ijms-logo

Journal Browser

Journal Browser

CAR-T Cell Therapy

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pathology, Diagnostics, and Therapeutics".

Deadline for manuscript submissions: closed (30 November 2019) | Viewed by 120310

Special Issue Editor

Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
Interests: cancer metastasis; cancer drug resistance; epigenetics; non-coding RNAs; miRNAs; breast cancer; prostate cancer; lung cancer; meningioma; pancreatic cancer; cancer health disparity
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

CAR (Chimeric Antigen Receptor) T-cell therapy is revolutionizing cancer treatment. Several monoclonal antibody-based immunotherapies are available to treat various cancers, but CAR T-cell therapy belongs to the class of ‘adoptive cell transfer’ therapy that makes use of a patient’s own T-cells to fight back against cancer. It involves the harvesting of T-cells and their genetic modification to express an antigen receptor that is normally not present. This creates a chimeric molecule—a T-cell with the combined specificity of an antibody. The receptor chosen to create a CAR T-cell depends on the cancer in question and the identification of a surface protein that is unique to the cancer cells, so that CAR T-cells, once introduced back into the patient, can specifically target and kill cancer cells. This therapy is particularly effective against liquid malignancies with the recent FDA approval of drugs against leukemia and non-Hodgkin lymphoma. The solid tumors, with their protective microenvironment, are a challenge to be targeted by CAR T-cells. Additionally, certain toxicities, primarily neurological complications and cytokine release syndrome, have been associated with this therapy. Finally, attempts are being made to find the utility of CAR T-cell therapy in diseases other than cancer. This Special Issue takes a look at the CAR T-cell therapy—its evolution, progress, and promises, as well as its associated challenges.

Dr. Aamir Ahmad
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Chimeric Antigen Receptor
  • T lymphocyte
  • CAR T-cell therapy
  • adoptive cell transfer therapy
  • immunotherapy
  • Kymriah
  • Yescarta
  • Cytokine Release Syndrome

Related Special Issue

Published Papers (16 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review, Other

3 pages, 146 KiB  
Editorial
CAR-T Cell Therapy
by Aamir Ahmad
Int. J. Mol. Sci. 2020, 21(12), 4303; https://doi.org/10.3390/ijms21124303 - 17 Jun 2020
Cited by 14 | Viewed by 6900
Abstract
CAR-T therapy has revolutionized the treatment of select hematological malignancies, namely, acute lymphoblastic leukemia and large B-cell lymphomas [...] Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)

Research

Jump to: Editorial, Review, Other

18 pages, 5068 KiB  
Article
Rapid and Effective Generation of Nanobody Based CARs using PCR and Gibson Assembly
by Stijn De Munter, Alexander Van Parys, Layla Bral, Joline Ingels, Glenn Goetgeluk, Sarah Bonte, Melissa Pille, Lore Billiet, Karin Weening, Annick Verhee, Jose Van der Heyden, Tom Taghon, Georges Leclercq, Tessa Kerre, Jan Tavernier and Bart Vandekerckhove
Int. J. Mol. Sci. 2020, 21(3), 883; https://doi.org/10.3390/ijms21030883 - 30 Jan 2020
Cited by 20 | Viewed by 6061
Abstract
Recent approval of chimeric antigen receptor (CAR) T cell therapy by the European Medicines Agency (EMA)/Federal and Drug Administration (FDA) and the remarkable results of CAR T clinical trials illustrate the curative potential of this therapy. While CARs against a multitude of different [...] Read more.
Recent approval of chimeric antigen receptor (CAR) T cell therapy by the European Medicines Agency (EMA)/Federal and Drug Administration (FDA) and the remarkable results of CAR T clinical trials illustrate the curative potential of this therapy. While CARs against a multitude of different antigens are being developed and tested (pre)clinically, there is still a need for optimization. The use of single-chain variable fragments (scFvs) as targeting moieties hampers the quick generation of functional CARs and could potentially limit the efficacy. Instead, nanobodies may largely circumvent these difficulties. We used an available nanobody library generated after immunization of llamas against Cluster of Differentiation (CD) 20 through DNA vaccination or against the ectodomain of CD33 using soluble protein. The nanobody specific sequences were amplified by PCR and cloned by Gibson Assembly into a retroviral vector containing two different second-generation CAR constructs. After transduction in T cells, we observed high cell membrane nanoCAR expression in all cases. Following stimulation of nanoCAR-expressing T cells with antigen-positive cell lines, robust T cell activation, cytokine production and tumor cell lysis both in vitro and in vivo was observed. The use of nanobody technology in combination with PCR and Gibson Assembly allows for the rapid and effective generation of compact CARs. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Graphical abstract

