Qing Chen, M.D., Ph.D.

Metastasis is the spread of cancer to one or more different organs of the body from where it started. The brain is one of the common organs for cancer recurrence. Even with aggressive treatments, brain metastasis is increasingly becoming a significant clinical problem. To find new therapeutic targets to treat brain metastasis, we need to first understand the progression of the disease.

Metastases are generally site specific. The environment of each organ is different. Cancer cells may only be able to colonize one or more specific organs, depending on the primary tumor from which the cells derive. As illustrated in the ‘seed and soil’ theory, tumor cells behave like seeds that can only successfully colonize selective organs that offer the right soil for their survival and growth. Thus, we plan to understand brain metastasis by focusing on the complex conversation between cancer cells (the seed) and brain cells (the soil). Using advanced microscopy techniques, we will directly visualize the metastatic brain tumors in the living animals. Meanwhile, we will detect therapeutic responses when newly designed treatments are applied. From these studies, we will obtain dynamic longitudinal changes in the cancer cells and the surrounding brain cells. This will allow “reconstruction” of the brain metastasis process, as well as therapeutic response. We strongly believe that these studies will yield new ways of fighting brain metastasis.

Andrei Thomas-Tikhonenko, Ph.D.

Funded by the Dick Vitale Gala

Over the past several decades, there has been a steady increase in cure rates in children with the so- called B-cell acute lymphoblastic leukemia (B-ALL), a type of blood cancer. Yet many B-ALL patients who failed the initial chemotherapy still die from their disease. Five years ago many of these high-risk patients began to benefit from immunotherapy, whereby patients’ own immune systems are trained to recognize and destroy the leukemic cells. One common form of immunotherapy is based on recognition of CD19, a protein residing on the surface of most leukemic cells. However, even this breakthrough treatment fails in about a third of patients, suggesting that other leukemia proteins need to be targeted in parallel. One alternative protein target is called CD22. CD22-directed immunotherapies show promise, but are not without their own record of failures. Our previous studies led us to believe that one common cause of treatment failure is improper assembly of the CD22 protein, resulting in re-shuffling of its key parts called ectodomains. This re-shuffling could result in CD22 becoming unrecognizable to the immune system. On the other hand, improperly assembled CD22 could be targeted using a new type of immunotherapeutics, which are trained to recognize improper junctions between ectodomains. The proposed work will test these ideas using leukemic cells grown in Petri dishes and in mice and samples from ongoing clinical trials. It will also extend our current studies to other cell surface proteins. In the end, the TVF-funded work would lead to a more precise matching of future patients to best possible treatments and thus much better outcomes.

Yanis Boumber, M.D., Ph.D.

Funded by the Stuart Scott Memorial Cancer Research Fund

Lung cancer is the main cause of death in the world. For unknown reasons, African Americans (AA) have more aggressive lung cancer compared to Caucasians. Recently, immunotherapy demonstrated that one out of five of patents have tumor shrinkage. Long term remissions are happening in one out of seven lung cancer patients. This is very exciting, but combinations of 2 or 3 immunotherapy drugs are needed to cure more patients. 

We proposed the lung cancer treatment combination that can block tumor blood vessel growth, and boost immune system. We think that this combination approach will cure more lung cancers. We will soon start a clinical study combining two immunotherapy drugs. One out of four patients on our study will be AA. We hope to find immune or blood vessel growth related markers to help predict who would benefit from this drug combination. This can help to use the right drugs for the right patients. In this study, we also plan to investigate why AA have more aggressive lung cancer. 

In Aim 1, we will perform detailed analysis of blood proteins and white cells from the blood of patients participating in our study. In Aim 2, we will correlate genes and other markers with response to immunotherapy combination. In Aim 3, we will compare blood proteins and tissue gene levels between AA and Caucasians.

Jessie Villanueva, Ph.D.

