Cancer Immunotherapies Don’t Work for Everyone: HLA Gene May Explain Why


Hundreds of thousands of people with cancer take immune checkpoint inhibitors, such as nivolumab (Opdivo) and pembrolizumab (Keytruda), every year. But for the majority of those people, the medicines—a type of immunotherapy—don’t work to treat their cancer.

Now NCI scientists think they’ve found a specific form of a gene that may make immune checkpoint inhibitors less effective for some people. The gene form, known as HLA-A*03, is found in 2% to 16% of the US population.

If the findings are confirmed in further studies, doctors could use the gene as a marker to help decide if a patient should or shouldn’t take an immune checkpoint inhibitor, said the scientist who led the study, Mary Carrington, Ph.D., of NCI’s Center for Cancer Research.

That could spare many patients from getting treatments that don’t work for them and that can cause harsh side effects, Dr. Carrington added. But, she cautioned, “we need more data before [doctors would] actually use that [marker] in the clinic.”

In their analysis of data from thousands of patients, the researchers found that people with HLA-A*03 died sooner after immune checkpoint inhibitor treatment than people with other forms of the HLA-A gene. The study, published December 9 in Lancet Oncology, showed that this pattern held true for different immune checkpoint inhibitors and different types of cancer, including kidney, bladder, and skin cancer.

Timothy Chan, M.D., Ph.D., director of the Cleveland Clinic’s Center for Immunotherapy and Precision Immuno-Oncology, called the findings “exciting.” The next step, continued Dr. Chan, who was not involved in the study, is “to work out how [A*03] can be used most efficiently with other biomarkers” that are currently used to predict how well checkpoint inhibitors will work. 

Different forms of HLA genes

HLA-A is a member of a set of genes—including HLA-B and HLA-C—that help the immune system find and destroy cells that are cancerous or infected with a virus or bacteria.

These HLA genes make proteins called human leukocyte antigens (HLA), which take bits and pieces of proteins from inside the cell and display them on the cell’s surface. If the cell is cancerous or infected, the HLA proteins display abnormal fragments that trigger immune cells to destroy that cell and any others displaying the same fragment.

Across the human population, “there are thousands of different forms of these HLA genes, which we call alleles,” explained Dr. Carrington, who leads NCI’s HLA immunogenetics research. 

A*03, for instance, is one of more than 2,000 alleles of HLA-A. That natural variation in HLA genes is, in part, what makes some people more susceptible than others to certain viral infections and autoimmune diseases. 

Because immune checkpoint inhibitors help immune cells find and attack cancer cells, scientists have long suspected that variations in HLA genes may influence how well these treatments work.

Possible links between various HLA alleles and immune checkpoint inhibitor effectiveness have been studied extensively. But, so far, A*03 is the only one that seems to work as a marker of treatment response for multiple types of cancer and different immune checkpoint inhibitors, Dr. Carrington noted. 

People with A*03 benefit less from immunotherapy

The team first sifted through data from a group of more than 1,000 people with 30 types of advanced cancer who were treated with an immune checkpoint inhibitor at Memorial Sloan Kettering Cancer Center. 

Among various HLA alleles, HLA-A*03 emerged as the strongest biomarker of immune checkpoint inhibitor effectiveness. People with A*03, regardless of what kind of cancer they had, died nearly 1.5 times sooner after starting treatment than those with a different allele, the researchers found. 

When the researchers separated the data by cancer type, A*03 was linked with a shorter time to death for people with bladder, brain (glioma), melanoma, lung, and kidney cancer. However, the impact of having A*03 was greatest in people with kidney cancer. 

Among participants of four clinical studies who received an immune checkpoint inhibitor for kidney cancer, those with A*03 didn’t live as long before their kidney cancer grew back or they died (progression-free survival), compared with those who didn’t have the allele.

Confirming the findings in randomized clinical trials is very good evidence that A*03 is a biomarker of immune checkpoint inhibitor effectiveness, Dr. Chan noted.

Dr. Carrington and her team also found that people with two copies of A*03 died sooner than people with one copy of A*03 plus a copy of a different HLA-A allele (2.3 and 1.5 times sooner, respectively, than people with no copies of A*03).

The researchers validated their findings in two independent groups of patients who had been treated with immune checkpoint inhibitors—1,326 patients with various types of cancer treated at the Dana-Farber Cancer Institute and 169 people with bladder cancer who had participated in an international clinical trial. In both groups, those with A*03 died sooner after starting treatment than those without this form of the gene (1.2 and 1.4 times sooner, respectively).

The link between A*03 and immune checkpoint inhibitor effectiveness held true regardless of a person’s age, sex, ancestry, the type of immune checkpoint inhibitor they took, and whether they got chemotherapy at the same time. However, the allele wasn’t linked with the outcomes of other kinds of cancer treatment.

Combining biomarkers for immunotherapy

“While data from multiple clinical trials consistently showed the association of HLA-A*03 with less clinical benefit in patients treated with immunotherapy, these results should be validated in a prospective clinical study,” noted James Gulley, M.D., Ph.D., of NCI’s Center for Cancer Research and a leader of the study.

