New Discoveries and Honors

Read about the latest discoveries by Damon Runyon scientists and honors received by scientists in the Damon Runyon scientific community.

December 4, 2017

Craig J. Ceol, PhD (Damon Runyon Fellow '05-'07) of the University of Massachusetts Medical School, Worcester, and colleagues, have identified a new protein that is involved in metastatic melanoma, the most deadly form of skin cancer. The protein, GDF6, is part of a class of proteins called “growth differentiation factors” that helps cells divide and differentiate into specific cell types.

November 30, 2017

Chuan He, PhD (Damon Runyon Fellow '00-'02) of the University of Chicago, Chicago, was named one of this year's winners of the 2017 Paul Marks Prize for Cancer Research. The award recognizes promising investigators aged 45 or younger at the time of nomination for their efforts in advancing cancer research.


November 23, 2017

Matthew L. Meyerson, MD, PhD (Damon Runyon Fellow '95-'98), of the Dana-Farber Cancer Institute, Boston, and colleagues, reported a study that provides clues to the role Fusobacteria may play in the development of human colon cancers. Meyerson was one of the first scientists to discover that Fusobacteria, which normally inhabit the mouth, are closely associated with colon cancer cells but not normal colon cells. Researchers have now confirmed the presence of Fusobacteria in up to half of all colon tumors.

November 9, 2017

This month, Damon Runyon scientists published promising findings on “liquid biopsies.” This non-invasive method isolates and studies circulating tumor DNA (ctDNA)-- free-floating pieces of DNA found in blood plasma that are shed from tumor cells. These studies demonstrated that liquid biopsies are becoming an important tool for monitoring cancer progression, as well as identifying treatment strategies and drug resistance earlier than traditional approaches.

November 8, 2017

Ronald J. Buckanovich, MD, PhD (Damon Runyon Clinical Investigator ’08-’11) of the University of Pittsburgh, and colleagues, have developed a process that can grow hundreds of cultured cell masses, called spheroids, from just a few tumor cells derived from a patient. This 3D method yields cells that grow and multiply just as they would inside the body. Currently, researchers are limited to two-dimensional cells grown in petri dishes, which often do not respond to medicines the same way as ovarian cancer cells inside the body.

November 2, 2017

Increasing evidence shows that diet plays a major role in the development of some cancers. Andrew T. Chan, MD, MPH (Damon Runyon Clinical Investigator ‘08-‘13), of Harvard Medical School and Massachusetts General Hospital, Boston, and colleagues, found that eating more fiber after colorectal cancer diagnosis is associated with a lower risk of dying from colorectal cancer.

October 18, 2017

A new class of treatments called CAR-T therapy is providing options for patients who have all but lost hope in their fight against cancer. This form of immunotherapy is based on genetically enhancing a patient’s own immune cells to target and kill their cancer. The Food and Drug Administration approved Yescarta for adults with a form of blood cancer called non-Hodgkins lymphoma.

October 16, 2017

The National Academy of Medicine announced the election of 80 new members. Election to the Academy is considered one of the highest honors in the fields of health and medicine and recognizes individuals who have demonstrated outstanding professional achievement and commitment to service. Damon Runyon congratulates the four alumni who were elected this year:


Scott A. Armstrong, MD, PhD (Clinical Investigator ’03-’08), Dana-Farber Cancer Institute

October 5, 2017

Gordon J. Freeman, PhD (Damon Runyon Fellow ’79-’81), Dana-Farber Cancer Institute, Boston, is one of five scientists honored by the 2017 Warren Alpert Foundation Prize for discoveries focused on “Immune Checkpoint Blockade and the Transformation of Cancer Therapy.” Collectively, their work has elucidated foundational mechanisms in cancer’s ability to evade immune recognition through the CTLA-4 and PD-1 pathways and, in doing so, has profoundly altered the understanding of disease development and treatment.