14 pages, 2018 KiB  
Article
Preclinical Activity of Embryonic Annexin A2-Specific Chimeric Antigen Receptor T Cells Against Ovarian Cancer
by Leonard Leong, Heng Liang Tan, Simeon Cua, Kylie Su Mei Yong, Qingfeng Chen and Andre Choo
Int. J. Mol. Sci. 2020, 21(2), 381; https://doi.org/10.3390/ijms21020381 - 07 Jan 2020
Cited by 11 | Viewed by 3643
Abstract
Chimeric antigen receptors (CARs) have found clinical success in B cell malignancies, but a dearth of potential targets limits their wider clinical application, especially in solid tumours. Here, we describe the development of an anti-annexin A2 CAR, CAR(2448), derived from an antibody found [...] Read more.
Chimeric antigen receptors (CARs) have found clinical success in B cell malignancies, but a dearth of potential targets limits their wider clinical application, especially in solid tumours. Here, we describe the development of an anti-annexin A2 CAR, CAR(2448), derived from an antibody found to have activity against epithelial ovarian cancer cell lines. The spacer length of CAR(2448) was optimised based on in vitro cytotoxic activity against ovarian cancer (OC) cell lines via a real-time cytotoxicity assay. The longer spacer CAR(2448)L T cells exhibit significant effector activity, inducing inflammatory cytokine release and cytotoxicity against OC cell lines. Furthermore, CAR(2448)L-BBz T cells induced enhanced survival in an in vivo OC xenograft model and reduced tumour volume by 76.6%. Our preclinical studies of CAR(2448) suggest its potential for the unmet need of novel strategies for the treatment of ovarian cancer. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Graphical abstract

16 pages, 1231 KiB  
Article
CSPG4-Specific CAR T Cells for High-Risk Childhood B Cell Precursor Leukemia
by Dennis C. Harrer, Gerold Schuler, Jan Dörrie and Niels Schaft
Int. J. Mol. Sci. 2019, 20(11), 2764; https://doi.org/10.3390/ijms20112764 - 05 Jun 2019
Cited by 18 | Viewed by 4212
Abstract
The advent of CD19-specific chimeric antigen receptor (CAR) T cells has proven to be a powerful asset in the arsenal of cancer immunotherapy of acute lymphoblastic leukemia and certain B cell lymphomas. However, a sizable portion of patients treated with CD19-CAR T cells [...] Read more.
The advent of CD19-specific chimeric antigen receptor (CAR) T cells has proven to be a powerful asset in the arsenal of cancer immunotherapy of acute lymphoblastic leukemia and certain B cell lymphomas. However, a sizable portion of patients treated with CD19-CAR T cells relapse with CD19-negative cancer cells, necessitating the quest for back-up antigens. Chondroitin sulfate proteoglycan 4 (CSPG4) expression has been reported on leukemic blasts bearing the ill-fated MLL 11q23 rearrangement. We aimed at exploring the use of CSPG4-specific CAR T cells against mixed-lineage leukemia (MLL)-rearranged leukemic blasts, using the precursor B cell leukemia cell line KOPN8 (MLL–MLLT1 translocation) as a model. First, we confirmed CSPG4 expression on KOPN8 cells. Bulk T cells electroporated with mRNA encoding a CSPG4-specific CAR upregulated activation markers and secreted the Th1 cytokines TNF and IFNγ in an antigen-specific manner upon co-culture with KOPN8 cells. More importantly, CSPG4-specific CAR T cells evinced specific degranulation towards KOPN8 cells and specifically lysed KOPN8 target cells in chromium lysis experiments. CSPG4 is a well-established CAR target in cutaneous melanoma. Here, we provide proof-of-principle data for the use of CSPG4-specific CAR T cells against MLL-translocated leukemias. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

