sonnemann
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Kyle R. Jackson
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Amjad H. Talukder
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Arjun S. Katailiha
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Gregory Lizee
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
- Department of Immunology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
236
|
Donohoue PD, Pacesa M, Lau E, Vidal B, Irby MJ, Nyer DB, Rotstein T, Banh L, Toh MS, Gibson J, Kohrs B, Baek K, Owen ALG, Slorach EM, van Overbeek M, Fuller CK, May AP, Jinek M, Cameron P. Conformational control of Cas9 by CRISPR hybrid RNA-DNA guides mitigates off-target activity in T cells. Mol Cell 2021; 81:3637-3649.e5. [PMID: 34478654 DOI: 10.1016/j.molcel.2021.07.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 05/28/2021] [Accepted: 07/28/2021] [Indexed: 12/26/2022]
Abstract
The off-target activity of the CRISPR-associated nuclease Cas9 is a potential concern for therapeutic genome editing applications. Although high-fidelity Cas9 variants have been engineered, they exhibit varying efficiencies and have residual off-target effects, limiting their applicability. Here, we show that CRISPR hybrid RNA-DNA (chRDNA) guides provide an effective approach to increase Cas9 specificity while preserving on-target editing activity. Across multiple genomic targets in primary human T cells, we show that 2'-deoxynucleotide (dnt) positioning affects guide activity and specificity in a target-dependent manner and that this can be used to engineer chRDNA guides with substantially reduced off-target effects. Crystal structures of DNA-bound Cas9-chRDNA complexes reveal distorted guide-target duplex geometry and allosteric modulation of Cas9 conformation. These structural effects increase specificity by perturbing DNA hybridization and modulating Cas9 activation kinetics to disfavor binding and cleavage of off-target substrates. Overall, these results pave the way for utilizing customized chRDNAs in clinical applications.
Collapse
Affiliation(s)
- Paul D Donohoue
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA.
| | - Martin Pacesa
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Elaine Lau
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Bastien Vidal
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Matthew J Irby
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - David B Nyer
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Tomer Rotstein
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Lynda Banh
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Mckenzi S Toh
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Jason Gibson
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Bryan Kohrs
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Kevin Baek
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Arthur L G Owen
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Euan M Slorach
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Megan van Overbeek
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Christopher K Fuller
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA
| | - Andrew P May
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA.
| | - Martin Jinek
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| | - Peter Cameron
- Caribou Biosciences, Inc., 2929 Seventh Street, Suite 105, Berkeley, CA 94710, USA.
| |
Collapse
|
237
|
Chimeric antigen receptor- and natural killer cell receptor-engineered innate killer cells in cancer immunotherapy. Cell Mol Immunol 2021; 18:2083-2100. [PMID: 34267335 PMCID: PMC8429625 DOI: 10.1038/s41423-021-00732-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T-cell (CAR-T) therapy has demonstrated impressive therapeutic efficacy against hematological malignancies, but multiple challenges have hindered its application, particularly for the eradication of solid tumors. Innate killer cells (IKCs), particularly NK cells, NKT cells, and γδ T cells, employ specific antigen-independent innate tumor recognition and cytotoxic mechanisms that simultaneously display high antitumor efficacy and prevent tumor escape caused by antigen loss or modulation. IKCs are associated with a low risk of developing GVHD, thus offering new opportunities for allogeneic "off-the-shelf" cellular therapeutic products. The unique innate features, wide tumor recognition range, and potent antitumor functions of IKCs make them potentially excellent candidates for cancer immunotherapy, particularly serving as platforms for CAR development. In this review, we first provide a brief summary of the challenges hampering CAR-T-cell therapy applications and then discuss the latest CAR-NK-cell research, covering the advantages, applications, and clinical translation of CAR- and NK-cell receptor (NKR)-engineered IKCs. Advances in synthetic biology and the development of novel genetic engineering techniques, such as gene-editing and cellular reprogramming, will enable the further optimization of IKC-based anticancer therapies.
