Elsevier

Seminars in Cancer Biology

Volume 69, February 2021, Pages 307-324
Seminars in Cancer Biology

Nanoparticle mediated cancer immunotherapy

https://doi.org/10.1016/j.semcancer.2020.03.015Get rights and content

Abstract

The versatility and nanoscale size have helped nanoparticles (NPs) improve the efficacy of conventional cancer immunotherapy and opened up exciting approaches to combat cancer. This review first outlines the tumor immune evasion and the defensive tumor microenvironment (TME) that hinders the activity of host immune system against tumor. Then, a detailed description on how the NP based strategies have helped improve the efficacy of conventional cancer vaccines and overcome the obstacles led by TME. Sustained and controlled drug delivery, enhanced cross presentation by immune cells, co-encapsulation of adjuvants, inhibition of immune checkpoints and intrinsic adjuvant like properties have aided NPs to improve the therapeutic efficacy of cancer vaccines. Also, NPs have been efficient modulators of TME. In this context, NPs facilitate better penetration of the chemotherapeutic drug by dissolution of the inhibitory meshwork formed by tumor associated cells, blood vessels, soluble mediators and extra cellular matrix in TME. NPs achieve this by suppression, modulation, or reprogramming of the immune cells and other mediators localised in TME. This review further summarizes the applications of NPs used to enhance the efficacy of cancer vaccines and modulate the TME to improve cancer immunotherapy. Finally, the hurdles faced in commercialization and translation to clinic have been discussed and intriguingly, NPs owe great potential to emerge as clinical formulations for cancer immunotherapy in near future.

Introduction

The current surge and significance of cancer immunotherapy can be realised by the 2018 Nobel Prize in Physiology or Medicine which was jointly awarded to James P. Allison and Tasuku Honjo for the discovery of inhibition of negative immune regulation. Cancer immunotherapy exclusively employs immune cells to target specific tumor cells sparing normal tissues. In comparison to conventional cancer treatment regimens, immunotherapy is more effective owing to need for activation of only a few thousand immune cells versus action against a million tumor cells in the former. It can systematically target both primary tumor cells as well as secondary metastatic tumors. Here both the host immune cells in lymphoid tissues and antitumor immune cells in tumor microenvironment (TME) are triggered to browse and destroy tumor cells. Besides, development of memory makes it even more promising against possible recurrence. The enhancement of the monitoring activity by the immune system can lead to an increase in the defense mechanisms of other parts of the body, which can cause inflammation [1]. Moreover, certain therapies may require multiple doses to achieve concentrations that are biologically relevant in the target tissues and this may lead to toxicities. Thus, the current efforts aim for further development to enhance specificity, effectiveness and reduce toxicity.

The advent of nanotechnology holds promising opportunities for safer, specific and more effective cancer immunotherapy. The flexibility to tune biodistribution, biocompatibility, specific targeting, immunogenicity, controlled loading, sustained release kinetics, minimal degradation of bioactive molecules, controlled spatiotemporal delivery profiles along with adjuvants has embraced the nanoparticle (NP) based therapy to emerge as an efficient tool [2,3].

Section snippets

Immune surveillance and tumor immune evasion

The immune system has a stringent control over ‘self’ and ‘non-self’ antigens. However, tumor cells arise due to mutations and altered pattern of differentiation. Hence, tumor cells can be better termed as ‘abnormal self’ cells. The two arms of immune system (nonspecific innate and specific adaptive) work in synergy to eliminate the ‘non-self’/ foreign or tumor cells. The ‘self’ cells are further protected in a process called negative selection in which the B and T lymphocyte clones that

NPs in cancer immunotherapy

The leaky vasculature in the TME results in enhanced permeation and retention effect for particles with small sizes ranging in few hundred nanometers. Smaller size and larger specific surface area provide NPs with efficient loading capacities. Several reports suggest that nanomaterials prevent degradation of payload from the acidic and proteolytic TME [35]. Antigens along with adjuvants can be co-delivered in NPs to optimize the effects, resulting in a sustained depot of antigen and therefore

