Skip to content
Parker Institute for Cancer Immunotherapy
Search
Search Close

Five Key Takeaways from the 2021 ASCO Annual Meeting

You could call it the Super Bowl of the cancer world. The American Society of Clinical Oncology Annual Meeting, traditionally drawing tens of thousands of people from all over the world to Chicago, is one of the biggest showcases of the year for the latest and most promising treatments and approaches to curing cancer. The meeting was held virtually once again this year due to the COVID pandemic, and that wasn’t the only impact current events had on the program.

“This year’s meeting not only featured cutting edge research, but also thought-provoking discussions about some of the biggest issues facing global health care systems and new drug development, including how we close the gaps that exist in access to clinical trials and innovative cancer medicines,” said Ute Dugan, MD, PICI’s Chief Medical Officer.

From reducing disparities to expanding immunotherapy options, here are five key takeaways from the 2021 ASCO Annual Meeting.

Diversity, Equity and Inclusion Takes Center Stage

Efforts to boost diversity, equity, inclusion and belonging are rightfully emerging as a key priority for many organizations, and the oncology world is no exception. The theme of this year’s meeting was “Equity: Every Patient. Every Day. Everywhere.” That meant more diversity on panels, greater scrutiny around disparities and a focus on access to care to ensure all cancer patients can benefit from the latest breakthroughs.

Importantly, this year’s meeting also focused on objectively analyzing the areas where the field is making progress, as well as where it’s still falling short. In 2018 and 2019, the U.S. Food and Drug Administration approved 28 new cancer treatments based on trial data from 8,700 patients. FDA reports show just four percent of those patients on study were Black. That’s in line with historical data. According to the FDA, just five percent of Black cancer patients are enrolled in clinical trials.

Researchers from the Abramson Cancer Center of the University of Pennsylvania presented data showing they were able to double the number of Black patients enrolled in their clinical trials. Importantly, they laid out the steps they took to make it happen, including changes in marketing strategies and an increased focus on community relations.

“Aligning the number of Black patients with cancer we care for with the number enrolled in our trials is how we can help bring more equitable care to the community, close gaps in disparities, and sustain trust,” said the study’s senior author Robert Vonderheide, MD, DPhil, director of the Abramson Cancer Center and a PICI member researcher. “There’s more work to be done to improve access and inclusion of minority groups, and the impact of this outreach and engagement effort is an important step forward.”

Progress for New Checkpoint Inhibitor LAG-3

Checkpoint inhibitors, which work by blocking certain signals in the body and allowing the immune system to attack cancer, have been revolutionary in many different disease types. Historically, the two most effective inhibitors have targeted proteins called PD-1 or CTLA-4. This year’s meeting included news of a groundbreaking new checkpoint option targeting a protein called LAG-3.

Bristol Myers Squibb reported data on a Phase 3 trial combining a LAG-3 inhibitor called relatlimab with the PD-1 inhibitor nivolumab in patients with melanoma. The global study found the median progression-free survival of patients who received both drugs was 10 months, compared to just four and a half months for patients who received nivolumab alone.

There were also significantly more serious side effects in the combination arm. The data showed adverse events serious enough to require hospitalization occurred in almost 19 percent of patients on both checkpoint inhibitors, compared to just under 10 percent on nivolumab alone. More patients in the combination arm had to stop treatment because of these issues – 14 percent, compared with less than 7 percent in the nivolumab group.

“This is encouraging progress that can expand immunotherapy treatment options for cancer patients, but there’s still work to do to better understand who is most likely to benefit,” said Lisa Butterfield, PhD, Vice President, Research and Development at PICI.

Drugging the “Undruggable” KRAS Mutation

Certain genetic mutations are known to be associated with cancer growth, which theoretically makes them attractive targets for new therapies. However, designing a treatment that hits its target is a complex challenge. That’s especially true of a gene called KRAS. It’s one of the most common genetic drivers of certain disease types like lung cancer, but it has historically been an evasive target, leading many to refer to it as “undruggable.”

That’s just one of the reasons why new data on a recently approved KRAS inhibitor called sotorasib are so encouraging. A Phase 2 study led by researchers at The University of Texas MD Anderson Cancer Center showed sotorasib, which targets a specific mutation called KRAS-G12C, led to a 37 percent objective response rate and a median overall survival of 12.5 months in non-small cell lung cancer patients who have undergone previous treatments.

“This is the first time a study has shown that a KRAS inhibitor may help cancer patients live longer. It’s especially encouraging in patients who had failed a previous therapy, which is a group of patients who generally have poor outcomes and who need options,” said Marko Spasic, MD, PICI’s Medical Director.

The FDA approved sotorasib in May based on previously reported data.

Moving Immunotherapy Earlier in Treatment

Figuring out who to treat with checkpoint inhibitors and expanding those options is only part of the equation. There’s also a question of timing. In most cases, like the KRAS inhibitor study, patients receive immunotherapy after failing a previous treatment like chemotherapy. But research presented at this year’s meeting showed the potential benefit of using immunotherapy earlier in a patient’s course.

Researchers treated kidney cancer patients with a PD-1 inhibitor following surgery to remove their tumors and found it cut the rate of disease returning or death by almost a third over a two-year period. The study, led by the Dana-Farber Cancer Institute and funded by Merck, was presented as part of the meeting’s plenary session, during which reviewers called it practice changing.

A separate study found adding a PD-1 inhibitor to pre-surgical chemotherapy can enhance responses in esophageal cancer patients. A third showed the benefit of adding PD-1 as a first-line therapy for patients with gastric cancers.

Radioactive Medicine in Prostate Cancer

The fast-growing field of nuclear medicine and cancer therapy saw a groundbreaking presentation at this year’s meeting. A Novartis-funded trial added a radioligand called Lu-PSMA-617, which uses a radioactive molecule that can kill cancer combined with a protein that can guide it toward tumors. That drug was added to standard of care treatments in prostate cancer patients who had exhausted all other treatment options. The combination improved median overall survival from 11 months to 15 months and progression free survival from 3.4 months to 8.7 months.

“This is a group of patients with a common cancer who need more options, and this study shows a potential new way to target their tumors,” said Theresa LaVallee, PhD, Vice President of Translational Medicine and Regulatory Affairs at PICI.

Novartis is studying the drug in other cancer types.

Stay in Touch

Keep up to date with the latest PICI news by signing up to receive email updates.