For more than 50 years, the University of Chicago has been a leader in cancer research. Our scientists and clinicians have consistently expanded the boundaries of knowledge and transformed cancer care and prevention.

Seminal discoveries made at UChicago have stimulated the development and introduction of many of the cancer treatments used today. As the examples below demonstrate, many of the roots of chemotherapy, hormonal therapy, gene therapy and bone marrow transplantation can be traced to UChicago.

1930s - 1940s

Charles B. Huggins, MD, demonstrated that prostate cancers are dependent on hormones, work that earned him the Nobel Prize in 1966 and transformed prostate and breast cancer research and treatment.

Leon O. Jacobson, MD, studied the effects of nitrogen mustard as an anticancer agent in leukemia patients, laying the foundation for widespread use of chemotherapy to treat cancer.

1950s

Eugene Goldwasser, PhD
Eugene Goldwasser, PhD, began work on the hormone that stimulates red blood cell production, erythropoietin. He was the first to purify it in 1977, leading to widespread use of the recombinant hormone to treat anemia from the 1980s to today.
Elwood Jensen, PhD, demonstrated that tissues responding to estrogens concentrate the hormone through a putative receptor, which he subsequently identified. He won the 2004 Lasker Award for Basic Medical Research for this groundbreaking work that led to estrogen receptor-based therapies for breast cancer.
Bernard Roizman, ScD, unraveled the genetic and protein structure of herpes simplex virus, leading to the first engineered virus, which has been used to study and target lethal tumors in humans.

1960s

Katherine Austin Lathrop, MS, Paul Harper, MD, and Robert Beck optimized an imaging technique using the radioisotope technetium-99m (99mTc). To this day 99mTc scans are used worldwide to detect cancer and other diseases.
Shutshung Liao, PhD
Shutsung Liao, PhD, began to map the pathway of testosterone production, activity and binding of the androgen receptor, the gene he successfully cloned in 1988. Disruption of this pathway became the mainstay for prostate cancer treatment.

1970s

Janet Rowley, MD, identified the first chromosomal translocation in leukemia, the t(9;22) or Philadelphia chromosome, leading to the recognition of the genetic basis of cancer and, ultimately, targeted therapy. She won numerous awards for her transformative contributions, including the 1998 Lasker Award for Clinical Medical Research and the Presidential Medal of Freedom in 2009.
John Ultmann, MD, our founding Cancer Center director and leading National Cancer Act proponent, led international efforts to stage subtypes of lymphoma, which led to the development of more effective, less toxic therapies.
Charles Metz, PhD
Charles Metz, PhD, with Kurt Rossman, PhD, pioneered radiology image analysis methods now used worldwide for quantitative evaluation of diagnostic performance. Pictured: Charles Metz, PhD
Leslie DeGroot, MD, and Samuel Refetoff, MD, were key figures in unraveling an epidemic of thyroid cancers. In 1975, they published a study showing that thyroid cancer developed frequently in people who were previously treated with X-rays. 
Elwood Jensen, PhD, and Geoffrey Greene, PhD
Geoffrey Greene, PhD, right, with Elwood Jensen, PhD, left, developed monoclonal antibodies to the estrogen receptor, leading to the standard biomarker test performed in modern breast cancer management.
George Block, MD, developed aggressive surgical techniques for treatment of colorectal cancer and promoted their widespread adoption based on improved outcomes observed in his patients.

1980s

Frank Fitch, MD, PhD
Frank Fitch, MD, PhD, defined the process of how CD4+ T cells help the cytolytic activity of CD8+ T cells’. This discovery paved the way to harnessing the anti-tumor capabilities of CD8+ T cells for cancer immunotherapy.
Harvey Golomb, MD, performed one of the first clinical trials with the cytokine a-interferon, leading to FDA approval for its use in treating hairy cell leukemia.
Everett Vokes, MD
Everett Vokes, MD, and Ralph Weichselbaum, MD, pioneered the combined use of chemo- and radiotherapy for the treatment of head and neck cancer, which becomes the standard of care. Pictured: Everett Vokes, MD

