Elsevier

Journal of Controlled Release

Volume 353, January 2023, Pages 875-888
Journal of Controlled Release

Cell delivery devices for cancer immunotherapy

https://doi.org/10.1016/j.jconrel.2022.11.041Get rights and content

Abstract

Adoptive cell therapy (ACT) that leverages allogeneic or autologous immune cells holds vast promise in targeted cancer therapy. Despite the tremendous success of ACT in treating hematopoietic malignancies, its efficacy is limited in eradicating solid tumors via intravenous infusion of immune cells. With the extending technology of cancer immunotherapy, novel delivery strategies have been explored to improve the therapeutic potency of adoptively transferred cells for solid tumor treatment by innovating the administration route, maintaining the cell viability, and normalizing the tumor microenvironment. In this review, a variety of devices for cell delivery are summarized. Perspectives and challenges of cell delivery devices for cancer immunotherapy are also discussed.

Introduction

Cancer immunotherapy educates, activates, and boosts the immune system to recognize, fight, and eliminate cancer cells [1]. To date, many treatment strategies have been developed, including immune checkpoint inhibitors [2], adoptively transferred immune cells [3], bispecific antibodies [4], oncolytic virus [5], bacteria [6], cancer vaccines [7], and immune system modulators [8]. Among these different approaches, the engineered immune cell-based ACT, especially the chimeric antigen receptor T cells (CAR-T) therapy, has emerged as a curative and powerful treatment. In addition, other promising types of ACT under investigation or in clinical application include T cell receptor engineered T cells (TCR-T), tumor-infiltrating lymphocytes (TIL), chimeric antigen receptor engineered natural killer cells (CAR-NK), and chimeric antigen receptor engineered macrophages (CAR-macrophages) [9].

In ACT therapy, the patient or donor immune cells are often genetically engineered to target cancer cells except using patient TIL [10]. After ex vivo expansion, the cells are infused into patients to eliminate cancer cells. Multiple hematologic malignancies, including B-cell acute lymphoblastic leukemia, B-cell non-Hodgkin lymphoma, and multiple myeloma, have been treated by ACT with remarkable clinical efficacy [[11], [12], [13]]. Notably, CAR-T therapy targeting B-lymphocyte antigen CD19 has exhibited unprecedented potency with 50–90% complete response rates for refractory B-cell malignancies in several clinical trials [[14], [15], [16], [17]]. This impressive treatment efficacy has prompted a focus on developing strategies to extend the application of CAR-T cells to other non-hematologic cancers. However, it remains challenging for CAR-T therapy to treat solid tumors due to the immunosuppressive tumor microenvironment, insufficient tumor infiltration of T cells, and tumor antigen heterogeneity [13]. In details, the compact structure of solid tumor contains numerous cell types, such as fibroblasts and immune inhibitory cells [18]. The nitration of the chemokine CCL2 is induced by reactive nitrogen species, resulting in the trapping of T cells in the stroma. Cancer-associated fibroblasts can produce CXCL12 to coat around tumor cells. The protection layer excludes T cells from attacking cancer cells. The tumor vasculature also expresses Fas ligand that induces apoptosis of effector T cells express Fas death receptor [19]. In addition, various immunosuppressive molecules in solid tumors, such as anti-inflammatory cytokines and immune checkpoints, can promptly downregulate the tumoricidal efficacy of T cells and further induce their exhaustion [20]. Furthermore, targeted tumor antigens may only distribute in a portion of tumor cells due to antigen heterogeneity in solid tumors or because cancer cells can evolve to reduce the expression of these antigens. In this case, T cells cannot recognize and eliminate all tumor cells, which eventually leads to tumor recurrences [21]. Current administration of CAR-T cells mainly relies on intravenous injection, which may result in insufficient tumor infiltration. Furthermore, side effects, such as acute kidney injury, bleeding episode, heart arrhythmia, cytokine storm, and neurotoxicity, also compromise the therapeutic outcome and prognosis [22,23].