Collapse
|
238
|
Wang H, Wang L, Li Y, Li G, Zhang X, Jiang D, Zhang Y, Liu L, Chu Y, Xu G. Nanobody-armed T cells endow CAR-T cells with cytotoxicity against lymphoma cells. Cancer Cell Int 2021; 21:450. [PMID: 34429118 PMCID: PMC8386010 DOI: 10.1186/s12935-021-02151-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/13/2021] [Indexed: 02/02/2023] Open
Abstract
Background Taking advantage of nanobodies (Nbs) in immunotherapy, we investigated the cytotoxicity of Nb-based chimeric antigen receptor T cells (Nb CAR-T) against lymphoma cells. Methods CD19 Nb CAR-T, CD20 Nb CAR-T, and Bispecific Nb CAR-T cells were generated by panning anti-human CD19- and CD20-specific nanobody sequences from a natural Nb-expressing phage display library, integrating Nb genes with a lentiviral cassette that included other CAR elements, and finally transducing T cells that were expanded under an optimization system with the above generated CAR lentivirus. Prepared Nb CAR-T cells were cocultured with tumour cell lines or primary tumour cells for 24 h or 5 days to evaluate their biological function. Results The nanobodies that we selected from the natural Nb-expressing phage display library had a high affinity and specificity for CD19 and CD20. CD19 Nb CAR-T, CD20 Nb CAR-T and Bispecific Nb CAR-T cells were successfully constructed, and these Nb CAR-T cells could strongly recognize Burkitt lymphoma cell lines (Raji and Daudi), thereby leading to activation, enhanced proliferation, and specific killing of target cells. Furthermore, similar results were obtained when using patient samples as target cells, with a cytotoxicity of approximately 60%. Conclusions Nanobody-based CAR-T cells can kill both tumour cell lines and patient-derived tumour cells in vitro, and Nb-based CAR-T cells may be a promising therapeutic strategy in future immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02151-z.
Collapse
Affiliation(s)
- Hongxia Wang
- General Hospital of Ningxia Medical University, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, China
| | - Liyan Wang
- General Hospital of Ningxia Medical University, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yanning Li
- General Hospital of Ningxia Medical University, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Guangqi Li
- General Hospital of Ningxia Medical University, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Xiaochun Zhang
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - Dan Jiang
- General Hospital of Ningxia Medical University, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yanting Zhang
- General Hospital of Ningxia Medical University, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Liyuan Liu
- General Hospital of Ningxia Medical University, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yuankui Chu
- General Hospital of Ningxia Medical University, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Guangxian Xu
- General Hospital of Ningxia Medical University, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China. .,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
239
|
Feasibility of Monitoring Tumor Response by Tracking Nanoparticle-Labelled T Cells Using X-ray Fluorescence Imaging-A Numerical Study. Int J Mol Sci 2021; 22:ijms22168736. [PMID: 34445443 PMCID: PMC8395984 DOI: 10.3390/ijms22168736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/28/2021] [Accepted: 08/10/2021] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy has been a breakthrough in cancer treatment, yet only a subgroup of patients responds to these novel drugs. Parameters such as cytotoxic T-cell infiltration into the tumor have been proposed for the early evaluation and prediction of therapeutic response, demanded for non-invasive, sensitive and longitudinal imaging. We have evaluated the feasibility of X-ray fluorescence imaging (XFI) to track immune cells and thus monitor the immune response. For that, we have performed Monte Carlo simulations using a mouse voxel model. Spherical targets, enriched with gold or palladium fluorescence agents, were positioned within the model and imaged using a monochromatic photon beam of 53 or 85 keV. Based on our simulation results, XFI may detect as few as 730 to 2400 T cells labelled with 195 pg gold each when imaging subcutaneous tumors in mice, with a spatial resolution of 1 mm. However, the detection threshold is influenced by the depth of the tumor as surrounding tissue increases scattering and absorption, especially when utilizing palladium imaging agents with low-energy characteristic fluorescence photons. Further evaluation and conduction of in vivo animal experiments will be required to validate and advance these promising results.