October 5, 2017

The National Institutes of Health’s High-Risk, High-Reward Research program, funded 86 awards to exceptionally creative scientists proposing to use highly innovative approaches to tackle major challenges in biomedical research. The program is designed to accelerate scientific discovery by supporting high-risk research proposals. Applicants of the program are encouraged to think outside-the-box and to pursue exciting, trailblazing ideas.

September 29, 2017

Liron Bar-Peled, PhD (Damon Runyon Fellow ‘14-‘17) of the Scripps Research Institute, La Jolla, developed a new proteomics-based approach to discover small-molecule inhibitors that could be used as anti-cancer therapies. The approach is based on the fact that certain amino acids on proteins have a special chemical reactivity that allows them to form irreversible covalent bonds with suitably designed probe or "scout" molecules.

September 13, 2017

Alexandra Zidovska, PhD (Damon Runyon Fellow ‘10-‘12) of New York University, New York, has discovered the “internal clock” of live human cells using state-of-the-art fluorescence microscopy. Previously, the only way to tell the precise point of a cell in its life cycle was by studying dead cells. Alexandra’s lab has found that the nuclear envelope, which separates the nucleus with the DNA from the rest of the cell, has a previously undetected type of motion: it fluctuates in shape every few seconds.

September 11, 2017

Kristopher R. Bosse, MD (Damon Runyon Physician Scientist '16-'20) of the Children's Hospital of Philadelphia, is engineering a new drug to selectively target neuroblastoma cells and deliver a chemotherapeutic agent into the cells. Neuroblastoma is a cancer of the developing nervous system that usually occurs as a solid tumor in a child's chest or abdomen, and is the most common cancer in infants.

August 15, 2017

Five researchers have been announced as the recipients of the Albany Medical Center Prize in Medicine and Biomedical Research for 2017. They have made important contributions to the development of CRISPR-Cas9, a gene engineering technology that harnesses a naturally occurring bacterial immune system process. The technology has revolutionized biomedical research and provided new hope for the treatment of genetic diseases and more.

August 1, 2017

The FDA approved Idhifa for acute myeloid leukemia (AML), the result of important contributions from Hai Yan, MD, PhD (Damon Runyon Scholar ‘05-‘07), of Duke University Medical Center, Durham, and Omar Abdel-Wahab, MD (Damon Runyon Clinical Investigator ‘13-‘16), of Memorial Sloan Kettering Cancer Center, New York. They made independent discoveries about mutations in the IDH2 gene and how these contribute to the development of AML.

July 21, 2017

Marcela V. Maus, MD, PhD (Damon Runyon-Rachleff Innovator ’17-’18) at Massachusetts General Hospital, Boston, published results from her clinical trial with glioblastoma patients showing for the first time that CAR (chimeric antigen receptor) T cells cross the blood-brain barrier to reach tumors and appeared to be safe. These CAR T cells were targeted to EGFR variant III in glioblastoma patients.

July 5, 2017

Catherine J.Wu, MD (Current Damon Runyon Physician-Scientist Mentor and Clinical Investigator '07-'12) of Dana-Farber Cancer Institute, Boston, has led one of the first studies that demonstrates the potential of personalized cancer vaccines. The study focused on six people with advanced melanoma, a type of skin cancer. The participants had surgery to remove their tumors, but about half of all such patients face a recurrence of cancer.

May 8, 2017

Three Damon Runyon scientists received 2017 Pershing Square Sohn Prizes for Young Investigators in Cancer Research. Recipients receive $200,000 per year for up to three years and opportunities to present their work to scientific and business audiences, helping to bridge the gap between the academic and business communities. This year, three of the six awards were granted to Damon Runyon scientists:  


Yimon Aye, PhD (Damon Runyon Fellow ’09 – ’12), Weil Cornell Medicine, New York


Daniel A. Heller, PhD (Damon Runyon Fellow ‘10 – ’12), Memorial Sloan Kettering Cancer Center, New York


Eirini P. Papapetrou, MD, PhD (Damon Runyon-Rachleff Innovator ’14 – ’17), Icahn School of Medicine at Mount Sinai, New York