To this end, cell delivery devices, including thin films, microparticles, scaffolds and microneedles have been exploited to enhance the viability and therapeutic efficacy of immune cells, while minimize side effects. The device assists immune cells to reach tumors directly, provides a location for delivered cells to reside, proliferate, and/or perform anti-tumor activities. Immune cells can be released into tumor tissues through active migration in response to the chemokines derived from tumor tissues or simply be released via diffusion with or without the facilitation of device degradation. Such manners can deliver essential amounts of therapeutic cells into tumor tissues within a short period of time or over days, which, to some extent, can improve the infiltration of immune cells into tumor tissues and/or solve the problem of cytokine storm. In addition, cell delivery devices can be loaded with immunomodulating drugs to support immune cells for enhanced proliferation and tumor elimination as well as normalize tumor immunosuppressive microenvironment for long-term preservation of cell bioactivities [24].

In this review, we summarize the devices for delivering diverse cell-based therapeutics (Fig. 1). First, we briefly discuss the design criteria for cell delivery devices such as key device characteristics and main types of device materials. Then, the typical devices that have been applied to cell delivery and cancer immunotherapy, particularly the scaffold, are discussed. Finally, we highlight the perspectives and challenges of cell delivery devices in cancer treatment, including the development of new delivery devices, evaluation of device biocompatibility and immunogenicity, and their applications in combination with other cancer therapies.

Section snippets

Design criteria of devices in cell delivery

Cell delivery devices should often meet the following criteria, including (1) excellent biocompatibility of devices and their degradation products, (2) adequate room for the cell loading, in-growth, and bio-substances transportation, (3) appropriate mechanical property, (4) optimal loading capacity of therapeutic agents such as antibodies and small drug molecules, (5) biological stability under physiological condition (temperature, biological activities, and signals), (6) suitable protection of

Major types of cell delivery devices

To date, a broad range of cell delivery devices have been developed. In this section, we review the reported cell delivery devices based on their shape and structure, which are the major factors to determine how devices are applied in cancer treatment. It is worth noting that same materials can be fabricated into devices with different shapes and structures.

Conclusion and perspectives

ACT has exhibited great potential in treating distinct hematologic tumors, but it has not met its expectation in solid tumor treatment due to a variety of limitations, such as poor infiltration in tumors, suppressed cell activity, and toxicity. Biomaterials-based delivery strategies that can maintain cell viability, increase cell activity, and reduce systemic toxicity are expected to break the bottleneck of ACT in solid tumor treatment.

To date, diverse strategies have been developed to meet

Author contributions

Z.G., H.L., H.C., and P.W. conceived the project. P.W. and W.W. contributed equally. H.Z., and Z.L. performed the schematic drawing. All authors contributed to writing and revising the manuscript and approved the final version.

Acknowledgments

Z.G. and H.L. would like to acknowledge the support from the National Key R&D Program of China (2021YFA0909900), the National Natural Science Foundation of China (52173142), and the grants from the Startup Package of Zhejiang University. H.C. thanks the support from Drexel University.

References (126)

  • Y.H. Ahn et al.

    A three-dimensional hyaluronic acid-based niche enhances the therapeutic efficacy of human natural killer cell-based cancer immunotherapy

    Biomaterials

    (2020)
  • A. Monette et al.

    Chitosan thermogels for local expansion and delivery of tumor-specific T lymphocytes towards enhanced cancer immunotherapies

    Biomaterials

    (2016)
  • S. Karki et al.

    Thin films as an emerging platform for drug delivery

    Asian J. Pharm. Sci.

    (2016)
  • M. Li et al.

    Electrospun protein fibers as matrices for tissue engineering

    Biomaterials

    (2005)
  • M. Santoro et al.

    Gelatin carriers for drug and cell delivery in tissue engineering

    J. Control. Release

    (2014)
  • M. Shayan et al.

    An overview of thin film nitinol endovascular devices

    Acta Biomater.

    (2015)
  • P. de Vos et al.

    Alginate-based microcapsules for immunoisolation of pancreatic islets

    Biomaterials

    (2006)
  • M. Farina et al.

    Cell encapsulation: overcoming barriers in cell transplantation in diabetes and beyond

    Adv. Drug Deliv. Rev.

    (2019)
  • P.B. Malafaya et al.

    Natural-origin polymers as carriers and scaffolds for biomolecules and cell delivery in tissue engineering applications

    Adv. Drug Deliv. Rev.

    (2007)
  • Y. Li et al.