15 pages, 2553 KiB  
Article
Tumor-Specific Reactive Oxygen Species Accelerators Improve Chimeric Antigen Receptor T Cell Therapy in B Cell Malignancies
by Hyeon Joo Yoo, Yibin Liu, Lei Wang, Maria-Luisa Schubert, Jean-Marc Hoffmann, Sanmei Wang, Brigitte Neuber, Angela Hückelhoven-Krauss, Ulrike Gern, Anita Schmitt, Carsten Müller-Tidow, Peter Dreger, Andriy Mokhir, Michael Schmitt and Leopold Sellner
Int. J. Mol. Sci. 2019, 20(10), 2469; https://doi.org/10.3390/ijms20102469 - 18 May 2019
Cited by 16 | Viewed by 5110
Abstract
Chimeric antigen receptor T cell (CART) therapy is currently one of the most promising treatment approaches in cancer immunotherapy. However, the immunosuppressive nature of the tumor microenvironment, in particular increased reactive oxygen species (ROS) levels, provides considerable limitations. In this study, we aimed [...] Read more.
Chimeric antigen receptor T cell (CART) therapy is currently one of the most promising treatment approaches in cancer immunotherapy. However, the immunosuppressive nature of the tumor microenvironment, in particular increased reactive oxygen species (ROS) levels, provides considerable limitations. In this study, we aimed to exploit increased ROS levels in the tumor microenvironment with prodrugs of ROS accelerators, which are specifically activated in cancer cells. Upon activation, ROS accelerators induce further generation of ROS. This leads to an accumulation of ROS in tumor cells. We hypothesized that the latter cells will be more susceptible to CARTs. CD19-specific CARTs were generated with a CD19.CAR.CD28.CD137zeta third-generation retroviral vector. Cytotoxicity was determined by chromium-51 release assay. Influence of the ROS accelerators on viability and phenotype of CARTs was determined by flow cytometry. The combination of CARTs with the ROS accelerator PipFcB significantly increased their cytotoxicity in the Burkitt lymphoma cell lines Raji and Daudi, as well as primary chronic lymphocytic leukemia cells. Exposure of CARTs to PipFcB for 48 h did not influence T cell exhaustion, viability, or T cell subpopulations. In summary, the combination of CARTs with ROS accelerators may improve adoptive immunotherapy and help to overcome tumor microenvironment-mediated treatment resistance. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

9 pages, 782 KiB  
Review
CAR-T Cell Therapies: An Overview of Clinical Studies Supporting Their Approved Use against Acute Lymphoblastic Leukemia and Large B-Cell Lymphomas
by Aamir Ahmad, Shahab Uddin and Martin Steinhoff
Int. J. Mol. Sci. 2020, 21(11), 3906; https://doi.org/10.3390/ijms21113906 - 30 May 2020
Cited by 43 | Viewed by 6191
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy is an exciting development in the field of cancer immunology, wherein immune T-cells from patients are collected, engineered to create ‘CAR’-T cells, and infused back into the same patient. Currently, two CAR-T-cell-based therapies, Tisagenlecleucel and Axicabtagene ciloleucel, [...] Read more.
Chimeric Antigen Receptor (CAR)-T cell therapy is an exciting development in the field of cancer immunology, wherein immune T-cells from patients are collected, engineered to create ‘CAR’-T cells, and infused back into the same patient. Currently, two CAR-T-cell-based therapies, Tisagenlecleucel and Axicabtagene ciloleucel, are approved by FDA for the treatment of hematological malignancies, acute lymphoblastic leukemia and large B-cell lymphomas. Their approval has been a culmination of several phase I and II clinical studies, which are the subject of discussion in this review article. Over the years, CAR-T cells have evolved to be significantly more persistent in patients’ blood, resulting in a much-improved clinical response and disease remission. This is particularly significant given that the target patient populations of these therapies are those with relapsed and refractory disease who have often progressed on multiple therapies. Despite the promising clinical results, there are still several challenges that need to be addressed. Of particular note are the associated toxicities exemplified by cytokine release syndrome (CRS) and the neurotoxicity. CRS has been addressed by an FDA-approved therapy of its own—tocilizumab. This article focuses on the progress related to CAR-T therapy: the pertinent clinical studies and their major findings, their associated adverse effects, and future perspective. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

