Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Review
. 2021 Sep;18(9):558-576.
doi: 10.1038/s41571-021-00507-y. Epub 2021 May 18.

Intratumoural administration and tumour tissue targeting of cancer immunotherapies

Affiliations
Review

Intratumoural administration and tumour tissue targeting of cancer immunotherapies

Ignacio Melero et al. Nat Rev Clin Oncol. 2021 Sep.

Abstract

Immune-checkpoint inhibitors and chimeric antigen receptor (CAR) T cells are revolutionizing oncology and haematology practice. With these and other immunotherapies, however, systemic biodistribution raises safety issues, potentially requiring the use of suboptimal doses or even precluding their clinical development. Delivering or attracting immune cells or immunomodulatory factors directly to the tumour and/or draining lymph nodes might overcome these problems. Hence, intratumoural delivery and tumour tissue-targeted compounds are attractive options to increase the in situ bioavailability and, thus, the efficacy of immunotherapies. In mouse models, intratumoural administration of immunostimulatory monoclonal antibodies, pattern recognition receptor agonists, genetically engineered viruses, bacteria, cytokines or immune cells can exert powerful effects not only against the injected tumours but also often against uninjected lesions (abscopal or anenestic effects). Alternatively, or additionally, biotechnology strategies are being used to achieve higher functional concentrations of immune mediators in tumour tissues, either by targeting locally overexpressed moieties or engineering 'unmaskable' agents to be activated by elements enriched within tumour tissues. Clinical trials evaluating these strategies are ongoing, but their development faces issues relating to the administration methodology, pharmacokinetic parameters, pharmacodynamic end points, and immunobiological and clinical response assessments. Herein, we discuss these approaches in the context of their historical development and describe the current landscape of intratumoural or tumour tissue-targeted immunotherapies.

PubMed Disclaimer

Conflict of interest statement

I.M. has received research grants from Alligator, Bioncotech, Bristol Myers Squibb (BMS), Leadartis, Pfizer and Roche; has received speaker’s bureau honoraria from MSD; and is a consultant or advisory board member for Alligator, AstraZeneca, Bayer, Bioncotech, BMS, F-Star, Genmab, Gossamer, Merck Serono, Numab, Pieris and Roche. E.C. is a consultant or advisory board member for AstraZeneca, Beigene, BMS, MSD and Roche. S.C. has received honoraria from Amgen, AstraZeneca, BMS, Janssen, Merck, MSD, Novartis and Roche; is an advisory board member for Amgen and AstraZeneca; has received funding for travel and conference attendance from AstraZeneca, MSD and Roche; and has received research grants from AstraZeneca, BMS, Boehringer Ingelheim, Janssen-Cilag, Merck, Novartis, Onxeo, Pfizer, Roche and Sanofi, and non-financial research support (investigational drugs) from AstraZeneca, Bayer, BMS, Boehringer Ingelheim, Medimmune, Merck, NH TherAGuiX, Onxeo, Pfizer and Roche. S.C. has been a principal investigator of academic or industry-sponsored clinical trials of intratumoural immunotherapies for Abbvie, AstraZeneca/Medimmune, BMS, Eisai/H3 Biomedicine, IDERA, Lytix Biopharma, MSD, Nanobiotix and Sanofi/BioNTech. A.M. has been a principal investigator of academic or industry-sponsored clinical trials of intratumoural immunotherapies from AstraZeneca, BMS, Eisai, IDERA, Lytix Biopharma, Merck/MSD, Roche and Transgene; is a member of the Data Safety and Monitoring Board of a trial of a intratumoural TLR3 agonist sponsored by Oncovir (NCT02423863); and has participated in scientific advisory boards or has provided consultancy services on the topic of intratumoural immunotherapies for Amgen, AstraZeneca, Banque Pour l’Investissement, Bayer, Eisai, eTheRNA, Lytix Biopharma, Medincell, MSD, Novartis, Oncosec, Pillar Partners, Rigontec and Sanofi/BioNTech. M.A. declares no competing interests.

