The Atlanta Journal-Constitution

‘Living drugs’ latest in cancer treatment

New type of immune therapy shows promise.

- By Lauran Neergaard

SEATTLE — Ken Shefveland’s body was swollen with cancer, treatment after treatment failing until doctors gambled on a radical approach: They removed some of his immune cells, engineered them into cancer assassins and unleashed them into his bloodstrea­m.

Immune therapy is the hottest trend in cancer care, and this is its next frontier — creating “living drugs” that grow inside the body into an army that seeks and destroys tumors.

Looking in the mirror, Shefveland saw “the cancer was just melting away.” A month later doctors at the Fred Hutchinson Cancer Research Center couldn’t find any signs of lymphoma in the Vancouver, Washington, man’s body.

“Today I find out I’m in full remission — how wonderful is that?” said Shefveland with a wide grin, giving his physician a quick embrace.

This experiment­al therapy marks an entirely new way to treat cancer — if scientists can make it work safely. Early-stage studies are stirring hope as one-time infusions of supercharg­ed immune cells help a remarkable number of patients with intractabl­e leukemia or lymphoma.

“It shows the unbelievab­le power of your immune system,” said Dr. David Maloney, Fred Hutch’s medical director for cellular immunother­apy who treated Shefveland with a type called CAR-T cells.

“We’re talking, really, patients who have no other options, and we’re seeing tumors and leukemias disappear over weeks,” added immunother­apy scientific director Dr. Stanley Riddell. But, “there’s still lots to learn,” he said.

T cells are key immune system soldiers. But cancer can be hard for them to spot, and it can put the brakes on an immune attack. Today’s popular immunother­apy drugs, called “checkpoint inhibitors,” release one brake so nearby T cells can strike. The new cellular immunother­apy approach aims to be more potent: Give patients stronger T cells to begin with.

Currently available only in studies at major cancer centers, the first CAR-T cell therapies for a few blood cancers could hit the market later this year.

The Food and Drug Administra­tion is evaluating one version developed by the University of Pennsylvan­ia and licensed to Novartis, and another created by the National Cancer Institute and licensed to Kite Pharma.

CAR-T therapy “feels very much like it’s ready for prime time” for advanced blood cancers, said Dr. Nick Haining of the Dana-Farber Cancer Institute and Broad Institute of MIT and Harvard, who isn’t involved in the developmen­t.

Now scientists are tackling a tougher next step, what Haining calls “the acid test”: Making T cells target more common cancers — solid tumors like lung, breast or brain cancer. Cancer kills about 600,000 Americans a year, including nearly 45,000 from leukemia and lymphoma.

“There’s a desperate need,” said NCI immunother­apy pioneer Dr. Steven Rosenberg, pointing to queries from hundreds of patients for studies that accept only a few.

No magic bullet, but exciting possibilit­ies

For all the excitement, there are formidable challenges.

Scientists still are unraveling why these living cancer drugs work for some people and not others.

Doctors must learn to manage potentiall­y life-threatenin­g side effects from an overstimul­ated immune system.

Also concerning is a small number of deaths from brain swelling, an unexplaine­d complicati­on that forced another company, Juno Therapeuti­cs, to halt developmen­t of one CAR-T in its pipeline.

And, made from scratch for every patient using their own blood, this is one of the most customized therapies ever and could cost hundreds of thousands of dollars.

“It’s a Model A Ford and we need a Lamborghin­i,” said CAR-T researcher Dr. Renier Brentjens of New York’s Memorial Sloan Kettering Cancer Center, which, like Hutch, has a partnershi­p with Juno.

In Seattle, Fred Hutch offered a behind-the-scenes peek at research underway to tackle those challenges. At a recently opened immunother­apy clinic, scientists are taking newly designed T cells from the lab to the patient and back again to tease out what works best.

“We can essentiall­y make a cell do things it wasn’t programmed to do naturally,” explained immunology chief Dr. Philip Greenberg.

“Your imaginatio­n can run wild with how you can engineer cells to function better,” he said.

Two long weeks to brew a dose

The first step is much like donating blood. When leukemia patient Claude Bannick entered a Hutch CAR-T study in 2014, nurses hooked him to a machine that filtered out his white blood cells, including the T cells.

Technician­s raced his bag of cells to a factory-like facility that’s kept so sterile they must pull on germ-deflecting suits, booties and masks just to enter. Then came 14 days of wait and worry, as his cells were reprogramm­ed.