May 3, 2017

C. Ryan Miller, MD, PhD (Damon Runyon Clinical Investigator ’09-’12) of the UNC Lineberger Comprehensive Cancer Center, Chapel Hill, and colleagues, reported two studies on the genetics underlying brain tumors. The first study showed that mutations in MAPK and PI3K affect how cancer starts in glial cells, brain cells that provide support and insulation for neurons. These mutations triggered tumor initiation and produced increasingly dense low-grade gliomas that quickly progressed to aggressive and often deadly glioblastoma (GBM).

May 2, 2017

Election to the National Academy of Sciences is one of the highest honors that can be earned by a U.S. scientist.  In recognition of their distinguished and continuing achievements in biomedical research, members of the Damon Runyon community of scientists were inducted this month:  


Ardem Patapoutian, PhD (Damon Runyon Scholar ’03-‘05, Fellow ’96-‘99), Scripps Research Institute, La Jolla


Guillermina Lozano, PhD (Former Fellowship Award Committee Member), M.D. Anderson Cancer Center, Houston


April 13, 2017

Feng Zhang, PhD (Damon Runyon-Rachleff Innovator ’12-’14) and colleagues at the Broad Institute, Cambridge, have developed a new CRISPR-based genetic diagnostic tool that may make it faster, less expensive, and easier to diagnose acute and chronic diseases like Zika, Ebola, cancer, and other hereditary disorders. The new tool dubbed SHERLOCK (Specific High-sensitivity Enzymatic Reporter unLOCKing) can detect extremely low concentrations of Zika virus and cancer DNA in blood, urine, and saliva samples.

April 10, 2017

Jedd D. Wolchok, MD, PhD (Damon Runyon-Lilly Clinical Investigator ‘03-‘08) at Memorial Sloan Kettering Cancer Center, New York, and colleagues, reported that matching the size of a tumor to the body's immune response could help doctors tailor immunotherapy treatments for melanoma patients whose disease has spread.

March 29, 2017

David M. Livingston, MD (Damon Runyon Board Member), of Dana-Farber Cancer Institute, Boston, has been named the 20th recipient of the Pezcoller Foundation-AACR International Award for Cancer Research. Dr. Livingston is honored for his fundamental contributions to the field of basic cancer research. His work has been pivotal to the understanding of retinoblastoma pathway of cell cycle control as well as the transcriptional co-activation function of the key regulatory proteins, p300 and CBP.

March 24, 2017

At the junction of growth and starvation stands a signaling protein called mechanistic Target of Rapamycin Complex 1 (mTORC1). Inside the cell, mTORC1 regulates metabolism, growth, protein and organelle recycling (autophagy), proliferation, and survival. When something goes wrong in the pathway, various diseases such as cancer, obesity, and type 2 diabetes, can develop.

March 14, 2017

Anjana Rao, PhD (Damon Runyon Fellow ’79), and James Scott-Browne, PhD (Damon Runyon Fellow ’11-’13), at the La Jolla Institute for Allergy and Immunology, La Jolla, and colleagues, are focusing on a key issue of how tumor-fighting T cells can lose their effectiveness or become “exhausted.”  The researchers identified two proteins, NFAT and Nr4a, that can bind to the DNA of T cells and shut down their tumor-fighting activity.  Next steps will be to determine if these processes can be interfered with or reversed in ord

February 27, 2017

Don X. Nguyen, PhD (Damon Runyon Fellow ’05-’08), of Yale Cancer Center, New Haven, and colleagues, reported new findings that explain the propensity of latent lung adenocarcinoma (LUAD) to relapse. They showed that differential expression of extracellular matrix (ECM) molecules and their interacting proteins contributes to risk of relapse in distinct LUAD subtypes. One protein called hyaluronan receptor HMMR, when overexpressed, was associated with inflammation and poor prognosis.

