Asmin Tulpule, MD, PhD

Funded by the Dick Vitale Pediatric Cancer Research Fund

We study a set of bone cancers that affect children and young adults. The treatment for these cancers has remained the same for the last 40 years – combinations of toxic medicines known as chemotherapy, followed by surgery or radiation to remove what is left of the cancer. While this strategy cures some patients, far too many children continue to die from these cancers. We believe we have found two specific weaknesses in these tumors: problems in their ability to repair damage to their DNA and a survival signal in a special pool of cancer cells known as “residual disease”. Through this research, we hope to bring new therapies to patients and cure more children with these bone tumors.

Peter Westcott, PhD

Funded with support from Carrie Collins in memory of Marty Collins

Immunotherapy helps the immune system recognize and kill cancer and it can cure patients where other treatments fail. Unfortunately, it still does not work for most patients. It is the goal of our research to understand why. Without a clear understanding of how cancer talks with the immune system, and how this conversation changes as cancer progresses, it is difficult to identify the root causes of why immunotherapy fails. Studying cancer evolution in patients is also challenging, as we rarely have the full history of tumor development and there is huge variability between tumors from one patient to the next. Through innovative genetic engineering, we are developing new mouse models of cancer that allow us to carefully study cancer development at all stages of the disease, especially at the moment when tumors acquire the ability to invade into other tissues—the reason cancer is so deadly. Why and how the immune system fails to stop cancer invasion and metastasis is not well understood and is a question of great importance. We will use the models we developed to study this question in creative and powerful new ways. We will also test exciting new immunotherapies, like cancer vaccines, in our models and determine why some tumors respond to treatment and others do not. Through this work, we hope to help match patients with the right immunotherapies and develop better immunotherapies that will be effective for many more patients.

Lindsay LaFave, PhD

Lung cancer is the deadliest cancer in the United States and lung adenocarcinoma is the most common type of lung cancer. While genetic mutations contribute to the development of cancer, cancer cells also activate gene programs over time that allow the cancer cells to become more aggressive and harder to treat. Advanced lung cancer cells evade current treatments such as chemotherapy or therapies that target the immune system. In our work, we have found that late-stage lung cancer cells expressed a unique transcription factor that activates gene programs which permit cancer cells to spread throughout the body. Of note is that these cancer cells also release molecules which we believe signal myeloid cells to enter the tumor. In doing so, the myeloid cells cause the immune system’s T-cells to be less effective and reduce how well current treatment strategies work. We seek to understand how late-stage cancer cells facilitate disease progression and how they limit response to current therapies. We have generated new mouse models which will allow us to investigate the gene programs that are active in these advanced cancer cells and to determine how these cells become resistant to therapy. Overall, our goal is to identify new options for targeting late-stage cancer cells which could be combined with, or used in place of, current treatment strategies so that we can increase how long patients with lung cancer live and improve their quality of life.

Hee Won Yang, PhD

NRAS mutations are found in about 30% of melanoma, a dangerous type of skin cancer. Although recent advancements in melanoma treatments have helped many patients, those with NRAS-mutant melanoma still face challenges. Available treatments for these patients are often not effective, and their cancer can quickly become resistant to treatment. Recently, scientists have developed new drugs called pan-RAS inhibitors that can directly target the NRAS mutations responsible for tumor growth. These drugs have the potential to greatly improve treatment for people with NRAS-mutant melanoma, but we need to learn more about potential resistance to these new drugs. This knowledge will help us develop better treatments for this type of cancer. Studying drug resistance is difficult because tumor can be very different from one another. To overcome these challenges, our study uses advanced technology to observe how individual melanoma cells grow and change. Our approach allows us to monitor the rare cells that adapt to the new pan-RAS inhibitors, helping us understand why some cells become resistant. We will also compare the genes in these adapting cells to those in cells that do not adapt to determine what makes them different. By learning how NRAS-mutant melanoma cells adapt to new treatments, we can design better therapies for patients with this type of cancer. This will help us meet the needs of people with limited treatment options and improve their chances of recovery. Our research aims to move the body of knowledge forward, positively impacting cancer patients and cancer research.

Poulikos Poulikakos, PhD

Funded by the Constellation Brands Gold Network Distributors

Cancer often occurs because some pathways in our body’s cells become too active, and these pathways are the same ones normal cells use to function properly. Researchers made drugs to target these pathways and slow down cancer growth. However a major problem is that these drugs can also affect normal cells and cause harmful side effects. Our research focuses on a specific type of cancer called RAS-mutant, which represents more than a third of human tumors, including lung, colorectal, pancreatic, and skin cancers. RAS mutations cause the RAS pathway in cells to go into overdrive, and that leads to uncontrolled cell growth, causing cancer. Scientists have developed drugs to target the  RAS pathway, like RAF and MEK inhibitors. However, these drugs have limitations because they can cause toxic effects in normal cells. The goal of our research is to find better ways to treat RAS-mutant cancers. We aim to understand why the drugs cause toxicities in normal cells by studying samples from patients and run experiments in the lab. We also found certain combinations of drugs that work better in cancer cells compared to normal cells. We will test these combinations in the lab and on animals to determine if they can effectively treat cancer without causing too many side effects. The ultimate goal of this research is to gather strong evidence to support quick clinical testing of these treatments in patients with RAS-mutant tumors, so we can develop better and safer treatments for people with these cancers.

