Team Award Program

2023 Team Awardees

Short Non-Coding RNAs as Biomarkers for Response to Immunotherapy in Metastatic Uveal Melanoma

Yuri Sykulev

Principal Investigator

Isidore Rigoutsos, PhD, 2023 Ocular Melanoma Foundation Team Awardee
Richard W. Hevner Professor in Computational Medicine
Director, Computational Medicine Center, Thomas Jefferson University
Professor, Department of Pathology and Genomic Medicine, Department of Biochemistry & Molecular
Biology, and Department of Cancer Biology

Ocular Melanoma Foundation logo This award was funded through a generous matching gift from the Ocular Melanoma Foundation and our patient families.

Melanoma of the eye (“uveal melanoma” – UM) is a rare tumor that originates from pigment-producing cells (melanocytes) in the eye. While UM is the most common primary eye malignancy in adults, it is rare compared to skin melanoma. Nearly one half of UM patients develop metastases that most often localize to the liver. Rapid tumor progression in the liver leads to death in 4 to 15 months. Metastatic UM (MUM) is highly resistant to chemotherapy. An alternative form of therapy called immunotherapy has also been explored in MUM. Immunotherapy activates the patient’s immune system to attack cancer cells in a targeted manner. While this approach has been very powerful in numerous contexts, and several drugs have already been approved by the US Food and Drug Administration, it has shown limited benefit for UM patients.

A new FDA-approved therapy for UM, Tebentafusp-tebn, is shown more promise. The therapy works by increasing the numbers of cancer-fighting cells and is currently the preferred treatment for 50% of Caucasians with MUM. Survival in patients receiving Tebentafusp-tebn was prolonged by approximately 6 months compared to standard care treatments. Response to treatment is typically evaluated through imaging. Imaging has its own challenges because of confounding factors. On the other hand, molecular markers that can gauge sensitively and specifically the response to treatment, and the speed of that response, could drastically improve the quality and speed of clinical decision making. In this project, we will explore a novel approach: we will develop biomarkers of response to treatment that are based on short non-coding RNAs (sncRNAs).

SncRNAs are important because they regulate the abundance of proteins, and, by extension, cell fate. Indeed, we now know that sncRNAs control numerous essential cellular processes, in health and disease. Advances during the last decade have demonstrated that the manner in which sncRNAs control proteins depends on tissue type and disease type. Thus, sncRNAs are very good candidates to serve as sensitive and specific diagnostic and prognostic tools, and this is why we chose them as the basis for this project.

The project has two parts. First, we will study liver tumors from MUM patients participating in a clinical trial at Jefferson. In these tumors, we will examine the levels of sncRNAs and of the RNA templates from which proteins are made. We will determine how these levels differ from what is seen in the primary tumors of UM patients who developed or did not develop metastases. We will determine how these levels change over time during treatment. Second, we will study sncRNAs in the blood of the same patients at different time points during treatment and determine how their levels differ from those in the blood of UM patients without metastases. We will determine how sncRNA blood levels change as treatment progresses and build a mathematical model to capture the speed of response to therapy. Once built, the model will allow us to accurately assess the response of patients while they are receiving the therapy.

Our short-term goal is to create biomarkers that leverage the power of sncRNAs in the MUM context. If successful, we expect to use the biomarkers in clinical settings to assist in clinical decision-making. Our long-term goal is to extend patient survival by creating novel targeted therapies for MUM. It is important to note that the methods we will develop in this project are very general in nature and can be used with other therapeutic regimens, and other cancers, where it is important to have molecular tests that accurately determine response to therapy in real-time.