NPs as delivery systems for antigens

NPs offer advantages of greater cellular uptake because of the EPR effect and enhanced immune response as compared to free peptide or protein-based cancer vaccines [52]. In conventional cancer vaccines, delivery of antigens and adjuvants separately in free forms may cause immune tolerance due to lack of danger signals to DCs at appropriate time [53,54]. NPs therefore promote activation of DCs by facilitating co-encapsulation of antigens and adjuvants [55]. Molino et al. [56] reported that E2

NPs assisted adoptive T cell therapy

Adoptive T cell therapy (ACT) involves isolation of immune cells from cancer or normal patients, followed by ex vivo genetic or chemical modulation of cells and reinjection back to the patient to activate the immune system and combat cancer. Two chimeric antigen receptor (CAR)-T therapies, Yescarta and Kymriah for lymphoma and leukemia respectively have been approved by FDA. However, the ACT suffers from major challenges which includes manufacturing difficulties, the immunosuppressive TME and

NPs mediated dissolution of ECM

The extracellular matrix (ECM) which comprises of hyaluronic acid, proteoglycan, fibronectin and collagen, acts as the main barrier for the diffusion of NPs. Use of inhibitors [116,117] or solubilising agents for these components enhances penetration of NPs [118]. Co-administration of hyaluronidase with NPs helpsincrease tumor penetration. A 4T1 breast tumor bearing mice when injected with hyaluronidase intratumorally, followed by PEG coated photosensitizer chlorine 6 (Ce6) containing

NPs mediated targeting of negative immune checkpoints

The checkpoint blockade with specific mAbs facilitates inhibition of pathways that keep the duration and strength of immune system in check. Inhibition of these checkpoint molecules work by re-educating the adaptive immune system and selectively eliminating primary and metastatic tumors. CTLA4, PD1, PDL-1, PDL-2 inhibitors are the major targets for immunotherapy. PD1 is considered a better target than CTLA4 owing to mitigated side effects and higher response rates demonstrated in tumor patient

NPs as artificial antigen presenting cells

Besides the targeting, modulation and reprogramming of innate immune cells present in TME, the immunosuppression can be relieved if the cytotoxic T cell population can be increased in the TME. One such approach involves delivering artificial antigen presenting cells (aAPCs) which can activate and promote expansion of antigen specific CD8+ T cells [208]. The NPs surface are coated with MHC-I with the peptide antigen (signal 1) and the costimulatory molecules B7.1, 4-1BBL (signal 2) [209]/

Challenges in commercialization

The major goal of the research and development in area of nanomedicines for cancer lies in its translation to clinics which is justified by FDA approval [215]. However, the path to clinical translation is sturdy and requires major hurdles to be passed at every level. This is one of the major reasons that despite much research and preliminary promising preclinical results, not many nanomedicines have reached the market [216]. It is imperative to assess the fate of nanomedicines and its

Concluding remarks

A plethora of literature is available on NP mediated cancer immunotherapy. Despite such an extensive documentation available, currently there are no NPs based prophylactic or therapeutic cancer vaccines translated to clinic. To translate the efficiency of NPs as clinical drug, advanced studies solving mechanism of NPs based strategies have to be worked upon at a faster pace. Also, in vivo studies involving stringent controls are to be performed to facilitate proper assessment about safety and

Funding source

None.

Declaration of Competing Interest

Authors declare that there exists no conflict of interest.

References (229)

  • T. Seremet et al.

    Tumor-specific antigens and immunologic adjuvants in cancer immunotherapy

    Cancer J.

    (2011)
  • S. Ahrens et al.

    F-actin is an evolutionarily conserved damage-associated molecular pattern recognized by DNGR-1, a receptor for dead cells

    Immunity

    (2012)
  • T.F. Gajewski et al.

    Innate and adaptive immune cells in the tumor microenvironment

    Nat. Immunol.

    (2013)
  • A.R. Lam et al.