Collapse
|
240
|
Miller IC, Zamat A, Sun LK, Phuengkham H, Harris AM, Gamboa L, Yang J, Murad JP, Priceman SJ, Kwong GA. Enhanced intratumoural activity of CAR T cells engineered to produce immunomodulators under photothermal control. Nat Biomed Eng 2021; 5:1348-1359. [PMID: 34385695 DOI: 10.1038/s41551-021-00781-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/11/2021] [Indexed: 12/17/2022]
Abstract
Treating solid malignancies with chimeric antigen receptor (CAR) T cells typically results in poor responses. Immunomodulatory biologics delivered systemically can augment the cells' activity, but off-target toxicity narrows the therapeutic window. Here we show that the activity of intratumoural CAR T cells can be controlled photothermally via synthetic gene switches that trigger the expression of transgenes in response to mild temperature elevations (to 40-42 °C). In vitro, heating engineered primary human T cells for 15-30 min led to over 60-fold-higher expression of a reporter transgene without affecting the cells' proliferation, migration and cytotoxicity. In mice, CAR T cells photothermally heated via gold nanorods produced a transgene only within the tumours. In mouse models of adoptive transfer, the systemic delivery of CAR T cells followed by intratumoural production, under photothermal control, of an interleukin-15 superagonist or a bispecific T cell engager bearing an NKG2D receptor redirecting T cells against NKG2D ligands enhanced antitumour activity and mitigated antigen escape. Localized photothermal control of the activity of engineered T cells may enhance their safety and efficacy.
Collapse
Affiliation(s)
- Ian C Miller
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Ali Zamat
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Lee-Kai Sun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Hathaichanok Phuengkham
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Adrian M Harris
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Lena Gamboa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Jason Yang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - John P Murad
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - Saul J Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA.,Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Gabriel A Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA. .,Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA. .,Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA. .,Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA, USA. .,Georgia Immunoengineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA, USA. .,Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
241
|
Wu K, Wang L, Liu M, Xiu Y, Hu Y, Fu S, Huang H, Xu B, Xiao H. The CD226-ERK1/2-LAMP1 pathway is an important mechanism for Vγ9Vδ2 T cell cytotoxicity against chemotherapy-resistant acute myeloid leukemia blasts and leukemia stem cells. Cancer Sci 2021; 112:3233-3242. [PMID: 34107135 PMCID: PMC8353902 DOI: 10.1111/cas.15014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/18/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
Vγ9Vδ2 T cells are attractive effector cells for immunotherapy with potent cytotoxic activity against a variety of malignant cells. However, the effect of Vγ9Vδ2 T cells on chemotherapy-resistant acute myeloid leukemia (AML) blasts, especially highly refractory leukemia stem cells (LSCs) is still unknown. In this study, we investigated the effect of cytotoxicity of allogeneic Vγ9Vδ2 T cells on chemotherapy-resistant AML cell lines, as well as on primary AML blasts and LSCs obtained from refractory AML patients. The results indicated that Vγ9Vδ2 T cells can efficiently kill drug-resistant AML cell lines in vitro and in vivo, and the sensitivity of AML cells to Vγ9Vδ2 T cell-mediated cytotoxicity is not influenced by the sensitivity of AML cells to chemotherapy. We further found that Vγ9Vδ2 T cells exhibited a comparable effect of cytotoxicity against LSCs to primary AML blasts. More importantly, we revealed that the CD226-extracellular signal-regulatory kinase1/2 (ERK1/2)-lysosome-associated membrane protein 1 (LAMP1) pathway is an important mechanism for Vγ9Vδ2 T cell-induced cytotoxicity against AML cells. First, Vγ9Vδ2 T cells recognized AML cells by receptor-ligand interaction of CD226-Nectin-2, which then induced ERK1/2 phosphorylation in Vγ9Vδ2 T cells. Finally, triggering the movement of lytic granules toward AML cells induced cytolysis of AML cells. The expression level of Nectin-2 may be used as a novel marker to predict the susceptibility/resistance of AML cells to Vγ9Vδ2 T cell treatment.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Cytotoxicity, Immunologic
- Drug Resistance, Neoplasm
- Female
- HL-60 Cells
- Humans
- Immunotherapy, Adoptive
- K562 Cells
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Lysosomal Membrane Proteins/metabolism
- MAP Kinase Signaling System
- Mice
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
- Treatment Outcome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kangni Wu
- Bone Marrow Transplantation CenterThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Department of HematologyThe First Affiliated Hospital of Xiamen University and Institute of HematologyMedical College of Xiamen UniversityXiamenChina