Bannick, 67, says he “was almost dead.” Chemothera­py, experiment­al drugs, even a bone-marrow transplant had failed, and “I was willing to try anything.”

The goal: Arm T cells with an artificial receptor, a tracking system that can zero in on identifyin­g markers of cancer cells, known as antigens. For many leukemias and lymphomas, that’s an antigen named CD19.

Every research group has its own recipe, but generally, scientists infect T cells with an inactive virus carrying genetic instructio­ns to grow the desired “chimeric antigen receptor.” That CAR will bind to its target cancer cells and rev up for attack.

Millions of copies of engineered cells are grown in incubators. If they work, those cells will keep multiplyin­g in the body. If they don’t, the doctors send blood and other samples back to researcher­s to figure out why.

What’s the data?

Small, early studies in the U.S. made headlines as 60 percent to 90 percent of patients trying CAR-Ts as a last resort for leukemia or lymphoma saw their cancer rapidly decrease or even become undetectab­le.

Last week, Chinese researcher­s reported similar early findings as 33 of 35 patients with another blood cancer, multiple myeloma, reached some degree of remission within two months.

Too few people have been studied so far to know how long such responses will last. A recent review reported up to half of leukemia and lymphoma patients may relapse.

There are long-term survivors. Doug Olson received the University of Pennsylvan­ia’s CAR-T version for leukemia in 2010. The researcher­s were frank — it had worked in mice but they didn’t know what would happen to him.

“Sitting here almost seven years later, I can tell you it works,” Olson, now 70, told a recent meeting of the Leukemia and Lymphoma Society. Bannick, the Hutch patient treated in 2014, recalls Maloney calling him “the miracle man.” He had some lingering side effects that required blood-boosting infusions but says CAR-T is “giving me a second life.”

Scary side effects

“The more side effects you have, that sort of tells everybody it’s working,” said Shefveland, who was hospitaliz­ed soon after his treatment at Hutch when his blood pressure collapsed. His last clear memory for days: “I was having a conversati­on with a nurse and all of a sudden it was gibberish.”

As CAR-T cells swarm the cancer, an immune overreacti­on called “cytokine release syndrome” can trigger high fevers and plummeting blood pressure and in severe cases organ damage. Some patients also experience confusion, hallucinat­ions or other neurologic symptoms.

Treatment is a balancing act to control those symptoms without shutting down the cancer attack.

Experience­d cancer centers have learned to expect and watch for these problems. “And, most importantl­y, we’ve learned how to treat them,” said Dr. Len Lichtenfel­d of the American Cancer Society, who is watching CAR-T’s developmen­t.

Fighting solid tumors will be harder

CAR-Ts cause collateral damage, killing some healthy white blood cells, called B cells, along with cancerous ones because both harbor the same marker. Finding the right target to kill solid tumors but not healthy organ tissue will be even more complicate­d.

“You can live without some normal B cells. You can’t live without your lungs,” Riddell explained.

Early studies against solid tumors are beginning, targeting different antigens. Timelapse photos taken through a microscope in Riddell’s lab show those new CAR-T cells crawling over aggressive breast cancer, releasing toxic chemicals until tumor cells shrivel and die.

CARs aren’t the only approach. Researcher­s also are trying to target markers inside tumor cells rather than on the surface, or even gene mutations that don’t form in healthy tissue.

How will patients get the first CAR-T therapies?

If the FDA approves Novartis’ or Kite’s versions, eligible leukemia and lymphoma patients would be treated at cancer centers experience­d with this tricky therapy. Their T cells would be shipped to company factories, engineered, and shipped back. Gradually, more hospitals could offer it.

Because only certain patients would qualify for the first drugs, others would have to search for CAR-T studies to try the treatment.

 ?? ELAINE THOMPSON / AP ?? Dr. Stanley Riddell describes how a patient’s large tumor rapidly shrank in an immunother­apy study at the Fred Hutchinson Cancer Research Center in Seattle. Riddell’s team studies “living drugs” that are geneticall­y engineered from patients’ own immune cells.
ELAINE THOMPSON / AP Dr. Stanley Riddell describes how a patient’s large tumor rapidly shrank in an immunother­apy study at the Fred Hutchinson Cancer Research Center in Seattle. Riddell’s team studies “living drugs” that are geneticall­y engineered from patients’ own immune cells.

Newspapers in English

Newspapers from United States