The V Foundation MRA Young Investigator Award

Co-funded with The Melanoma Research Alliance

Although significant progress has been made treating melanoma and the recent approval of several drugs for the treatment of advanced disease, several challenges remain.  For example, clinical responses are generally short-lived as tumors quickly become drug resistant and patients relapse. Moreover, tumors can develop drug resistance through a diverse number of molecular mechanisms, making the development of second-line therapies extremely daunting.  Therefore, it is critical to identify therapeutic targets that are common to the majority of resistant tumors.  We have recently found that a protein kinase called S6K is activated in melanomas resistant to BRAF and MEK inhibitors.  Moreover, we showed that inhibition of this protein using a triple drug combination blocked the growth of resistant tumors.  This provides strong rationale for establishing S6K as a novel target for melanoma therapy.  Notably, S6K is a common node for most resistance pathways.  We propose to investigate the role of S6K in melanoma and determine the therapeutic value of targeting this protein.  Towards these goals we will determine the consequences of blocking S6K in melanoma, identify the proteins that are regulated by S6K and use this knowledge to delineate combinatorial approaches that can lead to long-term tumor remission in a large number of melanomas, including those resistant to BRAF and MEK inhibitors.  We expect that the data generated by these studies can be quickly translated into new strategies aimed at maximizing the therapeutic efficacy of MAPK inhibitors in melanoma and provide actionable information that will guide the design of future clinical trials. 

Fiona Simpkins, M.D. & Eric Brown, Ph.D. & Payal Shah, M.D.

Funded by 2017 BRCA Fund-A-Need

Inhibitors of the poly(ADP-ribose) polymerase enzymes (PARPi) represent a significant advance in ovarian cancer treatment, particularly in those with inherited BRCA1 and BRCA2 mutations. These drugs are taken by mouth, are effective, and generally have fewer side effects than chemotherapy. However, responses to PARPi are generally limited and cancers develop treatment resistance. Inhibition of the ATR kinase offer a promising solution to this problem. Inhibitors of ATR (ATRi) and PARP have distinct and complementary effects. Data from our group shows that the PARPi-ATRi combination causes complete tumor regression in BRCA2-associated ovarian cancer animal models, an effect that is superior to that observed with PARPi alone. Based on these data, we plan a clinical trial of the PARPi, olaparib with an ATR inhibitor in patients with ovarian cancer. With support from the V Foundation, we have set out to further improve this therapeutic strategy. First, we will study tumor tissue from patients enrolled on the clinical trial to identify markers that predict if the treatment will be effective. Furthermore, we will use tumor tissue from patients to create animal models and use these animal models to test ATR inhibitor combinations with different PARP inhibitors, in addition to olaparib. We will determine which PARPi is most active in combination with ATRi, particularly in the BRCA1/2 mutation subset. Finally, we will use novel protein-based techniques to better understand exactly how the ATR and PARP inhibitors work together, which will permit further improvements of this therapeutic strategy. Our goal is to develop the most effective and well-tolerated treatment of BRCA1/2-mutant ovarian cancers for which standard therapies have faltered.

Nicola Mason, DVM, Ph.D. & Dana Clarke, VMD, DACECC

Funded in partnership with the American Kennel Club Canine Health Foundation

Bladder cancer or urothelial carcinoma (UC) affects approximately 40,000 dogs per year in the US with specific breeds including Scottish Terriers, West Highland White Terriers, Shetland Sheepdogs, Beagles, and Parson Russell Terriers being over-represented. Affected dogs usually display lower urinary tract clinical signs including bloody urine, frequent urination, difficulty and pain on urinating, and urinary outflow tract obstruction. Standard of care consists of anti-inflammatory drugs either alone or in combination with chemotherapy or radiation therapy. While these treatments can lead to stable disease for 6-12 months, they rarely lead to a cure, and most dogs eventually succumb to their disease. In human medicine, urinary bladder tumors have been shown to exhibit a high gene mutational burden which directly correlates with a favorable response to immune therapies. Canine UC exhibits a similar mutational load suggesting that the disease in dogs may also be immune responsive. In this study, the investigators will evaluate the safety and effectiveness of a novel targeted, immune therapy that aims to promote a powerful immune response against a specific gene mutation (V600E B-Raf) recently identified in up to 87% of dogs with UC. The investigators hypothesize that vaccine-induced anti-tumor immune responses will lead to tumor regression and that such favorable responses will correlate with the baseline mutational burden of the tumor. The investigators will use standard immunological methods and advanced genetic sequencing technology to study systemic and intra-tumoral immune responses to identify biomarkers that may predict immunological and clinical response in dogs.