    RAE1 ligands for the NKG2D receptor are regulated by STING-dependent DNA sensor pathways in lymphoma

    Cancer Res.

    (2014)
  • M.E. Bianchi

    Killing cancer cells, twice with one shot

    Cell Death Differ.

    (2014)
  • I. Cebrian et al.

    Sec22b regulates phagosomal maturation and antigen crosspresentation by dendritic cells

    Cell

    (2011)
  • M. Samie et al.

    The transcription factor TFEB acts as a molecular switch that regulates exogenous antigen-presentation pathways

    Nat. Immunol.

    (2015)
  • K.L. Rock

    The ins and outs of cross-presentation

    Nat. Immunol.

    (2003)
  • O.P. Joffre et al.

    Cross-presentation by dendritic cells

    Nat. Rev. Immunol.

    (2012)
  • S.L. Jongbloed et al.

    Human CD141 + (BDCA-3) + dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens

    J. Exp. Med.

    (2010)
  • J.A. Trapani et al.

    Functional significance of the perforin/granzyme cell death pathway

    Nat. Rev. Immunol.

    (2002)
  • J. Zhu et al.

    Differentiation of effector CD4 T Cell Populations

    Annu. Rev. Immunol.

    (2010)
  • K.M. Huster et al.

    Selective expression of IL-7 receptor on memory T cells identifies early CD40L-dependent generation of distinct CD8+ memory T cell subsets

    Proc. Natl. Acad. Sci. U. S. A.

    (2004)
  • S.P. Schoenberger et al.

    T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD4OL interactions

    Nature

    (1998)
  • B. Seliger et al.

    Antigen-processing machinery breakdown and tumor growth

    Immunol. Today

    (2000)
  • W. Zou

    Immunosuppressive networks in the tumour environment and their therapeutic relevance

    Nat. Rev. Cancer

    (2005)
  • A.S. Bear et al.

    Elimination of metastatic melanoma using gold nanoshell-enabled photothermal therapy and adoptive t cell transfer

    PLoS One

    (2013)
  • A. Wilkerson et al.

    Nanoparticle systems modulating myeloid-derived suppressor cells for cancer immunotherapy

    Curr. Top. Med. Chem.

    (2017)
  • L.C. Kennedy et al.

    T cells enhance gold nanoparticle delivery to tumors in vivo

    Nanoscale Res. Lett.

    (2011)
  • S.K. Rajendrakumar et al.

    Nanoparticle-based phototriggered cancer immunotherapy and its domino effect in the tumor microenvironment

    Biomacromolecules

    (2018)
  • M. Nesbit et al.

    Low-level monocyte chemoattractant protein-1 stimulation of monocytes leads to tumor formation in nontumorigenic melanoma cells

    J. Immunol.

    (2001)
  • M. Ruff et al.

    Neuropeptides are chemoattractants for human tumor cells and monocytes: a possible mechanism for metastasis

    Clin. Immunol. Immunopathol.

    (1985)
  • A. Mantovani et al.

    Role of tumor-associated macrophages in tumor progression and invasion

    Cancer Metastasis Rev.

    (2006)
  • M. Ray et al.

    Inhibition of Granzyme B by PI-9 protects prostate cancer cells from apoptosis

    Prostate

    (2012)
  • D.H. Munn et al.

    The tumor-draining lymph node as an immune-privileged site

    Immunol. Rev.

    (2006)
  • J. Gadiot et al.

    Overall survival and PD-L1 expression in metastasized malignant melanoma

    Cancer

    (2011)
  • G.D. Basu et al.

    Cyclooxygenase-2 inhibitor enhances the efficacy of a breast cancer vaccine: role of IDO

    J. Immunol.

    (2006)
  • S.Y. Lee et al.

    The immune tolerance of cancer is mediated by IDO that is inhibited by COX-2 inhibitors through regulatory T cells

    J. Immunother.

    (2009)
  • N. Obermajer et al.

