CIRSE Annual Congress

September 14-18 | Lisbon, Portugal

Patient centered - Science driven

September 14-18 | Lisbon, Portugal

Patient centered - Science driven

September 14-18 | Lisbon, Portugal

September 14-18 | Lisbon, Portugal

September 14-18 | Lisbon, Portugal

Patient centered - Science driven
Slider
ProgrammeSneak peeksUse of oncolytic viruses in IO

Use of oncolytic viruses in IO

Three reasons why you cannot miss my lecture

  1. You will learn how oncolytic viruses, injected into immune “cold” tumors, modulate the tumour microenvironment with strong local and systemic anticancer effects.
  2. I will show you how interventional radiologist play a central role in the planning and safe, accurate administration of intratumoral immunotherapy under imaging guidance.
  3. I will provide recommendations for optimizing your workflow including target lesion selection, needle type and size, injection volume, injection technique, and response assessment.

Dr. Nikolaos Fotiadis
Speaker bio 
 

Add this session to your calendar!

Background: Rationale and evidence for intratumoral immunotherapy

Systemic immunotherapy mainly in the form of immune checkpoint inhibitors has transformed the oncology treatment landscape, providing robust antitumor responses in various tumor types [1-3]. However, the vast majority of cancers do not respond to systemic immunotherapy due to a series of obstacles which include an immunosuppressive tumour microenvironment (i.e. immunologically “cold” tumors), physical barriers restricting immune cell invasion of tumors, insufficient drug penetration into tumours, and systemic toxicities [4, 5]. Direct intratumoral delivery of oncolytic immunotherapies could overcome many of these barriers by changing the tumor microenvironment and ignite a powerful patient specific immune response [6].

Oncolytic viruses (OVs) preferentially replicate in tumors as compared to normal tissue and promote immunogenic cell death and induction of host systemic anti-tumor immunity against the antigens released. Much of the existing body of contemporary clinical evidence surrounding intratumoral immunotherapy is derived from studies of talimogene laherparepvec (T-VEC), the only oncolytic immunotherapy to date approved by the US Food and Drug Administration and European Medicines Agency for intralesional treatment of melanoma. T-VEC is derived from a genetically modified herpes simplex virus type 1 engineered to expresses granulocyte-macrophage colony-stimulating factor an immune stimulatory protein, that selectively replicates in and lyses tumor cells [7]. Since its initial approval in 2015, T-VEC has been tested as monotherapy and combined with other therapies, such as immune checkpoint inhibitors, but many of these studies reported low response rates, revealing challenges associated with the clinical use of T-VEC, such as optimal dosing schedule [7]. Encouraging outcomes were reported in the phase 1b portion of the MASTERKEY-265 trial evaluating the staggered combination of intralesional T-VEC plus intravenous pembrolizumab in 21 patients with advanced melanoma (objective response rate: 62%; complete response rate: 43%) [8]. However, the phase 3 portion of the trial evaluating T-VEC plus pembrolizumab (initiated simultaneously) vs pembrolizumab alone failed to demonstrate a significant difference in the dual primary endpoints of progression-free survival and overall survival [9]. Next generation oncolytic viruses target multiple pathways with a single agent. For example, a series of herpes simplex virus type 1–based oncolytic immunotherapies (RP1–RP3) encodes sequences for multiple genes, including a fusogenic membrane glycoprotein (gibbon ape leukemia virus) that increases immunogenic cell death, granulocyte-macrophage colony-stimulating factor, an anti–cytotoxic T-lymphocyte antigen 4 antibody-like molecule, and immune costimulatory ligands (CD40L, 4-1BBL) in various combinations [10], with multiple ongoing clinical trials.

Practical considerations and optimizing workflow for intratumoral immunotherapy programs

Considerations for intratumoral immunotherapy programs include lesion selection and prioritization, delivery technique, needle design, and outcomes assessments.

The initial evaluation should include the assessment of all tumor locations. Common considerations include tumor size and accessibility for needle access. A tumor diameter below 1 cm creates additional challenges for accurate and reproducible intratumoral drug delivery. As the tumor depth affects the accuracy of needle placement , it is advisable to have a compromise between tumor size and tumor depth. In addition, tumor composition with necrosis, vascularity, and immune infiltration can modify drug exposure to viable tumor cells. Recent PET scans or DWI MRI can assist in identifying active tumor and can even be used to target a particularly active component of the tumor [11].

Regarding optimal needle design, a 21–23-gauge needle is recommended for deep intratumoral injections, whereas needles as small as 30-gauge may be suitable for skin lesions [12]. End-hole needles are generally appropriate for small (<1 cm) lesions, but are commonly used for lesions of all sizes. For larger deep-seated lesions, multiside-hole or multipronged needles may offer an advantage over an end-hole needle to distribute the injectate more uniformly [12-14]. Luer lock syringes prevent leakage and inadvertent displacement of the needle during injections. Biosafety concerns may be associated with the use of oncolytic immunotherapies. In clinical trials, few incidents of occupational exposure to herpes simplex virus type 1 through needlestick or splash were reported and those that occurred were effectively treated with acyclovir when appropriate [15,16]. Further, there have been no reported cases of viral transmission to close contacts. For patients who are immunosuppressed, additional safety warnings and procedures may be necessary [16].