15 pages, 1080 KiB  
Review
Finding the Keys to the CAR: Identifying Novel Target Antigens for T Cell Redirection Immunotherapies
by Rebecca C. Abbott, Ryan S. Cross and Misty R. Jenkins
Int. J. Mol. Sci. 2020, 21(2), 515; https://doi.org/10.3390/ijms21020515 - 14 Jan 2020
Cited by 48 | Viewed by 5727
Abstract
Oncology immunotherapy has been a significant advancement in cancer treatment and involves harnessing and redirecting a patient’s immune response towards their own tumour. Specific recognition and elimination of tumour cells was first proposed over a century ago with Paul Erlich’s ‘magic bullet’ theory [...] Read more.
Oncology immunotherapy has been a significant advancement in cancer treatment and involves harnessing and redirecting a patient’s immune response towards their own tumour. Specific recognition and elimination of tumour cells was first proposed over a century ago with Paul Erlich’s ‘magic bullet’ theory of therapy. In the past decades, targeting cancer antigens by redirecting T cells with antibodies using either bispecific T cell engagers (BiTEs) or chimeric antigen receptor (CAR) T cell therapy has achieved impressive clinical responses. Despite recent successes in haematological cancers, linked to a high and uniformly expressed CD19 antigen, the efficacy of T cell therapies in solid cancers has been disappointing, in part due to antigen escape. Targeting heterogeneous solid tumours with T cell therapies will require the identification of novel tumour specific targets. These targets can be found among a range of cell-surface expressed antigens, including proteins, glycolipids or carbohydrates. In this review, we will introduce the current tumour target antigen classification, outline existing approaches to discover novel tumour target antigens and discuss considerations for future design of antibodies with a focus on their use in CAR T cells. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Graphical abstract

15 pages, 3555 KiB  
Review
The Evolving Protein Engineering in the Design of Chimeric Antigen Receptor T Cells
by Hannah E. Hughes-Parry, Ryan S. Cross and Misty R. Jenkins
Int. J. Mol. Sci. 2020, 21(1), 204; https://doi.org/10.3390/ijms21010204 - 27 Dec 2019
Cited by 26 | Viewed by 19550
Abstract
The clinical success of chimeric antigen receptor (CAR) T cell immunotherapy in the treatment of haematological cancers has encouraged the extensive development of CAR design to improve their function and increase their applicability. Advancements in protein engineering have seen modifications to both the [...] Read more.
The clinical success of chimeric antigen receptor (CAR) T cell immunotherapy in the treatment of haematological cancers has encouraged the extensive development of CAR design to improve their function and increase their applicability. Advancements in protein engineering have seen modifications to both the ecto- and endo-domains of the CAR, with recent designs targeting multiple antigens and including inducible elements. These developments are likely to play an important role in inducing effective CAR T cell responses in a solid tumour context, where clinical responses have not been effective to date. This review highlights the spectrum of novel strategies being employed in CAR design, including for example variations in targeting tumour antigens by utilising different ectodomain designs such as dual chain CARs, natural receptor or ligand-based CARs, and T cell receptor fusion constructs, and also reviews some of the innovative approaches to a “universal” CAR and various multi-antigen targeting CAR strategies. We also explore how choices in the endodomain impact CAR function and how these need to be considered in the overall CAR design. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

