While major strides have been made in the development of therapies for melanoma, cases continue to rise, with more diagnoses occurring at an early stage where surgery is indicated for patients. However, even with standard of care anti-PD-1 adjuvant immunotherapy, patients still have a high risk of relapse (3-year relapse-free survival is 50-60%). Neoadjuvant anti-PD-1/anti-CTLA-4 immunotherapy has emerged as a potentially more efficacious alternative to adjuvant immunotherapy, however, widespread usage in these non-metastatic patients would be precluded due to significant toxicities. Therefore, there is a great unmet need to improve outcomes for locally/regionally advanced surgically resectable melanoma patients. Cytokine therapies such as interleukin (IL)- 12 and IL-15 have shown promise in pre-clinical in vitro and in vivo studies, however, when delivered systemically in bolus, their clinical utility is limited due to serious adverse effects as well as suboptimal pharmacokinetics and pharmacodynamics. Thus, we need to find novel ways to locally modulate the immune system early to limit toxicity and reduce the risk of recurrence. To address this challenge in melanoma in this NIH Phase I SBIR, Strand Therapeutics is proposing to engineer a tunable and programmable small molecule-regulated self-replicating mRNA (repRNA)-based combinatorial cytokine immunotherapy that mimics the physiological expression kinetics of IL-12 (expressed early) and IL-15 (expressed later). In doing so, this programmable repRNA immunotherapy is designed to enhance the patients own immune system to combat melanoma tumors and provide durable immune surveillance and remission with limited toxicity. In Aim 1, we will construct the mRNA circuit and validate expression kinetics of our programmed mRNAs using surrogate luciferase reporters, which will allow us to assess in vivo expression kinetics in real-time in a mouse model of melanoma. In Aim 2, we will encode IL-12 and IL-15 in the circuit validated in Aim 1 and assess expression kinetics and tunability of IL-12/IL-15 expression in a mouse model of melanoma. In Aim 3, we will test if our engineered circuit can eliminate tumors in vivo in mouse models of melanoma, and benchmark against non- circuit IL-12/IL-15 delivery approaches such as recombinant cytokines and constitutive expression from mRNAs. Successful completion of these studies will lead to a novel programmable circuit with IL-12/IL-15 for the treatment of melanoma. Through this project, we intend to program the natural kinetics of IL-12/IL-15, which will induce stronger and longer-lasting anti-cancer immune responses and increase the efficacy of anti-PD-1/PD-L1 therapies, without the side-effects linked to systemically delivered cytokines. Public Health Relevance Statement PROJECT NARRATIVE While there have been major strides in the development of therapies for melanoma, the number of cases is increasing, with more diagnoses occurring at an early stage where surgery is indicated; however, locally advanced patients are still at a high risk of relapse following surgery. To address this unmet need, Strand Therapeutics is engineering a programmable mRNA circuit with temporal expression of cytokines, interleukin-12 (IL-12) and interleukin-15 (IL-15), for effective stimulation of the adaptive immune response as a neoadjuvant therapy for melanoma. Our proprietary lipid nanoparticles will be used to deliver self-replicating mRNA, which will temporally express IL-12 and IL-15 directly to the tumor microenvironment (TME), to increase TME immunogenicity and increase the efficacy of anti-PD-1 therapy, without the side effects linked to systemically delivered cytokines.
Project Terms: Animals ; Malignant Neoplasms ; Cancers ; Malignant Tumor ; malignancy ; neoplasm/cancer ; Cells ; Cell Body ; Diagnosis ; Doxycycline ; Vibramycin ; alpha-6-Deoxyoxytetracycline ; Engineering ; Feedback ; Gene Expression ; Half-Life ; Immune system ; allergic/immunologic body system ; allergic/immunologic organ system ; Immunity ; Immunologic Surveillance ; Immune Surveillance ; Immunologic Surveillances ; Immunological Surveillance ; Immunological Surveillances ; Immunosurveillance ; Immunotherapy ; Immune mediated therapy ; Immunologically Directed Therapy ; immune therapeutic approach ; immune therapeutic interventions ; immune therapeutic regimens ; immune therapeutic strategy ; immune therapy ; immune-based therapies ; immune-based treatments ; immuno therapy ; In Vitro ; Incidence ; Kinetics ; Luciferases ; Luciferase Immunologic ; melanoma ; Malignant Melanoma ; Mus ; Mice ; Mice Mammals ; Murine ; United States National Institutes of Health ; NIH ; National Institutes of Health ; Neoplasm Metastasis ; Metastasis ; Metastasize ; Metastatic Lesion ; Metastatic Mass ; Metastatic Neoplasm ; Metastatic Tumor ; Secondary Neoplasm ; Secondary Tumor ; cancer metastasis ; tumor cell metastasis ; Patients ; Program Development ; Recurrence ; Recurrent ; Relapse ; Risk ; Messenger RNA ; mRNA ; T-Lymphocyte ; T-Cells ; thymus derived lymphocyte ; Testing ; Time ; Trimethoprim ; Proloprim ; Trimpex ; cytokine ; Interleukin-12 ; Edodekin Alfa ; IL-12 ; IL12 ; NKSF ; Natural Killer Cell Stimulatory Factor ; lumican ; Metastatic Neoplasm to the Lung ; Metastasis to the Lung ; Metastatic Tumor to the Lung ; lung metastasis ; metastasize to the lung ; pulmonary metastasis ; Fireflies ; Lampyridae ; base ; cytokine therapy ; Distal ; Clinical ; Phase ; Biological ; Physiological ; Physiologic ; Link ; Lesion ; Measurement ; Interleukin-15 ; IL-15 ; IL15 ; IL15 Protein ; Interleukin-15 Precursor ; MGC9721 ; Immunological response ; host response ; immune system response ; immunoresponse ; Immune response ; Therapeutic ; Reporter ; Intravenous ; programs ; subdermal ; subcutaneous ; Best Practice Analysis ; Benchmarking ; Operative Procedures ; Surgical ; Surgical Interventions ; Surgical Procedure ; surgery ; Operative Surgical Procedures ; Remission ; Disease remission ; American ; success ; tumor growth ; Induction Therapy ; NEOADJ ; Neoadjuvant ; Neoadjuvant Treatment ; Neoadjuvant Therapy ; Toxicities ; Toxic effect ; Adjuvant Therapy ; Immunomodulation ; immune modulation ; immune regulation ; immunologic reactivity control ; immunomodulatory ; immunoregulatory ; immunoregulation ; novel ; memory T lymphocyte ; T memory cell ; Pharmacodynamics ; Modeling ; Adverse Experience ; Adverse event ; Adverse effects ; Sea Pansy ; Renilla ; small molecule ; Address ; Dose ; Tumor Load ; Tumor Burden ; Bolus ; Bolus Infusion ; Data ; Resectable ; in vivo ; mRNA Expression ; Nonmetastatic ; Non-metastatic ; Small Business Innovation Research Grant ; SBIR ; Small Business Innovation Research ; Adjuvant ; Recombinant Cytokines ; Pathway interactions ; pathway ; pre-clinical ; preclinical ; preclinical study ; pre-clinical study ; tumor microenvironment ; cancer microenvironment ; immunogenicity ; design ; designing ; novel strategies ; new approaches ; novel approaches ; novel strategy ; manufacturing process ; Population ; innovation ; innovate ; innovative ; synthetic biology ; combinatorial ; mouse model ; murine model ; therapy development ; develop therapy ; intervention development ; treatment development ; prototype ; tumor ; high risk ; combat ; FDA approved ; standard of care ; Regimen ; T cell response ; Patient risk ; improved outcome ; objective response rate ; relapse risk ; adaptive immune response ; anti-PD1 therapy ; PD-1 antibody therapy ; PD-1 therapy ; PD1 antibody therapy ; PD1 based treatment ; aPD-1 therapy ; aPD-1 treatment ; aPD1 therapy ; aPD1 treatment ; anti-PD-1 therapy ; anti-PD-1 treatment ; anti-PD1 treatment ; anti-programmed cell death 1 therapy ; anti-programmed cell death protein 1 therapy ; programmed cell death protein 1 therapy ; PD-1 blockade ; PD1 blockade ; anti-PD-1 blockade ; anti-PD1 blockade ; anti-tumor immune response ; antitumor immune response ; Injections ; Abscopal effect ; abscopal activity ; abscopal response ; anti-PD-1 ; aPD-1 ; aPD1 ; anti programmed cell death 1 ; anti-PD1 ; anti-programmed cell death protein 1 ; antiPD-1 ; antiPD1 ; αPD-1 ; αPD1 ; anti-cancer ; anticancer ; nanoparticle delivery ; nano particle delivery ; nanoparticle delivered ; lipid nanoparticle ; side effect ; anti-CTLA4 ; aCTLA-4 ; aCTLA4 ; anti-CTLA-4 ; α-CTLA-4 ; α-CTLA4 ; αCTLA-4 ; αCTLA4 ; pharmacokinetics and pharmacodynamics ; PK/PD ; anti-PD-1/PD-L1 ; anti-PD-L1 therapy ; PD-L1 therapy ; PD-L1 treatment ; PDL1 therapy ; PDL1 treatment ; aPD-L1 therapy ; aPD-L1 treatment ; anti programmed cell death ligand 1 therapy ; anti programmed cell death ligand 1 treatment ; anti programmed cell death protein ligand 1 therapy ; anti programmed cell death protein ligand 1 treatment ; anti-PD-L1 treatment ; anti-PDL1 therapy ; anti-PDL1 treatment ; αPD-L1 therapy ; αPD-L1 treatment ;