1990s

Michelle Le Beau, PhD
Michelle Le Beau, PhD, with Janet Rowley, MD, mapped the first chromosome translocation using fluorescence in situ hybridization (FISH) which became a standard diagnostic tool for blood cancers. Pictured: Michelle Le Beau, PhD
Craig Thompson, MD, uncovered mechanisms regulating lymphocyte survival and death, including BCL-xL, an anti-apoptotic therapeutic target for cancer.
Jeffrey Bluestone, PhD, elucidated the mechanisms by which CTLA-4 signaling inhibits T-cell function, paving the way for the development of ipilimumab, an immunotherapy that effectively targets CTLA-4.
Elaine Fuchs, PhD
Elaine Fuchs, PhD, pioneered reverse genetics, now a commonly used tool, to understand how defective protein function causes skin cancer.

2000 - 2004

James Vardiman, MD, co-led the development of the World Health Organization classification of leukemias and lymphomas, which became the international diagnostic system.
John Crispino, PhD, identified a genetic defect that causes the development of leukemia in infants and children with Down syndrome, providing key insights into the molecular basis of the disease.
Thomas Gajewski, MD, showed immunization with interleukin-12 and engineered peripheral blood mononuclear cells has activity in melanoma patients, setting the stage for modern cancer immunotherapies.
Sonali Smith, MD
Sonali Smith, MD; Richard Larson, MD; Michelle Le Beau, PhD; and James Vardiman, MD, delineate the clinical, genetic, and morphologic subsets of therapy-related myeloid neoplasia arising after cytotoxic therapy for a primary malignant disease. Pictured: Sonali Smith, MD
Mark Ratain, MD, discovered genetic variants that predict which cancer patients are likely to experience severe side effects from the drug irinotecan and helped launch the field of pharmacogenomics.
Harinder Singh, PhD
Harinder Singh, PhD, mapped the regulatory networks controlling the commitment of hematopoietic stem cells into antibody-producing B cells, leading to insights into B cell tumor development.

2005 - 2009

Walter Stadler, MD, with Mark Ratain, MD, pioneered a clinical trial design (called randomized discontinuation trial) for anticancer agents, allowing for selection and analysis of patient subgroups most likely to benefit from a growth inhibitory treatment.
Marsha Rosner, PhD
Marsha Rosner, PhD, characterized a signaling inhibitor that impacts the mitotic spindle checkpoint and metastasis and is an attractive therapeutic target for cancer.
Habibul Ahsan, MD
Habibul Ahsan, MD, reported that arsenic in drinking water is associated with the risk of premalignant skin lesions, contributing to the EPA lowering the legal limit of arsenic in United States drinking water.
M. Eileen Dolan, PhD, with Nancy Cox, PhD, discovered genetic variants associated with chemotherapy-induced toxicities using novel cell lines from the HapMap Project, data from which they made publicly available to researchers worldwide, and demonstrated that these variants generally alter gene expression.
Michelle Le Beau, PhD, identified the first haploinsufficient (exerts its effect by loss of a single copy) tumor suppressor gene on chromosome 5 – EGR1 – in therapy-related myeloid neoplasms. The group went on to define a novel molecular mechanism by which leukemia with a del(5q) is the result of a “contiguous gene syndrome” arising from the combined loss of multiple haploinsufficient genes.  
Maryellen Giger, PhD, demonstrated that computer-aided diagnosis imaging tools are accurate for detecting breast cancer, starting a field now known as radiomics. In 2019, QuantX™ – developed by Dr. Giger – became the first FDA-cleared, artificial-intelligence-driven technology to assist radiologists in the diagnosis of breast cancer.
James Nachman, MD, and Wendy Stock, MD
Wendy Stock, MD, with James Nachman, MD, showed that adolescent and young adult acute lymphoblastic leukemia patients had better outcomes on pediatric trials compared to adult trials.
Olufunmilayo Olopade, MD, discovered that most breast cancers in women of African ancestry are the aggressive triple-negative subtype and began to identify genetic variants contributing to this disparity.
Susan Cohn, MD
Susan Cohn, MD, led efforts to establish a risk group classification system for neuroblastoma, a common pediatric cancer, and identified factors that contribute to the subgroup differences.
 