21 pages, 1576 KiB  
Review
Optimizing Manufacturing Protocols of Chimeric Antigen Receptor T Cells for Improved Anticancer Immunotherapy
by Sophia Stock, Michael Schmitt and Leopold Sellner
Int. J. Mol. Sci. 2019, 20(24), 6223; https://doi.org/10.3390/ijms20246223 - 10 Dec 2019
Cited by 79 | Viewed by 13686
Abstract
Chimeric antigen receptor (CAR) T cell therapy can achieve outstanding response rates in heavily pretreated patients with hematological malignancies. However, relapses occur and they limit the efficacy of this promising treatment approach. The cellular composition and immunophenotype of the administered CART cells play [...] Read more.
Chimeric antigen receptor (CAR) T cell therapy can achieve outstanding response rates in heavily pretreated patients with hematological malignancies. However, relapses occur and they limit the efficacy of this promising treatment approach. The cellular composition and immunophenotype of the administered CART cells play a crucial role for therapeutic success. Less differentiated CART cells are associated with improved expansion, long-term in vivo persistence, and prolonged anti-tumor control. Furthermore, the ratio between CD4+ and CD8+ T cells has an effect on the anti-tumor activity of CART cells. The composition of the final cell product is not only influenced by the CART cell construct, but also by the culturing conditions during ex vivo T cell expansion. This includes different T cell activation strategies, cytokine supplementation, and specific pathway inhibition for the differentiation blockade. The optimal production process is not yet defined. In this review, we will discuss the use of different CART cell production strategies and the molecular background for the generation of improved CART cells in detail. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

22 pages, 2171 KiB  
Review
CSPG4 as Target for CAR-T-Cell Therapy of Various Tumor Entities–Merits and Challenges
by Dennis C. Harrer, Jan Dörrie and Niels Schaft
Int. J. Mol. Sci. 2019, 20(23), 5942; https://doi.org/10.3390/ijms20235942 - 26 Nov 2019
Cited by 36 | Viewed by 5631
Abstract
Targeting cancer cells using chimeric-antigen-receptor (CAR-)T cells has propelled adoptive T-cell therapy (ATT) to the next level. A plentitude of durable complete responses using CD19-specific CAR-T cells in patients suffering from various lymphoid malignancies resulted in the approval by the food and drug [...] Read more.
Targeting cancer cells using chimeric-antigen-receptor (CAR-)T cells has propelled adoptive T-cell therapy (ATT) to the next level. A plentitude of durable complete responses using CD19-specific CAR-T cells in patients suffering from various lymphoid malignancies resulted in the approval by the food and drug administration (FDA) of CD19-directed CAR-T cells for the treatment of acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). A substantial portion of this success in hematological malignancies can be traced back to the beneficial properties of the target antigen CD19, which combines a universal presence on target cells with no detectable expression on indispensable host cells. Hence, to replicate response rates achieved in ALL and DLBCL in the realm of solid tumors, where ideal target antigens are scant and CAR-T cells are still lagging behind expectations, the quest for appropriate target antigens represents a crucial task to expedite the next steps in the evolution of CAR-T-cell therapy. In this review, we want to highlight the potential of chondroitin sulfate proteoglycan 4 (CSPG4) as a CAR-target antigen for a variety of different cancer entities. In particular, we discuss merits and challenges associated with CSPG4-CAR-T cells for the ATT of melanoma, leukemia, glioblastoma, and triple-negative breast cancer. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