Roger Greenberg, M.D., Ph.D.

Funded by 2017 BRCA Fund-A-Need

When an individual is born with one mutated (or abnormal) copy of BRCA1 or BRCA2, there is a high chance that they will develop cancer. Drugs called PARP inhibitors have been developed to take advantage of DNA repair deficiency in BRCA related cancer, but they do not work in all patients, and resistance to the medications frequently develops. There is a pressing need to more deeply analyze primary tumors in BRCA1/2 mutation carriers to see how often there are “subclones” that have other types of primary resistance to PARP inhibitors and other therapies. This information will be critical to rationally design new strategies that will overcome a broad spectrum of resistance mechanisms. Due to a higher burden of DNA mutations in BRCA1/2 related tumors, there is significant excitement about the prospect of using immune therapies for these cancers. However, further research is needed to understand the baseline immune status of BRCA1/2 tumors and the pathways in which the immune system can be turned ‘on’ in these tumors. We propose a novel strategy to use inflammatory and immune responses that target both sensitive and resistant cells within a tumor.

To address these important objectives, we present two integrated aims that utilize our collective expertise in cancer genomics, DNA repair, tumor immunology, and mouse models of cancer. Consistent with the goals of the Team Science Convergence Award application, researchers from multiple schools, departments and divisions at the University of Pennsylvania will work together to maximize innovation and productivity.

Shannon Maude, M.D., Ph.D.

Funded by Bristol-Myers Squibb

Cancer therapy has undergone a revolution in recent years as a result of the success of several immunotherapies, providing hope to patients with once-thought incurable cancers. One of the most powerful technologies is engineered T cell therapy. By reprogramming a T cell, a type of immune cell, to recognize and kill a cancerous cell, we have seen remissions in over 90% of children and young adults with very resistant acute lymphoblastic leukemia (ALL). Because T cells are living cells, they have the potential to last in the body for months or years and may provide long-term disease control. While most of these remissions are long-lasting, with 60% of responding patients still in remission at 1 year, relapse remains the key mode of treatment failure. One of the reasons for relapse is short lifespan of the engineered T cells in some patients. In this study, we will ask why T cells live many months to years in some patients while they live only weeks to a few months in others. We will use this information to open a clinical trial to test the combination of engineered T cells with a medication to augment T cell function. As we improve on the percentage of long-term remissions, we hope to not only increase the chance of cure but also reduce the number of patients who require intensive therapies, including bone marrow transplant.

Nester Esnaola, M.D., M.P.H.

Funded by Hooters of America, LLC

African Americans often do not take part in research.  To raise the community’s knowledge about breast cancer research, we will work with trusted members of the community through our Community Ambassador Training (CAT) program.  We will craft a training about the value of research and what it means to take part in research.  The training will build on their current knowledge through activities that highlight breast cancer.  Once trained, Community Ambassadors will take the information to the community spreading the word about the value of research.

Sameer Agnihotri, Ph.D.

Funded by WWE in honor of Connor’s Cure

Brain cancer is now the No. 1 cause of cancer-related deaths in children. A tumor known as pediatric high-grade glioma (PHGG) is the most deadly type. Even though children with PHGG get intense treatment, including surgery, radiation, and chemotherapy, most patients still die within two years of their initial brain cancer diagnosis. Part of the problem is that PHGG tumors are not all the same. However, our research has recently identified a clear group of PHGG tumors in which there is damage to the system of proteins that promote healthy cell growth. The system is supposed to work like the accelerator and brake pedals of a car, allowing the body to keep cell growth in control; but when gene mutations produce bad proteins, the system behaves as though the accelerator is stuck and the brakes have failed. The system becomes overactive and promotes unstoppable tumor growth. This system, called PI3K/AKT, is also a factor in many other aggressive cancers. We think that restoring the proper function of PI3K/AKT is possible and could halt or even shrink PHGG tumors. Our proposed research will test and validate new therapies to do this.

Mailing List Mailing List
Close Mailing List