| | - Li‐mengmeng Wang
- Bone Marrow Transplantation CenterThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Meng Liu
- Bone Marrow Transplantation CenterThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yanghui Xiu
- Eye Institute and Xiamen Eye Center Affiliated to Xiamen UniversityXiamenChina
| | - Yongxian Hu
- Bone Marrow Transplantation CenterThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Shan Fu
- Bone Marrow Transplantation CenterThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - He Huang
- Bone Marrow Transplantation CenterThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Bing Xu
- Department of HematologyThe First Affiliated Hospital of Xiamen University and Institute of HematologyMedical College of Xiamen UniversityXiamenChina
| | - Haowen Xiao
- Department of HematologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
242
|
Dolan JG, Paessler ME, Rheingold SR, Pillai V. Hematopathologic Correlates of CAR T-Cell Therapy. Clin Lab Med 2021; 41:325-339. [PMID: 34304768 DOI: 10.1016/j.cll.2021.03.012] [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] [Indexed: 11/25/2022]
Abstract
CD19-targeting chimeric antigen rector (CAR) T-cell products are used for the treatment of relapsed/refractory B-acute lymphoblastic leukemia, diffuse large B-cell lymphoma, and mantle cell lymphoma. The success of CD19-CAR-T cells has led to the investigation of CAR T-cell products targeting different antigens in other hematological malignancies and solid tumors. Clinical laboratories play an important role in the manufacture, distribution, and monitoring of CAR T-cell therapy. Hence, it is important for laboratory professionals to be cognizant of clinicopathologic aspects of CAR T-cell therapy.
Collapse
Affiliation(s)
- J Gregory Dolan
- Division of Oncology and Cellular Therapy, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michele E Paessler
- Division of Hematopathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan R Rheingold
- Division of Oncology and Cellular Therapy, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vinodh Pillai
- Division of Hematopathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
243
|
Jan M, Sperling AS, Ebert BL. Cancer therapies based on targeted protein degradation - lessons learned with lenalidomide. Nat Rev Clin Oncol 2021; 18:401-417. [PMID: 33654306 PMCID: PMC8903027 DOI: 10.1038/s41571-021-00479-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 02/08/2023]
Abstract
For decades, anticancer targeted therapies have been designed to inhibit kinases or other enzyme classes and have profoundly benefited many patients. However, novel approaches are required to target transcription factors, scaffolding proteins and other proteins central to cancer biology that typically lack catalytic activity and have remained mostly recalcitrant to drug development. The selective degradation of target proteins is an attractive approach to expand the druggable proteome, and the selective oestrogen receptor degrader fulvestrant served as an early example of this concept. Following a long and tragic history in the clinic, the immunomodulatory imide drug (IMiD) thalidomide was discovered to exert its therapeutic activity via a novel and unexpected mechanism of action: targeting proteins to an E3 ubiquitin ligase for subsequent proteasomal degradation. This discovery has paralleled and directly catalysed myriad breakthroughs in drug development, leading to the rapid maturation of generalizable chemical platforms for the targeted degradation of previously undruggable proteins. Decades of clinical experience have established front-line roles for thalidomide analogues, including lenalidomide and pomalidomide, in the treatment of haematological malignancies. With a new generation of 'degrader' drugs currently in development, this experience provides crucial insights into class-wide features of degraders, including a unique pharmacology, mechanisms of resistance and emerging therapeutic opportunities. Herein, we review these past experiences and discuss their application in the clinical development of novel degrader therapies.
Collapse
Affiliation(s)
- Max Jan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Adam S Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
244
|
Wehrli M, Maus MV. New CARs on and off the road: challenges and new developments in CAR-T cell therapy. Curr Opin Pharmacol 2021; 59:116-126. [PMID: 34198161 DOI: 10.1016/j.coph.2021.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 11/19/2022]
Abstract
Building on the tremendous success of chimeric antigen receptor T-cell therapy in hematological malignancies, there are efforts under way to overcome the challenges associated with this treatment and expand its application to solid tumors. In this fast-evolving field, new therapeutic options are constantly generated, tested in model systems, and further evaluated in clinical trials. In this review, we provide an overview of recent challenges and developments associated with engineered T cells and chimeric antigen receptor T-cell applications. We report on the most recent progress in hematological malignancies and highlight technical advances for applications in solid tumors.
