Peri-tumor administration of controlled release anti-CTLA-4 synergizes with systemic anti-PD-1 to induce systemic antitumor immunity while sparing autoimmune toxicity

Cancer Immunol Immunother. 2020 Sep;69(9):1737-1749. doi: 10.1007/s00262-020-02579-8. Epub 2020 Apr 24.

Abstract

Combination immunotherapy targeting the PD-1 and CTLA-4 checkpoint inhibitor pathways provides substantial clinical benefit in patients with advanced-stage cancer but at the risk of dose-limiting inflammatory and autoimmune toxicity. The delicate balance that exists between unleashing tumor killing and promoting systemic autoimmune toxicity represents a major clinical challenge. We hypothesized that targeting anti-CTLA-4 so that it perfuses tumor-draining lymph nodes would provide a significant therapeutic advantage and developed an injectable hydrogel with controlled antibody release characteristics for this purpose. Injection of hydrogel-encapsulated anti-CTLA-4 at a peri-tumor location (MC-38 tumor model) produced dose-dependent antitumor responses and survival that exceeded those by anti-CTLA-4 alone (p < 0.05). Responses to 100 µg of targeted anti-CTLA-4 also equaled or exceeded those observed with a series of systemic injections delivering 600 µg (p < 0.05). While preserving antitumor activity, this approach resulted in serum anti-CTLA-4 exposure (area under the curve) that averaged only 1/16th of that measured with systemic therapy. Consistent with the marked differences in systemic exposure, systemic anti-CTLA-4 stimulated the onset of autoimmune thyroiditis in iodide-exposed NOD.H-2h4 mice, as measured by anti-thyroglobulin antibody titer, while hydrogel-encapsulated anti-CTLA-4 had a minimal effect (p ≤ 0.01). At the same time, this targeted low-dose anti-CTLA-4 approach synergized well with systemic anti-PD-1 to control tumor growth and resulted in a high frequency of complete responders that were immune to tumor re-challenge at a distant site. We conclude that targeted and controlled delivery of low-dose anti-CTLA-4 has the potential to improve the benefit-risk ratio associated with combination checkpoint inhibitor therapy.

Keywords: Autoimmune toxicity; CTLA-4; Checkpoint inhibitor blockade; Hydrogel; NOD.H-2h4; Peri-tumor injection.

MeSH terms

  • Animals
  • Antineoplastic Agents / pharmacology*
  • Autoimmunity / drug effects
  • CTLA-4 Antigen / immunology*
  • Cell Line, Tumor
  • Cell Proliferation / drug effects
  • Combined Modality Therapy / methods
  • Delayed-Action Preparations / pharmacology*
  • Drug Synergism
  • Female
  • Immunity / drug effects*
  • Immunotherapy / methods
  • Mice
  • Mice, Inbred C57BL
  • Mice, Inbred NOD
  • Programmed Cell Death 1 Receptor / antagonists & inhibitors*

Substances

  • Antineoplastic Agents
  • CTLA-4 Antigen
  • Delayed-Action Preparations
  • Pdcd1 protein, mouse
  • Programmed Cell Death 1 Receptor