28 pages, 1233 KiB  
Review
Beyond the Cell Surface: Targeting Intracellular Negative Regulators to Enhance T cell Anti-Tumor Activity
by Poojitha Sitaram, Bradley Uyemura, Subramaniam Malarkannan and Matthew J. Riese
Int. J. Mol. Sci. 2019, 20(23), 5821; https://doi.org/10.3390/ijms20235821 - 20 Nov 2019
Cited by 21 | Viewed by 7594
Abstract
It is well established that extracellular proteins that negatively regulate T cell function, such as Cytotoxic T-Lymphocyte-Associated protein 4 (CTLA-4) and Programmed Cell Death protein 1 (PD-1), can be effectively targeted to enhance cancer immunotherapies and Chimeric Antigen Receptor T cells (CAR-T cells). [...] Read more.
It is well established that extracellular proteins that negatively regulate T cell function, such as Cytotoxic T-Lymphocyte-Associated protein 4 (CTLA-4) and Programmed Cell Death protein 1 (PD-1), can be effectively targeted to enhance cancer immunotherapies and Chimeric Antigen Receptor T cells (CAR-T cells). Intracellular proteins that inhibit T cell receptor (TCR) signal transduction, though less well studied, are also potentially useful therapeutic targets to enhance T cell activity against tumor. Four major classes of enzymes that attenuate TCR signaling include E3 ubiquitin kinases such as the Casitas B-lineage lymphoma proteins (Cbl-b and c-Cbl), and Itchy (Itch), inhibitory tyrosine phosphatases, such as Src homology region 2 domain-containing phosphatases (SHP-1 and SHP-2), inhibitory protein kinases, such as C-terminal Src kinase (Csk), and inhibitory lipid kinases such as Src homology 2 (SH2) domain-containing inositol polyphosphate 5-phosphatase (SHIP) and Diacylglycerol kinases (DGKs). This review describes the mechanism of action of eighteen intracellular inhibitory regulatory proteins in T cells within these four classes, and assesses their potential value as clinical targets to enhance the anti-tumor activity of endogenous T cells and CAR-T cells. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

23 pages, 1059 KiB  
Review
CAR-Based Strategies beyond T Lymphocytes: Integrative Opportunities for Cancer Adoptive Immunotherapy
by Ramona Rotolo, Valeria Leuci, Chiara Donini, Anna Cykowska, Loretta Gammaitoni, Giovanni Medico, Giorgio Valabrega, Massimo Aglietta and Dario Sangiolo
Int. J. Mol. Sci. 2019, 20(11), 2839; https://doi.org/10.3390/ijms20112839 - 11 Jun 2019
Cited by 32 | Viewed by 6342
Abstract
Chimeric antigen receptor (CAR)-engineered T lymphocytes (CAR Ts) produced impressive clinical results against selected hematological malignancies, but the extension of CAR T cell therapy to the challenging field of solid tumors has not, so far, replicated similar clinical outcomes. Many efforts are currently [...] Read more.
Chimeric antigen receptor (CAR)-engineered T lymphocytes (CAR Ts) produced impressive clinical results against selected hematological malignancies, but the extension of CAR T cell therapy to the challenging field of solid tumors has not, so far, replicated similar clinical outcomes. Many efforts are currently dedicated to improve the efficacy and safety of CAR-based adoptive immunotherapies, including application against solid tumors. A promising approach is CAR engineering of immune effectors different from αβT lymphocytes. Herein we reviewed biological features, therapeutic potential, and safety of alternative effectors to conventional CAR T cells: γδT, natural killer (NK), NKT, or cytokine-induced killer (CIK) cells. The intrinsic CAR-independent antitumor activities, safety profile, and ex vivo expansibility of these alternative immune effectors may favorably contribute to the clinical development of CAR strategies. The proper biological features of innate immune response effectors may represent an added value in tumor settings with heterogeneous CAR target expression, limiting the risk of tumor clonal escape. All these properties bring out CAR engineering of alternative immune effectors as a promising integrative option to be explored in future clinical studies. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