    Positive feedback between PGE2 and COX2 redirects the differentiation of human dendritic cells toward stable myeloid-derived suppressor cells

    Blood

    (2011)
  • A. Sica et al.

    Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy

    Eur. J. Cancer

    (2006)
  • J. Liu et al.

    Nanoparticle-Based Nanomedicines to Promote Cancer Immunotherapy: Recent Advances and Future Directions

    Small

    (2019)
  • I.H. Lee et al.

    Imageable antigen-presenting gold nanoparticle vaccines for effective cancer immunotherapy in vivo

    Angew. Chemie - Int. Ed.

    (2012)
  • N.H. Cho et al.

    A multifunctional core-shell nanoparticle for dendritic cell-based cancer immunotherapy

    Nat. Nanotechnol.

    (2011)
  • M.Z. Iqbal et al.

    Silica-coated super-paramagnetic iron oxide nanoparticles (SPIONPs): a new type contrast agent of T1 magnetic resonance imaging (MRI)

    J. Mater. Chem. B

    (2015)
  • A.Y. Lin et al.

    High-density sub-100-nm peptide-gold nanoparticle complexes improve vaccine presentation by dendritic cells in vitro

    Nanoscale Res. Lett.

    (2013)
  • Q. Chen et al.

    Photothermal therapy with immune-adjuvant nanoparticles together with checkpoint blockade for effective cancer immunotherapy

    Nat. Commun.

    (2016)
  • K. Chamoto et al.

    Combination immunotherapy with radiation and CpG-based tumor vaccination for the eradication of radio- and immuno-resistant lung carcinoma cells

    Cancer Sci.

    (2009)
  • J. Bugno et al.

    Tumor penetration of Sub-10 nm nanoparticles: effect of dendrimer properties on their penetration in multicellular tumor spheroids, Nanomedicine Nanotechnology

    Biol. Med.

    (2019)
  • N. Hoshyar et al.

    The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction

    Nanomedicine

    (2016)
  • S.T. Reddy et al.

    In vivo targeting of dendritic cells in lymph nodes with poly(propylene sulfide) nanoparticles

    J. Control. Release

    (2006)
  • Cited by (46)

    • Targeted nanoscale drug delivery systems for melanoma therapy

      2023, Journal of Drug Delivery Science and Technology
    • Nanomedicine-based immunotherapy for Alzheimer's disease

      2023, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      Nanomedicine includes engineered materials and devices with nanoscale (1–100 nm) minimum feature sizes composed of a variety of materials, such as biodegradable polymers, lipids, proteins, viruses, and organometallic compounds (Gupta et al., 2019). The NPs are versatile formulations that can be fine tuned in their size, charge, shape, valency, biodistribution, biocompatibility, site-specific targeting, immunogenicity, controlled synthesis, loading, sustained release, precise spatiotemporal delivery, and toxicity (Gowd et al., 2022; Gupta et al., 2021, 2019). Nanomedicines can therefore be designed to interact with the BBB cells and enhance cargo transport across the barrier.

    • Prodrug nanoparticles potentiate tumor chemo-immunometabolic therapy by disturbing oxidative stress

      2022, Journal of Controlled Release
      Citation Excerpt :

      Quantitative analysis of metastatic lung nodules posts DHCRJ treatments were 14.3 and 4.8-fold lower than control and DOX@HA + JQ1 group, respectively, demonstrating the activation of the systemic antitumor immune response stimulated by DHCRJ for metastasis rejection (Fig. S12, Supporting Information). Immune exhaustion (“cold”) tumor microenvironment, that is, lack of infiltrated cytotoxic T lymphocytes (CTLs) but plenty of immunosuppressive regulatory T lymphocytes (Tregs), myeloid-derived suppressive cells (MDSCs), and tumor-associated macrophages (TAMs), which lead to limit immunotherapy effectiveness in TNBC [50–53]. To investigate the antitumor immune response activation and immune resistance reduction among immune contexts after different treatments, tumor-infiltrating immune cells and cytokine production were analyzed post three times treatments.

    View all citing articles on Scopus
    View full text