Collapse
Affiliation(s)
- Marc Wehrli
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
245
|
Wang N, Li X, Wang R, Ding Z. Spatial transcriptomics and proteomics technologies for deconvoluting the tumor microenvironment. Biotechnol J 2021; 16:e2100041. [PMID: 34125481 DOI: 10.1002/biot.202100041] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/31/2022]
Abstract
The tumor microenvironment (TME) harbors heterogeneous contents and plays critical roles in tumorigenesis, metastasis, and drug resistance. Therefore, the deconvolution of the TME becomes increasingly essential to every aspect of cancer research and treatment. Novel spatially-resolved high-plex molecular profiling technologies have been emerging rapidly as powerful tools to obtain in-depth understanding from TME perspectives due to their capacity to allow high-plex protein and RNA profiling while keeping valuable spatial information. Based on our practical experience, we review a variety of available spatial proteogenomic technologies, including 10X Visium, GeoMx Digital Spatial Profiler (DSP), cyclic immunofluorescence-based CODEX and Multi-Omyx, mass spectrometry (MS)-based imaging mass spectrometry (IMS) and multiplex ion-beam imaging (MIBI). We also discuss FISSEQ, MERFISH, Slide-seq, and HDST, some of which may become commercially available in the near future. In particular, with our experience, we elaborate on DSP for spatial proteogenomic profiling and discuss its unique features designed for immuno-oncology and propose anticipation towards its future direction. The emerging spatially technologies are rapidly reshaping the magnitude of our understanding of the TME.
Collapse
Affiliation(s)
- Nan Wang
- Fynn Biotechnologies Ltd., Mills Institute for Personalized Cancer Care, Jinan City, Shandong Province, P. R. China
| | - Xia Li
- Fynn Biotechnologies Ltd., Mills Institute for Personalized Cancer Care, Jinan City, Shandong Province, P. R. China
| | - Rongshui Wang
- Fynn Biotechnologies Ltd., Mills Institute for Personalized Cancer Care, Jinan City, Shandong Province, P. R. China
| | - Zhiyong Ding
- Fynn Biotechnologies Ltd., Mills Institute for Personalized Cancer Care, Jinan City, Shandong Province, P. R. China
| |
Collapse
|
246
|
Targeted delivery of CRISPR-Cas9 and transgenes enables complex immune cell engineering. Cell Rep 2021; 35:109207. [PMID: 34077734 PMCID: PMC8236216 DOI: 10.1016/j.celrep.2021.109207] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/09/2021] [Accepted: 05/11/2021] [Indexed: 01/04/2023] Open
Abstract
As genome engineering advances cell-based therapies, a versatile approach to introducing both CRISPR-Cas9 ribonucleoproteins (RNPs) and therapeutic transgenes into specific cells would be transformative. Autologous T cells expressing a chimeric antigen receptor (CAR) manufactured by viral transduction are approved to treat multiple blood cancers, but additional genetic modifications to alter cell programs will likely be required to treat solid tumors and for allogeneic cellular therapies. We have developed a one-step strategy using engineered lentiviral particles to introduce Cas9 RNPs and a CAR transgene into primary human T cells without electroporation. Furthermore, programming particle tropism allows us to target a specific cell type within a mixed cell population. As a proof-of-concept, we show that HIV-1 envelope targeted particles to edit CD4+ cells while sparing co-cultured CD8+ cells. This adaptable approach to immune cell engineering ex vivo provides a strategy applicable to the genetic modification of targeted somatic cells in vivo.
