AACR President, 2022-2023
Hildegard Lamfrom Endowed Chair in Basic Science Professor and Chair, Department of Cell, Developmental & Cancer Biology
Associate Director for Basic Research, Knight Cancer Institute
Oregon Health & Science University Portland, Oregon

There has been tremendous progress in our fundamental understanding of cancer biology during my decades-long career as a scientist. Remarkable discoveries in basic research have led to the development of lifesaving treatments for patients with cancer, resulting in a steady reduction in the overall U.S. cancer mortality rate year after year. I am also very optimistic about the future of cancer science and medicine, because of the new wave of innovations that are just over the horizon and are set to fundamentally improve cancer detection and treatment.

One of the most important lessons learned over the past two decades is that cancer is not a single disease, but a collection of over 200 different types of diseases. We have also learned that, even within a single tumor, a multitude of cell types with different mutations exists. This complexity is one of the reasons that cancers are so difficult to treat. The contributions of the tumor microenvironment, which includes the immune system as well as other cells, further increase the complexity of cancers. With the tremendous boom in sequencing technology over the past two decades, we can further understand these complexities using cutting-edge tools, such as tumor microdissection, single cell sequencing, and proteomics. These technologies have led to an explosion of data, necessitating cross-disciplinary approaches that require cancer biologists, bioinformaticians, computational biologists, and chemists working together to identify the cellular and molecular pathways important for cancer pathogenesis. This type of research is essential for investigators to create drugs to target those vulnerabilities and stop cancer in its tracks. I anticipate that the costs of these sequencing technologies will become even more affordable to the point where personalized medicine approaches are applied to every future patient, dramatically improving their treatment options and transforming patient outcomes.

One area of cancer medicine where we have evidenced unprecedented progress is immune oncology, which leverages the power of immune cells to thwart cancer progression and tumor growth. In particular, the area of adoptive cell therapies, such as CAR T-cell therapies, has changed the treatment landscape for patients with certain hematologic malignancies. We are now beginning to appreciate the potential of other immune cells beyond T cells that can be leveraged to fight tumors, such as natural killer cells and macrophages. Many of these therapies are currently being tested in clinical trials and are set to vastly increase the diversity of immunotherapy options for patients within the next decade. Other immune-based therapies, such as immune checkpoint inhibitors, unleash the power of the immune system to fight cancer, and have improved patient outcomes across many types of cancer. It is important to note that every single advance in developing new and effective treatments for cancer has its roots in a basic research discovery. For instance, the development of checkpoint inhibitors is rooted in the study of basic T-cell biology, which led to the knowledge of the molecular pathways important for their cancer-fighting capabilities, paving the way for this class of immunotherapies.

One of the biggest challenges in cancer research and patient care continues to be the lack of diversity in oncology clinical trials and in genetic databases, such as The Cancer Genome Atlas, which predominantly contains samples from patients of European ancestry. It has been only within the last 25 years that we have begun to appreciate how genetic variants found in populations of shared ancestry contribute to cancer incidence and mortality. Biologists, population scientists, and epidemiologists are working together to decode genetic data and address social determinants of health for better understanding and treatment of cancers in diverse groups of people. In addition to racial and ethnic minorities, increasing our knowledge of the challenges faced by other medically underserved populations, such as sexual and gender minorities and those who are socioeconomically deprived, is required for equitable and culturally appropriate cancer care. Increased funding from our government is necessary to better understand these populations and their unique risk factors so that appropriate intervention strategies can be implemented.

Increasing the participation of medically underserved populations in scientific studies and clinical trials demands a more diverse cancer workforce. While we have made some gains in closing the gender gap in medical science, we still lack racial, ethnic, socioeconomic, and geopolitical diversity. Increasing diversity among researchers and clinicians is crucial for expanding the conversation across the cancer continuum, affording unique perspectives to tackle new questions. I am extremely proud that AACR is playing a leading role in crafting solutions to help increase diversity in cancer research and science by improving the gender balance, as well as by identifying the problems surrounding the lack of diversity.

Basic research is integral to understanding why normal cells become cancerous and it requires continued support both from governmental bodies, such as NIH and NCI, and nonprofit and philanthropic organizations, such as AACR. I often tell my students that you cannot understand how to fix something unless you know why it is broken. Such questions are only addressed through basic research, making it vital that we continually increase our investments in this area. It is these investments that have led to remarkable gains in reducing cancer incidence by implementing effective prevention strategies; in lowering the chances of late-stage disease by detecting cancers earlier; and in reducing cancer mortality by improving treatment options.