Digestive Disease Interventions 2021; 05(01): 050-054
DOI: 10.1055/s-0040-1722267
Review Article

Intratumoral and Oncoviral Immunotherapy

Rahul A. Sheth
1   Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
› Institutsangaben

Abstract

Immunotherapy is a paradigm-shifting advance in cancer care, but despite their success, contemporary immunotherapy strategies are limited by several factors. The objective response rates to checkpoint inhibitor therapy remain below 50%, and the toxicities associated with systemically administered immunotherapies, particularly combination immunotherapy regimens, can be devastating. Intratumoral immunotherapies represent a burgeoning paradigm in immuno-oncology designed to address these limitations. The direct delivery of intratumoral immunotherapies can result in concentrations of the therapeutic agent at levels that would likely be far too toxic if administered systemically. In this review, I summarize the sound biological framework underlying intratumoral immunotherapies as well as provide an overview of the major categories of intratumoral agents currently under investigation. I also emphasize the components of this treatment strategy that are relevant for image-guided interventionalists based on existing clinical experience.



Publikationsverlauf

Eingereicht: 01. Juli 2020

Angenommen: 25. November 2020

Artikel online veröffentlicht:
19. Januar 2021

© 2021. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015; 161 (02) 205-214
  • 2 Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015; 348 (6230): 56-61
  • 3 Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017; 377 (25) 2500-2501
  • 4 Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med 2018; 378 (02) 158-168
  • 5 Marabelle A, Tselikas L, de Baere T, Houot R. Intratumoral immunotherapy: using the tumor as the remedy. Ann Oncol 2017; 28 (Suppl. 12) xii33-xii43
  • 6 Galluzzi L, Vitale I, Warren S. et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer 2020; 8 (01) e000337
  • 7 Andtbacka RHI, Kaufman HL, Collichio F. et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol 2015; 33 (25) 2780-2788
  • 8 Andtbacka RHI, Ross M, Puzanov I. et al. Patterns of clinical response with Talimogene laherparepvec (T-VEC) in patients with melanoma treated in the OPTiM phase III clinical trial. Ann Surg Oncol 2016; 23 (13) 4169-4177
  • 9 Louie RJ, Perez MC, Jajja MR. et al. Real-world outcomes of Talimogene laherparepvec therapy: a multi-institutional experience. J Am Coll Surg 2019; 228 (04) 644-649
  • 10 Ribas A, Dummer R, Puzanov I. et al. Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy. Cell 2017; 170 (06) 1109-1119.e10
  • 11 Hecht JR, Pless M, Cubillo A. et al. Early safety from a phase I, multicenter, open-label clinical trial of Talimogene laherparepvec (T-VEC) injected (inj) into liver tumors in combination with pembrolizumab (pem). J Clin Oncol 2020; 38: 3015
  • 12 Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov 2015; 14 (09) 642-662
  • 13 Twumasi-Boateng K, Pettigrew JL, Kwok YYE, Bell JC, Nelson BH. Oncolytic viruses as engineering platforms for combination immunotherapy. Nat Rev Cancer 2018; 18 (07) 419-432
  • 14 Russell SJ, Barber GN. Oncolytic viruses as antigen-agnostic cancer vaccines. Cancer Cell 2018; 33 (04) 599-605
  • 15 Aznar MA, Tinari N, Rullán AJ, Sánchez-Paulete AR, Rodriguez-Ruiz ME, Melero I. Intratumoral delivery of immunotherapy-act locally, think globally. J Immunol 2017; 198 (01) 31-39
  • 16 Sheth RA, Hesketh R, Kong DS, Wicky S, Oklu R. Barriers to drug delivery in interventional oncology. J Vasc Interv Radiol 2013; 24 (08) 1201-1207
  • 17 Bhatia S, Miller NJ, Lu H. et al. Intratumoral G100, a TLR4 agonist, induces antitumor immune responses and tumor regression in patients with merkel cell carcinoma. Clin Cancer Res 2019; 25 (04) 1185-1195
  • 18 Brody JD, Ai WZ, Czerwinski DK. et al. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J Clin Oncol 2010; 28 (28) 4324-4332
  • 19 Frank MJ, Reagan PM, Bartlett NL. et al. In situ vaccination with a TLR9 agonist and local low-dose radiation induces systemic responses in untreated indolent lymphoma. Cancer Discov 2018; 8 (10) 1258-1269
  • 20 Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol 2015; 15 (12) 760-770
  • 21 Meric-Bernstam F, Sandhu SK, Hamid O. et al. Phase Ib study of MIW815 (ADU-S100) in combination with spartalizumab (PDR001) in patients (pts) with advanced/metastatic solid tumors or lymphomas. J Clin Oncol 2019; 37: 2507
  • 22 Sheth RA, Murthy R, Hong DS. et al. Assessment of image-guided intratumoral delivery of immunotherapeutics in patients with cancer. JAMA Netw Open 2020; 3 (07) e207911