Figures

Fig. 1
Fig. 1. Strengths, weaknesses, opportunities and threats of intratumoural delivery of immunotherapies.
a | Comparison of immunotherapy delivery strategies graphically depicting the typical biodistribution of intravenously administered systemic and tumour tissue-targeted immunotherapies and intratumourally administered immunotherapies. Intravenous delivery has certain practical advantages but also carries a higher risk of adverse events, particularly on-target, off-tumour toxicities related to systemic exposure to the active compound. On the contrary, intratumoural delivery presents technical and logistical challenges but can increase the therapeutic index of immunotherapies within the treated lesions, typically with a low risk of on-target, off-tumour toxicities. b | Summary of the internal strengths and weaknesses as well as external opportunities and threats (SWOT analysis) of intratumoural immunotherapy, all of which need to be balanced against the current clinical drug development landscape of cancer immunotherapy, which encompasses a multitude of novel agents. irAEs, immune-related adverse events; itRECIST, Response Criteria for Intratumoral Immunotherapy in Solid Tumors.
Fig. 2
Fig. 2. Current landscape of active clinical trials of intratumoural immunotherapies.
a | Classification of the different types of immunotherapy agents that are currently being investigated in clinical trials involving intratumoural administration as of 1 December 2020 (data obtained from the ClinicalTrials.gov database using the search term “intratumoral OR intralesional AND cancer AND immunotherapy”). b | Visualization of the number of clinical trials for each type of agent outlined in the classification by circle packing, whereby the circle diameter indicates the relative proportion of ongoing or completed clinical trials identified. The trials included in this figure are listed in Supplementary Tables 1–7. CAR, chimeric antigen receptor; DCs, dendritic cells; ICD, immunogenic cell death; LL37, 37-residue cathelicidin antimicrobial peptide; mAb, monoclonal antibody; NK, natural killer; PRR, pattern recognition receptor; RLR, RIG-I-like receptor; STING, stimulator of interferon genes; TAA, tumour-associated antigen; TCR, T cell receptor; TILs, tumour-infiltrating lymphocytes; TLR, Toll-like receptor.
Fig. 3
Fig. 3. Boosting the intratumoural cancer immunity cycle.
By selecting therapeutic agents based on their immunological properties, local immunotherapy — achieved either directly through intratumoural administration or indirectly through selective delivery to or activation in the tumour following systemic administration — can specifically enhance each step of the cancer immunity cycle described by Chen and Mellman. Examples of key cell types, processes and immunotherapy agents that are relevant to each step of this cycle are noted in the figure. Importantly, in accordance with steps 4 and 5 of the cycle, local immunotherapies need to result in redistribution of effector immune cells or antibodies via the circulation for abscopal or anenestic responses against distant untreated lesions and micrometastases. ADCs, antibody–drug conjugates; APCs, antigen-presenting cells; CAR, chimeric antigen receptor; CLEVER1, common lymphatic endothelial and vascular endothelial receptor 1 (also known as stabilin 1); PRR, pattern recognition receptor; Siglec-15, sialic acid-binding Ig-like lectin 15; SIRPα, signal-regulatory protein-α; TCR, T cell receptor, TILs, tumour-infiltrating lymphocytes; Treg, regulatory T.
Fig. 4
Fig. 4. Targeted approaches to immunotherapy against solid tumours.
Multiple strategies are currently being developed to promote the selective homing, accumulation and/or activation of immunotherapeutic agents inside tumours following systemic administration. These novel drugs, which include bispecific biologics, antibody Fab fragments, full antibodies, DARPins, immunocytokines and probodies, have diverse structures and target various factors present on tumour cells, in the tumour stroma and/or on immune cells to facilitate selective activation of immune responses in malignant tissues. Examples of targets for tumour cell valency or immune cell valency are listed in the figure. Such targeting strategies could also be of great interest for local immunotherapy owing to the potential to improve tumour exposure through tissue-tethering and retention of the agent in the tumour microenvironment following intratumoural administration. CEA, carcinoembryonic antigen; DC, dendritic cell; FAP, fibroblast-activation protein; NK, natural killer; Treg, regulatory T.

References

    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–1355. doi: 10.1126/science.aar4060. - DOI - PMC - PubMed
    1. Guedan S, Ruella M, June CH. Emerging cellular therapies for cancer. Annu. Rev. Immunol. 2019;37:145–171. doi: 10.1146/annurev-immunol-042718-041407. - DOI - PMC - PubMed
    1. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–68. doi: 10.1126/science.aaa4967. - DOI - PMC - PubMed
    1. Cuesta AM, Sainz-Pastor N, Bonet J, Oliva B, Alvarez-Vallina L. Multivalent antibodies: when design surpasses evolution. Trends Biotechnol. 2010;28:355–362. doi: 10.1016/j.tibtech.2010.03.007. - DOI - PubMed
    1. Melero I, Rouzaut A, Motz GT, Coukos G. T-cell and NK-cell infiltration into solid tumors: a key limiting factor for efficacious cancer immunotherapy. Cancer Discov. 2014;4:522–526. doi: 10.1158/2159-8290.CD-13-0985. - DOI - PMC - PubMed

Publication types

MeSH terms