Team Members

Eric Londin, PhD

Eric Londin, PhD
Assistant Professor
Department of Pathology, Anatomy and Cellular Biology

Marlana Orloff, MD

Marlana Orloff, MD
Alexander and Johnston Family Endowed Clinical Director of Uveal Melanoma
Associate Professor, Department of Medical Oncology

Mizue Terai

Mizue Terai, PhD
Research Assistant Professor
Department of Medical Oncology
Thomas Jefferson University 


Immunotherapy of Metastatic Uveal Melanoma Using iPSC Cell-Derived NK Cells

Mizue Terai, PhD

Principal Investigator

Mizue Terai, PhD
Research Assistant Professor
Department of Medical Oncology
Thomas Jefferson University 

Uveal melanoma (UM) is a rare eye tumor that originates from pigment-producing cells (melanocytes) in the eye. Although rare compared to skin melanoma, it is the most common primary intraocular malignancy in adults. Over the past decade, substantial progress has been achieved in the treatment of primary eye lesions while preserving the affected eye. Nevertheless, up to 50% of UM patients subsequently develop systemic spread (metastasis) after successful therapy of the primary tumor in the eye. Once UM cells escape from the eye, about 80-90% of them spread to the liver where tumors rapidly progress, and patients die within four to 15 months. Metastatic UM (MUM) is highly resistant to chemotherapy and other tumor inhibitors. Therefore, there is still an unmet need for better treatment for MUM patients.

Immunotherapy utilizes the body’s immune system to recognize and destroy cancer cells. In the past decade, immunotherapy has become a powerful approach in treating various cancers including metastatic skin melanoma. However, some cancer cells create an immune-suppressive environment by either reducing the expression of antigens on their cell surface recognized by immune cells or by other mechanisms that escape the attack by the immune system.

Adoptive Cell Therapy (ACT) is a new form of immunotherapy that uses human or genetically engineered immune cells to eliminate cancer. ACT is a promising approach, especially for solid tumors, like MUM, which have shown low response to other therapies. ACT has been approved by the Food and Drug Administration (FDA) to treat hematologic (blood) malignancies. Natural killer (NK) cells are one of the key components in the immune system and are an ideal candidate for ACT, because MUM cells tend to express molecules recognized by NK cells. To overcome the technical challenge of conventional NK cell therapy, which is the limitation of NK cells obtained from human or cell lines, we collaborated with HEALIOS K.K. from Japan, one of the leaders in engineered stem cell (iPSC, induced pluripotent blood cells) therapy. iPSC benefits include genetic engineering, increased expression of cytokines and chemokines and their receptors, and a loweredrisk of infection and rejection, because there are no donor cell requirements. We will explore the potential of iPSC-derived NK cells in the treatment of MUM.

To achieve the goal, we proposed the following two projects in this team research award application:

Aim 1: Investigate the anti-tumor activity of iPSC-NK cells against MUM cells. To investigate the activities of iPSC-NK cells against MUM cells, at least three MUM cell lines will be employed to study the killing activities and the production of immune stimulants by iPSC-NK cells. In addition, we will investigate the migration of iPSC-NK cells toward MUM cells.

Aim 2: Investigate in vivo activity of iPSC-NK cells in MUM mouse models. This aim is to test the accumulation and killing activity of iPSC-NK cells against MUM in the MUM metastasis mouse model. We have already established a MUM liver metastatic mouse model using NSG mice without mouse immune cells. We will investigate MUM tumor growth suppression by iPSC-NK cells in mouse models.

Our future goal of this project is to conduct a clinical trial of liver direct infusion therapy for MUM patients using immune cells, such as NK cells, to eliminate MUM effectively. Our overall goal is to completely eradicate MUM cells in patients with MUM.