20 pages, 935 KiB  
Review
HBV Immune-Therapy: From Molecular Mechanisms to Clinical Applications
by Carolina Boni, Valeria Barili, Greta Acerbi, Marzia Rossi, Andrea Vecchi, Diletta Laccabue, Amalia Penna, Gabriele Missale, Carlo Ferrari and Paola Fisicaro
Int. J. Mol. Sci. 2019, 20(11), 2754; https://doi.org/10.3390/ijms20112754 - 05 Jun 2019
Cited by 43 | Viewed by 7091
Abstract
Chronic hepatitis B virus (HBV) infection represents a worldwide public health concern with approximately 250 million people chronically infected and at risk of developing liver cirrhosis and hepatocellular carcinoma. Nucleos(t)ide analogues (NUC) are the most widely used therapies for HBV infection, but they [...] Read more.
Chronic hepatitis B virus (HBV) infection represents a worldwide public health concern with approximately 250 million people chronically infected and at risk of developing liver cirrhosis and hepatocellular carcinoma. Nucleos(t)ide analogues (NUC) are the most widely used therapies for HBV infection, but they often require long-lasting administration to avoid the risk of HBV reactivation at withdrawal. Therefore, there is an urgent need to develop novel treatments to shorten the duration of NUC therapy by accelerating virus control, and to complement the effect of available anti-viral therapies. In chronic HBV infection, virus-specific T cells are functionally defective, and this exhaustion state is a key determinant of virus persistence. Reconstitution of an efficient anti-viral T cell response may thus represent a rational strategy to treat chronic HBV patients. In this perspective, the enhancement of adaptive immune responses by a checkpoint inhibitor blockade, specific T cell vaccines, lymphocyte metabolism targeting, and autologous T cell engineering, including chimeric antigen receptor (CAR) and TCR-redirected T cells, constitutes a promising immune modulatory approach for a therapeutic restoration of protective immunity. The advances of the emerging immune-based therapies in the setting of the HBV research field will be outlined. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Graphical abstract

28 pages, 1952 KiB  
Review
Dendritic Cell-Mediated Th2 Immunity and Immune Disorders
by Sunil Kumar, Yideul Jeong, Muhammad Umer Ashraf and Yong-Soo Bae
Int. J. Mol. Sci. 2019, 20(9), 2159; https://doi.org/10.3390/ijms20092159 - 01 May 2019
Cited by 62 | Viewed by 9621
Abstract
Dendritic cells (DCs) are the professional antigen-presenting cells that recognize and present antigens to naïve T cells to induce antigen-specific adaptive immunity. Among the T-cell subsets, T helper type 2 (Th2) cells produce the humoral immune responses required for protection against helminthic disease [...] Read more.
Dendritic cells (DCs) are the professional antigen-presenting cells that recognize and present antigens to naïve T cells to induce antigen-specific adaptive immunity. Among the T-cell subsets, T helper type 2 (Th2) cells produce the humoral immune responses required for protection against helminthic disease by activating B cells. DCs induce a Th2 immune response at a certain immune environment. Basophil, eosinophil, mast cells, and type 2 innate lymphoid cells also induce Th2 immunity. However, in the case of DCs, controversy remains regarding which subsets of DCs induce Th2 immunity, which genes in DCs are directly or indirectly involved in inducing Th2 immunity, and the detailed mechanisms underlying induction, regulation, or maintenance of the DC-mediated Th2 immunity against allergic environments and parasite infection. A recent study has shown that a genetic defect in DCs causes an enhanced Th2 immunity leading to severe atopic dermatitis. We summarize the Th2 immune-inducing DC subsets, the genetic and environmental factors involved in DC-mediated Th2 immunity, and current therapeutic approaches for Th2-mediated immune disorders. This review is to provide an improved understanding of DC-mediated Th2 immunity and Th1/Th2 immune balancing, leading to control over their adverse consequences. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