Collapse
|
247
|
Absolute lymphocyte count proliferation kinetics after CAR T-cell infusion impact response and relapse. Blood Adv 2021; 5:2128-2136. [PMID: 33881465 DOI: 10.1182/bloodadvances.2020004038] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/24/2021] [Indexed: 12/18/2022] Open
Abstract
CD19-directed chimeric antigen receptor (CAR) T cells show characteristic proliferation kinetics after infusion that correlate with response. Clearance of circulating disease, B-cell aplasia (BCA), and cytokine release syndrome (CRS) are used to observe CAR T-cell function, given the lack of commercial CAR T-cell measurement assays. We investigated the utility of common hematology laboratory parameters in 166 patients with B-cell acute lymphoblastic leukemia (B-ALL) who were treated with CAR T-cell therapy targeting CD19. CAR T-cell infusion was followed by disappearance of circulating blasts in 86% of patients at a median of 6 days. After a lag phase, there was a rapid expansion in absolute lymphocyte count (ALC) in the second week that coincided with the appearance of atypical lymphocytes. The expansion phase was followed by a contraction phase with a concomitant decrease in atypical lymphocytes. In vitro CAR T-cell studies showed similar kinetics and morphological changes. Peak ALC and overall expansion was greater in sustained responders compared with that in nonresponders. Patients with early loss of BCA and those with eventual CD19+ minimal residual disease/relapse showed lower overall lymphocyte expansion compared with the controls. Pleomorphic lymphocytosis was noted in the cerebrospinal fluid at post-CAR time points. We conclude that lymphocyte counts and differential can also be used to evaluate CAR T-cell expansion after infusion, along with BCA and CRS. This is the first report to characterize the morphology of CAR T cells and determine the utility of lymphocyte kinetics.
Collapse
|
248
|
Gong Y, Klein Wolterink RGJ, Wang J, Bos GMJ, Germeraad WTV. Chimeric antigen receptor natural killer (CAR-NK) cell design and engineering for cancer therapy. J Hematol Oncol 2021; 14:73. [PMID: 33933160 PMCID: PMC8088725 DOI: 10.1186/s13045-021-01083-5] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
Due to their efficient recognition and lysis of malignant cells, natural killer (NK) cells are considered as specialized immune cells that can be genetically modified to obtain capable effector cells for adoptive cellular treatment of cancer patients. However, biological and technical hurdles related to gene delivery into NK cells have dramatically restrained progress. Recent technological advancements, including improved cell expansion techniques, chimeric antigen receptors (CAR), CRISPR/Cas9 gene editing and enhanced viral transduction and electroporation, have endowed comprehensive generation and characterization of genetically modified NK cells. These promising developments assist scientists and physicians to design better applications of NK cells in clinical therapy. Notably, redirecting NK cells using CARs holds important promise for cancer immunotherapy. Various preclinical and a limited number of clinical studies using CAR-NK cells show promising results: efficient elimination of target cells without side effects, such as cytokine release syndrome and neurotoxicity which are seen in CAR-T therapies. In this review, we focus on the details of CAR-NK technology, including the design of efficient and safe CAR constructs and associated NK cell engineering techniques: the vehicles to deliver the CAR-containing transgene, detection methods for CARs, as well as NK cell sources and NK cell expansion. We summarize the current CAR-NK cell literature and include valuable lessons learned from the CAR-T cell field. This review also provides an outlook on how these approaches may transform current clinical products and protocols for cancer treatment.
Collapse
Affiliation(s)
- Ying Gong
- Division of Hematology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.,GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Roel G J Klein Wolterink
- Division of Hematology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.,GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China.,National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Gerard M J Bos
- Division of Hematology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.,GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,CiMaas BV, Maastricht, The Netherlands
| | - Wilfred T V Germeraad
- Division of Hematology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands. .,GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands. .,CiMaas BV, Maastricht, The Netherlands.
| |
Collapse
|
249
|
Pearlman AH, Hwang MS, Konig MF, Hsiue EHC, Douglass J, DiNapoli SR, Mog BJ, Bettegowda C, Pardoll DM, Gabelli SB, Papadopoulos N, Kinzler KW, Vogelstein B, Zhou S. Targeting public neoantigens for cancer immunotherapy. NATURE CANCER 2021; 2:487-497. [PMID: 34676374 PMCID: PMC8525885 DOI: 10.1038/s43018-021-00210-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 04/13/2021] [Indexed: 02/06/2023]
Abstract
Several current immunotherapy approaches target private neoantigens derived from mutations that are unique to individual patients' tumors. However, immunotherapeutic agents can also be developed against public neoantigens derived from recurrent mutations in cancer driver genes. The latter approaches target proteins that are indispensable for tumor growth, and each therapeutic agent can be applied to numerous patients. Here we review the opportunities and challenges involved in the identification of suitable public neoantigen targets and the development of therapeutic agents targeting them.