Team Members

Vitali Alexeev, PhD

Vitali Alexeev, PhD
Research Assistant Professor
Departments of Dermatology and Medical Oncology
Thomas Jefferson University

Sota Deguchi, MD, PhD

Sota Deguchi, MD, PhD
Postdoctoral Fellow
Department of Medical Oncology
Thomas Jefferson University

Yuri Sykulev, MD, PhD

Yuri Sykulev, MD, PhD
Professor, Department of Microbiology & Immunology
Thomas Jefferson University


2022 Team Awardees

Yuri Sykulev

Principal Investigator

Yuri Sykulev, MD, PhD
Professor, Department of Microbiology & Immunology
Thomas Jefferson University

Watch Video »

Yuri Sykulev, MD, PhD, is leading a team with a focus on developing effective T cell therapeutics for treatment of metastatic uveal melanoma (UM). T cell therapies have shown promising responses in multiple refractory solid tumors including skin melanoma. Thus far, the most promising T cell therapy to treat skin melanoma is infusion of tumor infiltrating lymphocytes (TIL), which recognize tumor-specific neo-antigens abundant in skin melanoma. The barrier to successful treatment of UM with TIL is much lower frequency of tumor-reactive T cells within the metastases and strength of their anti-tumor response. To overcome the barriers for production of highly effective UM-specific T cell therapeutics, researchers propose to utilize TCR genes that recognize driver mutations of UM (GNAQ/GNA11 mutations). The researchers design to incorporate these TCR genes to otherwise non-specific T cells to manufacture UM-specific TCR-T cells to produce “off-shelf” products. The research team will investigate various strategies to generate these melanoma-specific TCR-T cells and will compare the ability of these cells to control the growth of human UM cells in vitro and in mouse models. The goal of this multi-phased project is to begin the development of effective T cell therapeutics, which would change the treatment for future uveal melanoma patients.

Team Members

Vitali Alexeev

Vitali Alexeev, PhD
Research Assistant Professor
Departments of Dermatology and Medical Oncology
Thomas Jefferson University 

Nadia Anikeyeva

Nadia Anikeyeva, PhD
Research Instructor
Department of Microbiology and Immunology
Thomas Jefferson University 

Mizue Terai

Mizue Terai, PhD
Research Assistant Professor
Department of Medical Oncology
Thomas Jefferson University 

Gyorgy Hajnoczky

Principal Investigator

Gyorgy Hajnoczky, MD, PhD
Director, Mitochondrial Research Center
Professor, Thomas Jefferson University

Watch Video »

Gyorgy Hajnoczky, MD, PhD, is leading a team with a focus on mitochondrial diagnostics and therapy to look into the development of novel therapeutics for uveal melanoma. This research team brings together expertise in uveal melanoma liver metastases (Mizue Terai, PhD, Assistant Professor, Thomas Jefferson University), the inherited defect (Philip B. Wedegaertner, PhD, Professor, Thomas Jefferson University), and therapeutic cell death activation (Gyorgy Hajnoczky, MD, PhD, Professor, Thomas Jefferson University) to test this hypothesis and understand the disease process and therapy of uveal melanoma. Piyush Mishra, PhD, a senior fellow, is the primary driving force in moving this uveal melanoma project further.

The group discovered that primary liver cancer and normal liver have distinctive cell death signatures, and developed an approach based on this difference to kill hepatocarcinoma without harming normal liver. Because uveal melanoma mortality is primarily due to its liver metastases, they speculated whether the liver metastases are also different from normal liver in their cell death signature and can be selectively targeted with the therapy that is effective for primary liver cancer. This project has the opportunity to develop new therapeutics for this disease, which would have an immeasurable impact on metastatic uveal melanoma patients.

Team Members

Sergei Koshkin

Sergei Koshkin, PhD
Postdoctoral Research Fellow
Department of Medical Oncology
Sidney Kimmel Cancer Center
Thomas Jefferson University

Jiansong Luo

Jiansong Luo, PhD
Research Associate
Biochemistry & Molecular Biology
Thomas Jefferson University

Piyush Mishra

Piyush Mishra, PhD
Postdoctoral Research Fellow
Pathology
Thomas Jefferson University

Mizue Terai

Mizue Terai, PhD
Research Assistant Professor
Department of Medical Oncology
Thomas Jefferson University

Philip Wedegaertner

Philip Wedegaertner, PhD
Professor
Department of Biochemistry & Molecular Biology
Sidney Kimmel Cancer Center
Thomas Jefferson University