Oops, you're using an old version of your browser so some of the features on this page may not be displaying properly.

MINIMAL Requirements: Google Chrome 24+Mozilla Firefox 20+Internet Explorer 11Opera 15–18Apple Safari 7SeaMonkey 2.15-2.23

Mini oral session - Investigational immunotherapy

962MO - A phase I clinical trial on intratumoral injection of autologous CD1c (BDCA-1)+/CD141 (BDCA-3)+ myeloid dendritic cells (myDC) in combination with talimogene laherparepvec (T-VEC) in patients with advanced pretreated melanoma

Date

17 Sep 2021

Session

Mini oral session - Investigational immunotherapy

Topics

Immunotherapy

Tumour Site

Melanoma

Presenters

Julia Katharina Schwarze

Citation

Annals of Oncology (2021) 32 (suppl_5): S829-S866. 10.1016/annonc/annonc705

Authors

J.K. Schwarze1, J. Tijtgat1, G. Awada1, L. Cras2, I. Dufait3, R. Forsyth2, I. Van Riet4, S. Tuyaerts5, B. Neyns1

Author affiliations

  • 1 Department Of Medical Oncology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), 1090 - Brussels/BE
  • 2 Department Of Pathology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), 1090 - Brussels/BE
  • 3 Department Of Radiotherapy/translational Radiation Oncology, Supportive Care And Physics (trop), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), 1090 - Brussels/BE
  • 4 Stem Cell Laboratory/department Of Clinical Hematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), 1090 - Brussels/BE
  • 5 Department Of Medical Oncology/laboratory Of Medical And Molecular Oncology (lmmo), Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), 1090 - Brussels/BE

Resources

Login to get immediate access to this content.

If you do not have an ESMO account, please create one for free.

Abstract 962MO

Background

Intratumoral (IT) myDC play a pivotal role in initiating antitumor immune responses within the tumor microenvironment. IT injection of the oncolytic virus T-VEC may lead to the release of tumor antigens and maturation signals that can be captured and processed by CD1c (BDCA-1)+/CD141 (BDCA-3)+ myDC, thereby reinvigorating the cancer immunity cycle.

Methods

Patients (pts) with ICI-refractory melanoma received IT injections of ≥1 non-visceral metastases with T-VEC (106 PFU/mL; max 4 mL) on day 1 followed by IT injection of CD1c (BDCA-1)+ (cohort C1) or CD1c (BDCA-1)+/CD141 (BDCA-3)+ myDC (cohort C2) on day 2. Injection of T-VEC (108PFU/mL; max 4 mL) was repeated on day 21, and Q2w thereafter. In C1, the number of CD1c (BDCA-1)+myDCs was escalated from 0.5x106, to 1x106, and 10x106 cells. In C2, pts received all isolated CD1c (BDCA-1)+/CD141 (BDCA-3)+ myDCs. Primary objectives were safety and feasibility. Immunohistochemistry (IHC), gene expression profiling (GEP), and multiplexed immunofluorescence (mIF) of baseline and on-treatment biopsies was performed.

Results

13 pts were enrolled (C1: n=7 [respectively 2, 2, and 3 pts per dose-level of myDC]; C2: n=6). Pts received the predefined dose of myDCs and a median of 6 (range 3-8) T-VEC injections. Most frequent AEs were fatigue in 11 pts (85%), injection-site pain in 9 pts (69%), fever in 8 pts (62%), and chills and flu-like symptoms in 6 pts (46%). There were no G4 or G5 AEs. AEs of special interest were a G3 eosinophilia and a G2 purpuric rash at the injection-site; 2 pts (C1, dose level 3) developed a pathological complete remission that is ongoing at 24 months following treatment initiation. One pt in C2 had an unconfirmed partial response (iRECIST); a mixed response was observed in 2 pts. Responses were observed in both injected and non-injected lesions. In responder pts, infiltration of lymphocytes was observed on IHC. GEP and mIF on biopsies are ongoing.

Conclusions

IT co-injection of CD1c (BDCA-1)+ +/- CD141 (BDCA-3)+ myDC plus T-VEC is feasible, tolerable, and resulted in encouraging early signs of durable antitumor activity in pts with ICI-refractory melanoma.

Clinical trial identification

NCT03747744.

Editorial acknowledgement

Legal entity responsible for the study

Department of Medical Oncology, Universitair Ziekenhuis Brussel.

Funding

Kom op Tegen Kanker (Stand up to Cancer), the Flemish cancer society.

Disclosure

J.K. Schwarze: Non-Financial Interests, Personal, Other: Amgen; Financial Interests, Personal, Invited Speaker: Novartis; Non-Financial Interests, Personal, Other: MSD Oncology. G. Awada: Financial Interests, Personal, Invited Speaker: Novartis; Financial Interests, Personal, Advisory Board: Novartis; Non-Financial Interests, Personal, Other: MSD Oncology; Non-Financial Interests, Personal, Other: Astellas Pharma; Non-Financial Interests, Personal, Other: Novartis; Non-Financial Interests, Personal, Other: Pfizer. B. Neyns: Financial Interests, Personal, Invited Speaker: Roche; Financial Interests, Personal, Invited Speaker: Bristol-Myers Sqibb; Financial Interests, Personal, Invited Speaker: Merck Sharp & Dohme; Financial Interests, Personal, Invited Speaker: Novartis; Financial Interests, Personal, Invited Speaker: AstraZeneca; Financial Interests, Institutional, Funding: Pfizer; Financial Interests, Institutional, Funding: Novartis; Financial Interests, Institutional, Funding: Roche; Financial Interests, Institutional, Funding: Merck Sharp & Dohme. All other authors have declared no conflicts of interest.

This site uses cookies. Some of these cookies are essential, while others help us improve your experience by providing insights into how the site is being used.

For more detailed information on the cookies we use, please check our Privacy Policy.

Customise settings
  • Necessary cookies enable core functionality. The website cannot function properly without these cookies, and you can only disable them by changing your browser preferences.