February 23, 2017

Christine Iok In Chio, PhD (Damon Runyon Shirley Stein Fellow ’13-’17), in the laboratory of her sponsor David Tuveson, MD, PhD, and colleagues at Cold Spring Harbor Laboratory, Cold Spring Harbor, created a new 3D model of pancreatic cancer, which allowed them to identify two distinct stroma cell populations called cancer-associated fibroblasts  (CAF’s), that work together with cancer cells to protect and help the tumor grow.

January 25, 2017

Gregory L. Beatty, MD, PhD (Damon Runyon-Nadia’s Gift Foundation Innovator ’12-’15) and colleagues at the Abramson Cancer Center at University of Pennsylvania, Philadelphia, reported high levels of inflammatory compounds in mice with pancreatic tumors. These included CCL2, a signaling molecule that promotes recruitment of inflammatory white blood cells by tumors. This likely contributes to the protective tumor microenvironment that makes most pancreatic tumors resistant to treatment.

January 24, 2017

Theodora S. Ross, MD, PhD (Damon Runyon Scholar ’01-’03), and colleagues at UT Southwestern, Dallas, reported that the BRCA1 gene is required for the survival of blood forming stem cells. This could explain why patients with BRCA1 mutations do not have an elevated risk for leukemia; the stem cells die before they have an opportunity to transform into a blood cancer. These results also suggest that these patients may be at higher risk for the serious side effects of chemotherapy. The study was published in Cell Reports.


January 23, 2017

Akinyemi I. Ojesina, MBBS, PhD (Damon Runyon Fellow ’08-’11), of University of Alabama at Birmingham, worked with The Cancer Genome Atlas Research Network to identify novel genomic and molecular characteristics of cervical cancer that will aid in the subclassification of the disease and may help define personalized therapies for each individual patient.

January 23, 2017

Elaine V. Fuchs, PhD (Damon Runyon Board Member, Damon Runyon Fellow ‘77-‘79) of The Rockefeller University, New York, has been named the recipient of the 2017 McEwen Award for Innovation. The prize, given by the International Society for Stem Cell Research, recognizes groundbreaking work pertaining to stem cells or regenerative medicine. Dr. Fuchs studies adult skin stem cells, how they make and repair tissues, and how cancers develop.


January 23, 2017

Pardis C. Sabeti, MD, DPhil (Damon Runyon Fellow ‘04-‘06) of Harvard University, Cambridge, will receive the 2017 Richard Lounsbery Award from the National Academy of Sciences.

January 13, 2017

Elaine V. Fuchs, PhD (Damon Runyon Board Member, Damon Runyon Fellow ‘77-‘79) and Shruti Naik, PhD (Damon Runyon Fellow ’14-’18) at The Rockefeller University, New York, and colleagues, found that skin squamous cell carcinomas alter the protein-making machinery to preferentially use tumor-related mRNAs, leading to the production of proteins important for cancer progression. This switch is linked to a ribosome initiation factor called eIF2 and transition initiation factor eIF2A.

January 12, 2017

Trudy G. Oliver, PhD (Damon Runyon-Rachleff Innovator ’13-’15), and colleagues at the Huntsman Cancer Institute at the University of Utah, Salt Lake City, reported the generation of a new mouse model for studying small cell lung cancer (SCLC). They demonstrated that Myc oncogene expression cooperates with Rb1 and Trp53 loss in the mouse lung to promote aggressive, highly metastatic tumors that are initially sensitive to chemotherapy followed by relapse.

January 5, 2017

Feng Zhang, PhD (Damon Runyon-Rachleff Innovator ‘12-‘14) of the Broad Institute, Cambridge, and colleagues, reported the discovery of new types of RNA-targeting CRISPR systems, which utilize a novel Cas enzyme called Cas13b. Cas13b is capable of targeting and degrading RNA (rather than DNA, which is targeted by previous CRISPR systems), which will enable researchers to specifically manipulate RNA in a high-throughput manner and manipulate gene function more broadly.