Jonathan Peled, MD, PhD

Multiple myeloma is a type of cancer that affects the blood and bone marrow. Although there are many treatments, it almost always comes back. Scientists are looking for new treatments. Studies have shown, perhaps surprisingly, that the body’s ability to control a cancer is affected by the microbiome. The microbiome is the collection of bacteria that live in the gut. We hypothesize that myeloma, and how the immune system fights it, might respond to signals from the gut microbiome.

We are planning a new clinical trial to see if a fermented food product that may protect the microbiome will help nurture the microbiomes of patients getting bone marrow transplants. We will use samples collected from the trial participants to determine the effect of the fermented food on the microbiome and the metabolites that get into the patients’ bodies. Then we will study what happens to the immune system of patients in the trial. Finally, we will give myeloma to mice in the laboratory, while treating them humanely, and ask if antibiotics affect how the cancer behaves.

This study is important because it could help us understand how the microbiome affects MM and how to prevent cancer from coming back after treatment. The findings may also be helpful for developing new treatments for other types of cancer. Ultimately, we hope this will make people feel better and live longer.

Marina Konopleva, MD, PhD

Nick Valvano Translational Research Grant*

Acute Myeloid Leukemia (AML) is a blood cancer that arises from cells that normally fight infections in the body. However, these cells can become fast-growing and hard to kill, which causes their over-production. Eventually, healthy cells in the blood stop working because the diseased cells take over. Patients with AML are often treated with a novel drug called Venetoclax, which kills the majority of AML cells. However, residual cancer cells that did not die eventually re-populate the body leading to the patient’s death. In this research proposal, we identified the protein BAX as a key effector of cancer cell killing by Venetoclax. We also made several scientific observations about Venetoclax and BAX that are critical to understanding why this drug works for some cancer cells and not others. A scientific goal of this V Foundation Award is to provide the scientific reasoning for why Venetoclax does not always work in AML patients. At the same time, a therapeutic goal is to examine the new drugs that directly activate BAX, which restores its ability to kill AML cells. Our scientific goal is to work together and to provide a deeper knowledge of cancer therapies with the aim of cancer cures.

Eva Hernando-Monge, PhD

Most people who die from skin cancer died because their cancer has spread to the brain. Recent progress in treating patients whose cancer has spread to other organs has not kept pace for patients in whom skin cancer spread to the brain. At least part of the reason for this is likely because the environment in the brain is so different from other parts of the body. To address this urgent need for better treatments developed specifically for patients with skin cancer who then develop brain tumors, we looked for genes that might help cancer cells that spread from the skin to adapt so they can do well in the brain. We have identified a molecule that explain how it might help skin cancer cells to adapt to the brain. Already, we have encouraging evidence of how this molecule allows tumor cells to survive and grow inside the brain. Equally exciting is that there are already ways to block this gene function by taking a pill or injection, which will allow us to test if we can prevent or reverse the spread of skin cancer to the brain in our models, and eventually in patients. However, we first need to better understand exactly how important this process is in helping skin cancer cells to adapt to the brain microenvironment, and gather more information about how this gene seems to help skin cancer cells to invade the brain and adapt to a new environment.

Alexandra Miller, MD, PhD

Funded by the Dick Vitale Pediatric Cancer Research Fund

Primary brain tumors are the most common solid tumors in children. They are also the most frequent cause of cancer-related death in children and teens. Genetic profiling is an important tool in the treatment of these tumors. DNA sequencing provides information for proper diagnosis. It can also be used to understand how tumors change over time and to monitor response to treatment. However, performing biopsies is very challenging for brain tumors.  Many tumors are in important areas of the brain and can’t be fully removed or repeatedly sampled.

“Liquid biopsy” is a new tool that can be used to diagnose cancer and track response for some systemic tumors. It works by detecting small pieces of DNA that break off from tumors. These can be found in the cerebrospinal fluid (CSF) and in blood (circulating tumor DNA, ctDNA). Accessing these “liquids” is usually easier and has fewer complications than surgery.

We previously showed that CSF ctDNA can be used to diagnose brain tumors and that ctDNA is associated with active disease. But there are instances where CSF ctDNA is not informative due to technical limitations. We propose to improve how these samples are analyzed so CSF liquid biopsies can help more patients. Our prior work was retrospective. For this project, CSF ctDNA monitoring will be added to a clinical trial. We will investigate whether there is a relationship between CSF ctDNA and disease burden. Validating CSF liquid biopsy could greatly improve how pediatric primary brain tumors are diagnosed and treated.

Christopher R. Vakoc, MD, PhD

Co-funded by the Dick Vitale Pediatric Cancer Research Fund and the Jeff Gordon Children’s Foundation

What big question(s) will your work answer? Rhabdomyosarcoma is a deadly cancer that occurs in children and young adults. Several decades of research points to a specific molecule (called PAX3-FOXO1) as the most compelling drug target in this disease. However, we simply do not understand the molecular details of PAX3-FOXO1 enough to made a medicine that exploits this target. The big question addressed in this project will be to understand this compelling target with atomic detail by applying innovative technology. • Why does this question matter? Children continue to die of rhabdomyosarcoma and yet the medicines used in the clinic are woefully inadequate and toxic. A new therapy tailor-made for this disease could change everything. • How will your work answer the big question? Our work has the potential to provide a basic science foundation upon which a drug discovery campaign could be launched.

Mailing List Mailing List
Close Mailing List