2010 - 2014

Chuan He, PhD, with Lucy Godley, MD, PhD, developed the first tool to identify genome-wide changes in the epigenetic modification 5-methylcytosine in cancer.
Kenan Onel, MD, PhD, identified genetic variants associated with radiation therapy-induced secondary cancers. 
Ernst Lengyel, MD, PhD
Ernst Lengyel, MD, PhD, uncovered the importance of the ovarian cancer microenvironment in promoting tumor progression and metastasis and molecular pathways involved, some of which are attractive therapeutic targets.
 
Chun-Su Yuan, MD, PhD, identified the molecular mechanisms responsible for ginseng's chemopreventive activity in colorectal cancer.
Sonali Smith, MD, led a multi-center study that shows hematopoietic cell transplantation is beneficial as an early therapy for T-cell non-Hodgkin lymphoma.
 
Megan McNerney, MD, PhD, and Kevin White, PhD
Megan McNerney, MD, PhD, and Kevin White, PhD, cloned CUX1, a long-sought tumor suppressor gene on chromosome 7 frequently inactivated in acute myeloid leukemia.
Yusuke Nakamura, MD, PhD,
Yusuke Nakamura, MD, PhD, developed a novel anti-cancer therapy targeting the T-lymphokine-activated killer cell-originated protein kinase (TOPK) and demonstrated potent activity in preclinical lung cancer studies.
 

2015 - 2019

Chuan He, PhD, developed methods to investigate mRNA epitranscriptomic modifications, leading to understanding of their role in mRNA stability and gene expression, and demonstrated their role in oncogenesis.

Thomas Gajewski, MD, PhD, demonstrated the role of β-catenin signaling in tumor immune exclusion and PD1 checkpoint inhibitor therapy resistance, showing that the T cell-inflamed tumor microenvironment is a biomarker of response to immunotherapy.

Suzanne Conzen, MD, discovered a role for the stress hormone, glucocorticoid, as an alternative biological pathway used by breast and prostate cancer cells to escape dependence on estrogen or testosterone.
Rita Nanda, MD
Rita Nanda, MD, led a clinical study showing that checkpoint inhibitors are an effective therapy for treating the aggressive triple-negative breast cancer.
Ralph Weichselbaum, MD
Wenbin Lin, PhD, and Ralph Weichselbaum, MD, developed advanced nanoscale metal-organic frameworks that deliver chemotherapy and photodynamic drugs designed to synergize with radiotherapy and immunotherapy. Pictured: Ralph Weichselbaum, MD
Richard Larson, MD
Richard Larson, MD, demonstrated that the addition of the drug midostaurin to the standard of care for acute myeloid leukemia (AML) therapy, improved survival in patients with FLT3 mutations, strengthening targeted therapeutic approaches for AML.
 
Tara Henderson, MD, MPH, championed novel approaches to childhood cancer survivorship care to address the needs of this growing population and improve their long-term care.
 
Hedy Kindler, MD
Hedy Kindler, MD, discovered that PARP inhibition successfully stopped disease progression for metastatic pancreatic cancer in patients with BRCA1/2 mutations. 
Mark Ratain, MD, and Russell Szmulewitz, MD, found that taking a lower dose of the expensive prostate cancer drug abiraterone with food lowers costs, addressing financial toxicity, or the economic burden placed on patients by the high cost of care.

2020 -

Karen Kim, MD, developed novel approaches to implement colon cancer screening in an effort to address disparities and increase use of the live-saving intervention.
 
Eric Pamer, MD
Eric Pamer, MD, identified patterns of microbiota disruption that occur in cancer patients during hematopoietic-cell transplant and determined how to use these patterns as predictors of outcome following transplantation.