    Advances in biodegradable and injectable hydrogels for biomedical applications

    J. Control. Release

    (2021)
  • A.S. Hoffman

    Hydrogels for biomedical applications

    Adv. Drug Deliv. Rev.

    (2012)
  • J.D. Kretlow et al.

    Injectable matrices and scaffolds for drug delivery in tissue engineering

    Adv. Drug Deliv. Rev.

    (2007)
  • T. Sheng et al.

    Microneedle-mediated vaccination: innovation and translation

    Adv. Drug Deliv. Rev.

    (2021)
  • A.D. Waldman et al.

    A guide to cancer immunotherapy: from T cell basic science to clinical practice

    Nat. Rev. Immunol.

    (2020)
  • D.M. Pardoll

    The blockade of immune checkpoints in cancer immunotherapy

    Nat. Rev. Cancer

    (2012)
  • S.A. Rosenberg et al.

    Adoptive cell transfer: a clinical path to effective cancer immunotherapy

    Nat. Rev. Cancer

    (2008)
  • A.F. Labrijn et al.

    Bispecific antibodies: a mechanistic review of the pipeline

    Nat. Rev. Drug Discov.

    (2019)
  • H. Fukuhara et al.

    Oncolytic virus therapy: a new era of cancer treatment at dawn

    Cancer Sci.

    (2016)
  • C.R. Gurbatri et al.

    Engineered probiotics for local tumor delivery of checkpoint blockade nanobodies

    Sci. Transl. Med.

    (2020)
  • M. Saxena et al.

    Therapeutic cancer vaccines

    Nat. Rev. Cancer

    (2021)
  • I. Mellman et al.

    Cancer immunotherapy comes of age

    Nature

    (2011)
  • M. Klichinsky et al.

    Human chimeric antigen receptor macrophages for cancer immunotherapy

    Nat. Biotechnol.

    (2020)
  • D. Li et al.

    Genetically engineered T cells for cancer immunotherapy

    Signal Transduct. Target. Ther.

    (2019)
  • S. Atrash et al.

    CAR-T treatment for hematological malignancies

    J. Investig. Med.

    (2020)
  • R.A. Morgan et al.

    Cancer regression in patients after transfer of genetically engineered lymphocytes

    Science

    (2006)
  • L. Labanieh et al.

    Programming CAR-T cells to kill cancer

    Nat. Biomed. Eng.

    (2018)
  • C.J. Turtle et al.

    Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells

    Sci. Transl. Med.

    (2016)
  • R.J. Brentjens et al.

    CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia

    Sci. Transl. Med.

    (2013)
  • C.J. Turtle et al.

    CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients

    J. Clin. Invest.

    (2016)
  • M. Binnewies et al.

    Understanding the tumor immune microenvironment (TIME) for effective therapy

    Nat. Med.

    (2018)
  • J.A. Joyce et al.

    T cell exclusion, immune privilege, and the tumor microenvironment

    Science

    (2015)
  • R. Fu et al.

    Delivery techniques for enhancing CAR T cell therapy against solid tumors

    Adv. Funct. Mater.

    (2021)
  • A.J. Hou et al.

    Navigating CAR-T cells through the solid-tumour microenvironment

    Nat. Rev. Drug Discov.

    (2021)
  • N.N. Shah et al.

    Mechanisms of resistance to CAR T cell therapy

    Nat. Rev. Clin. Oncol.

    (2019)
  • Y. Ding et al.

    Recent advances in overcoming barriers to cell-based delivery systems for cancer immunotherapy

    Exploration

    (2022)
  • G. Orive et al.

    Cell encapsulation: promise and progress

    Nat. Med.

    (2003)
  • T. Garg et al.

    Biomaterials-based nanofiber scaffold: targeted and controlled carrier for cell and drug delivery

    J. Drug Target.

    (2015)
  • O.S. Fenton et al.

    Advances in biomaterials for drug delivery

    Adv. Mater.

    (2018)
  • Y. Tabata

    Biomaterial technology for tissue engineering applications

    J. R. Soc. Interface

    (2009)
  • J. Ma et al.

    Bioactive inorganic particles-based biomaterials for skin tissue engineering

    Exploration

    (2022)
  • Cited by (7)

    View all citing articles on Scopus
    1

    These authors made equal contributions to this work.

    View full text