15 pages, 1130 KiB  
Review
CAR-T with License to Kill Solid Tumors in Search of a Winning Strategy
by Benedetto Sacchetti, Andrea Botticelli, Luca Pierelli, Marianna Nuti and Maurizio Alimandi
Int. J. Mol. Sci. 2019, 20(8), 1903; https://doi.org/10.3390/ijms20081903 - 17 Apr 2019
Cited by 15 | Viewed by 6271
Abstract
Artificial receptors designed for adoptive immune therapies need to absolve dual functions: antigen recognition and abilities to trigger the lytic machinery of reprogrammed effector T lymphocytes. In this way, CAR-T cells deliver their cytotoxic hit to cancer cells expressing targeted tumor antigens, bypassing [...] Read more.
Artificial receptors designed for adoptive immune therapies need to absolve dual functions: antigen recognition and abilities to trigger the lytic machinery of reprogrammed effector T lymphocytes. In this way, CAR-T cells deliver their cytotoxic hit to cancer cells expressing targeted tumor antigens, bypassing the limitation of HLA-restricted antigen recognition. Expanding technologies have proposed a wide repertoire of soluble and cellular “immunological weapons” to kill tumor cells; they include monoclonal antibodies recognizing tumor associated antigens on tumor cells and immune cell checkpoint inhibition receptors expressed on tumor specific T cells. Moreover, a wide range of formidable chimeric antigen receptors diversely conceived to sustain quality, strength and duration of signals delivered by engineered T cells have been designed to specifically target tumor cells while minimize off-target toxicities. The latter immunological weapons have shown distinct efficacy and outstanding palmarès in curing leukemia, but limited and durable effects for solid tumors. General experience with checkpoint inhibitors and CAR-T cell immunotherapy has identified a series of variables, weaknesses and strengths, influencing the clinical outcome of the oncologic illness. These aspects will be shortly outlined with the intent of identifying the still “missing strategy” to combat epithelial cancers. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

Other

11 pages, 2222 KiB  
Case Report
Oligoclonal T Cells Transiently Expand and Express Tim-3 and PD-1 Following Anti-CD19 CAR T Cell Therapy: A Case Report
by Christopher Ronald Funk, Christopher T. Petersen, Neera Jagirdar, Sruthi Ravindranathan, David L. Jaye, Christopher R. Flowers, Amelia Langston and Edmund K. Waller
Int. J. Mol. Sci. 2018, 19(12), 4118; https://doi.org/10.3390/ijms19124118 - 19 Dec 2018
Cited by 7 | Viewed by 4680
Abstract
Clinical trials of chimeric antigen receptor (CAR) T cells in hematologic malignancy associate remissions with two profiles of CAR T cell proliferation kinetics, which differ based upon costimulatory domain. Additional T cell intrinsic factors that influence or predict clinical response remain unclear. To [...] Read more.
Clinical trials of chimeric antigen receptor (CAR) T cells in hematologic malignancy associate remissions with two profiles of CAR T cell proliferation kinetics, which differ based upon costimulatory domain. Additional T cell intrinsic factors that influence or predict clinical response remain unclear. To address this gap, we report the case of a 68-year-old woman with refractory/relapsed diffuse large B cell lymphoma (DLBCL), treated with tisagenlecleucel (anti-CD19), with a CD137 costimulatory domain (4-1BB) on an investigational new drug application (#16944). For two months post-infusion, the patient experienced dramatic regression of subcutaneous nodules of DLBCL. Unfortunately, her CAR T exhibited kinetics unassociated with remission, and she died of DLBCL-related sequelae. Serial phenotypic analysis of peripheral blood alongside sequencing of the β-peptide variable region of the T cell receptor (TCRβ) revealed distinct waves of oligoclonal T cell expansion with dynamic expression of immune checkpoint molecules. One week prior to CAR T cell contraction, T cell immunoglobulin mucin domain 3 (Tim-3) and programmed cell death protein 1 (PD-1) exhibited peak expressions on both the CD8 T cell (Tim-3 ≈ 50%; PD-1 ≈ 17%) and CAR T cell subsets (Tim-3 ≈ 78%; PD-1 ≈ 40%). These correlative observations draw attention to Tim-3 and PD-1 signaling pathways in context of CAR T cell exhaustion. Full article
(This article belongs to the Special Issue CAR-T Cell Therapy)
Show Figures

Figure 1

Back to TopTop