Collapse
Affiliation(s)
- Alexander H Pearlman
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Michael S Hwang
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Genentech, Inc., South San Francisco, CA, USA
| | - Maximilian F Konig
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emily Han-Chung Hsiue
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Jacqueline Douglass
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Sarah R DiNapoli
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Brian J Mog
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Chetan Bettegowda
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Drew M Pardoll
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Sandra B Gabelli
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas Papadopoulos
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth W Kinzler
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
- Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bert Vogelstein
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
250
|
Pimenta C, Bettiol V, Alencar-Silva T, Franco OL, Pogue R, Carvalho JL, Felipe MSS. Advanced Therapies and Regulatory Framework in Different Areas of the Globe: Past, Present, and Future. Clin Ther 2021; 43:e103-e138. [PMID: 33892966 DOI: 10.1016/j.clinthera.2021.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/02/2021] [Accepted: 02/23/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE The field of human medicine is in a constant state of evolution, developing and incorporating technological advances from diverse scientific fields. In recent years, cellular and gene therapies have come of age, challenging regulatory agencies to define the path for commercial registration. Approval necessarily demands robust evidence for safety and efficacy, but these exigencies must not be such that they render unviable the development and testing of the therapeutic agent. Furthermore, reimbursement strategies are required to guarantee commercial viability of these products, to avoid the risk that they will be removed from the market or become unavailable to most patients through lack of financial resources. To address such challenges, several countries have created strategies to manage advanced therapy products. METHODS Based on official documents published by regulatory agencies worldwide, this review summarizes the current scenario in the United States, Europe, Brazil, Japan, South Korea, and China in this regard, discussing the harmonized and dissonant aspects of the regulatory framework in different regions of the world and exploring perspectives for the future. FINDINGS The technical aspects of advanced therapies are increasingly complex, bringing challenges for high mass commercialization and demanding specific regulation. The regulatory framework of the analyzed regions is mainly recent and discordant, but many harmonizing initiatives were observed. IMPLICATIONS The comparative analysis of regulatory frameworks in different parts of the world is informative, as scientists must be aware of the rationale of regulators to assertively develop new technology and products that will be commercialized. The comparative analysis also provides insight into the main dissonances that must be addressed, fostering the harmonization of local regulatory frameworks. Many unanswered questions still lie ahead for the field of advanced therapies, and empirical evidence will be the most effective way to separate hype from hope and to establish the most sustainable mechanisms to regulate and finance such products in each part of the world.
Collapse
Affiliation(s)
- Cleila Pimenta
- Public Health Program, University of Brasília, DF, Brazil
| | - Vitória Bettiol
- Genomic Sciences and Biotechnology Program, Catholic University of Brasília, DF, Brazil
| | - Thuany Alencar-Silva
- Genomic Sciences and Biotechnology Program, Catholic University of Brasília, DF, Brazil
| | - Octavio Luiz Franco
- Genomic Sciences and Biotechnology Program, Catholic University of Brasília, DF, Brazil; Catholic University of Dom Bosco, Campo Grande, MS, Brazil
| | - Robert Pogue
- Genomic Sciences and Biotechnology Program, Catholic University of Brasília, DF, Brazil
| | - Juliana Lott Carvalho
- Genomic Sciences and Biotechnology Program, Catholic University of Brasília, DF, Brazil; Faculty of Medicine, University of Brasília, DF, Brazil
| | - Maria Sueli Soares Felipe
- Public Health Program, University of Brasília, DF, Brazil; Genomic Sciences and Biotechnology Program, Catholic University of Brasília, DF, Brazil.
| |
Collapse
|