Intratumoral injection of oncolytic viruses for solid tumours could help circumvent many of the barriers associated with systemic administration while still inducing a robust systemic antitumor response beyond the injection site. Interventional radiologist in close collaboration with medical and surgical oncologists, play a central role in optimizing workflows and logistics for intratumoral administration of immunotherapeutics and ensuring optimal treatment outcomes for patients.

 

Nikolaos Fotiadis

Royal Marsden Hospital, London/GB

Dr. Fotiadis has been a Consultant and Clinical Lead in Interventional Radiology at the Royal Marsden Hospital in London since 2012 and a Reader/ Associate Professor in Interventional Oncology at the Institute of Cancer Research, London UK. Before that, he had clinical and academic affiliations in interventional radiology as Consultant & Honorary Senior Lecturer at Barts and QMUL from 2007-2012 and as a Clinical Lecturer at King’s College London as well as Guy’s and St Thomas’ Hospital 2005-2007. Dr. Fotiadis leads the largest interventional oncology service in the UK which offers the whole range of oncological interventions. Together with his team, he has developed the first robotic guided interventional oncology service in the UK and has performed more than 400 robotic interventions (ablations and biopsies). Dr. Fotiadis’ research interests include functional imaging guided precision biopsies, robotic interventions(ablation and biopsies) and image-guided delivery of intra-tumoral immunotherapies.

 

References

  1. Zhang Y, Zhang Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor infiltrating immune cells and their therapeutic implications. Cell Mol Immunol 2020; 17 (8) : 807-821.
  2. Larkin J, Chiarion-Sileni V, Gonzalez R et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015 Jul 2;373(1):23-34.
  3. Forde PM, Chaft JE, Pardoll DM. Neodjuvant PD-1 blockade in resectable lung cancer. N Eng J Med 2018; 379(22): 2108-2121.
  4. Haslam A, Prasad V, Estimation of the Percentage of US Patients With Cancer Who Are Eligible for and Respond to Checkpoint Inhibitor Immunotherapy Drugs. JAMA Netw Open 2019 May 3; 2(5): e 192535.
  5. Abdelhafeez AAM, Sgohdy KS, Ibrahim W. Safety of combination immune checkpoint inhibitors compared to monotherapy; a systematic review and meta-analysis. Cancer Invest 2020; 38 (3): 150-157.
  6. Ghosn N, Tselikas L, Champiat S et al. Intratumoral immunotherapy: is it ready for prime time? Curr Oncol Rep 2023 Aug ; 25(8): 857-867.
  7. Shalhout SZ, Miller DM, Emerick KS, Kaufman HL. Therapy with oncolytic viruses: progress and challenges. Nat Rev Clin Oncol 2023; Mar 20(3): 160-177.
  8. Long G, Dummer R, Johnson D, Michielin O, Martin-Algarra S, Treichel S, Chan E, Diede S, Ribas A. 429 Long-term analysis of MASTERKEY-265 phase 1b trial of talimogene laherparepvec (T-VEC) plus pembrolizumab in patients with unresectable stage IIIB-IVM1c melanoma. J Immunotherap Cancer 2020; 8(Suppl 3). A261-A261.
  9. Chesney JA, Ribas A, Long GV et al. Randomized, double-blind, placebo controlled, global phase III trial of Talimogene laherparepvec combined with pembrolizumab for advanced melanoma. J Clin Oncol 2023 Jan 20; 41(3): 528-540.
  10. Thomas S, Kuncheria L, Roulstone V et al. Development of a new fusion-enhanced oncolytic immunotherapy platform based on herpes simplex virus type 1. J Immunother Cancer 2019;7(1): 214.
  11. Tselikas L, Champiat S, Sheth RA et al. Interventional Radiology for Local Immunotherapy in Oncology. Clin Cancer Res 2021 May 15; 27(10): 2698-2705.
  12. Marabelle A, Andtbacka R, Harrington K et al. Starting the fight in the tumor: expert recommendations for the development of human intratumoral immunotherapy (HIT-IT). Ann Oncol 2018;29(11):2163-2174.
  13. 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(7):e207911.
  14. Tselikas L, Dardenne A, de Baere T, et al. Feasibility, safety and efficacy of human intra-tumoral immuno-therapy. Gustave Roussy’s initial experience with its first 100 patients. Eur J Cancer 2022;172:1-12.
  15. Robilotti EV, Kumar A, Glickman MS, Kamboj M. Viral oncolytic immunotherapy in the war on cancer: infection control considerations. Infect Control Hosp Epidemiol 2019;40(3):350-354.
  16. Macedo N, Miller DM, Haq R, Kaufman HL. Clinical landscape of oncolytic virus research in 2020. J Immunother Cancer 2020;8(2)