Testing for a specific HLA allele can be fast and reliable, the researchers noted. It’s done routinely to match donors and recipients for organ transplants, and as a way to tell who might have life-threatening allergic reactions to certain HIV and epilepsy treatments.

And HLA genes are included in several genetic biomarker tests that are commonly given to people with cancer, Dr. Chan noted. 

But, Dr. Carrington cautioned, just because someone has A*03 doesn’t necessarily mean that they won’t benefit from immune checkpoint inhibitors.

In all of the data the researchers looked at, there were some people with A*03 for whom immune checkpoint inhibitor treatment worked. “It’s when we look at the entire population of [people with] A*03 that we see, overall, there’s less benefit,” she said. 

Ultimately, models that combine multiple biomarkers might better predict whether an immune checkpoint inhibitor is likely to work for an individual, Dr. Chan said. “That’s where this [field] is headed,” he added.

“Immunity is complex, and we know HLA is one part of the story,” he said. But many factors, from the total number of mutations in the tumor (tumor mutational burden) to the microbes that live in the gut, also influence how well immune checkpoint inhibitors work. And “it’s clear that all these things work together,” Dr. Chan explained. 

In a recent study, he and his colleagues built a model that takes into account a person’s genetics, biology, age, and tumor stage, among other factors, to predict whether checkpoint inhibitors are likely to work. The combination model was more accurate than tumor mutational burden alone at predicting whether a checkpoint inhibitor would be effective, the researchers found.

As for Dr. Carrington, her group is focused on understanding why immune checkpoint inhibitors don’t work well for people with A*03. They are currently sifting through several plausible explanations, but none has surfaced as the winner yet. 

“We’ve been ruling things out one after another,” she said. But that’s the exciting part of science, she added, figuring out a great unknown.

Green Light for Cancer Immunotherapies in Europe


The European Medicines Agency (EMA) has recommended granting marketing authorization for three cancer immunotherapies ― pembrolizumab (Keytruda, Merck & Co, Inc) for melanoma, nivolumab (Opdivo, Bristol-Myers Squibb Company) for lung cancer, and dinutuximab (Unituxin, United Therapeutics Corporation) for neuroblastoma.

In addition, the EMA recommended approval for the additional indication of Waldenstrӧm’s macroglobulinaemia for ibrutinib (Imbruvica, Pharmacyclics, Inc, Janssen Biotech, Inc) and also for a generic version of bortezomib (Bortezomib Accord, Accord Healthcare) for use in the treatment of multiple myeloma and mantle cell lymphoma.

Pembrolizumab for Advanced Melanoma

Pembrolizumab will be the second drug that acts as a programmed death inhibitor for the treatment of advanced melanoma in Europe, joining nivolumab, which was recommended for approval for this indication just a few weeks ago. Both drugs are already approved in the United States for use in advanced melanoma.

The data supporting the recommendation for approval come from one uncontrolled study and early results from two ongoing randomized, controlled trials, one comparing pembrolizumab with standard chemotherapy, and the other comparing pembrolizumab with ipilimumab (Yervoy, Bristol-Myers Squibb Company), another immunotherapy but with a slightly different mechanism of action.

The Committee for Medicinal Products for Human Use (CHMP) says the data so far have demonstrated the efficacy of pembrolizumab in adults with unresectable or metastatic melanoma, both in patients who had and in those who had not previously received ipilimumab. The committee also looked at safety information from more than 1000 patients enrolled in clinical studies and regarded the safety profile to be manageable.

Nivolumab for Lung Cancer

Nivolumb was recommended for approval for use in the treatment of squamous non–small cell lung cancer (NSCLC) when the disease is advanced and the patient has already been treated with chemotherapy. It is the first immunotherapy to be approved for use in lung cancer, and was welcomed by experts when this approval was announced in the United States.

In Europe, for this indication the product will have the trade name Nivolumab BMS, whereas for the treatment of melanoma, it will be marketed as Opvido, the tradename used in other countries.

The CHMP notes that this recommendation for approval is based on data from a phase 3 trial in 272 patients with squamous NSCLC in whom chemotherapy had failed. These patients were randomly assigned to receive either nivolumab or chemotherapy with docetaxel. This study found that nivolumab improved overall survival compared with docetaxel (median, 9.2 months, compared with 6.0 months). After 12 months, 42% of patients treated with nivolumab were still alive compared with 24% of patients treated with docetaxel, the CHMP notes. Details of this study are due to be presented next week at the annual meeting American Society of Clinical Oncology.

There were also further data from an uncontrolled study involving 117 patients with squamous NSCLC who had undergone at least two previous chemotherapy treatments and who were then treated with nivolumab, the CHMP noted.

Dinutuximab for Neuroblastoma

Dinutuximab was recommended for approval for use in the treatment of high-risk neuroblastoma in children who had already responded to an induction treatment with chemotherapy, followed by myeloablative therapy and autologous stem-cell transplant. The product should be administered in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-2 (IL-2), and isotretinoin.

The product was recently approved in the United States.

Neuroblastoma is a rare cancer, so dinutuximab has orphan drug designation for this indication. The tumor forms from immature nerve cells and is usually seen as a lump in the abdomen or around the spine. It typically occurs in children younger than 5 years, the EMA explained in a press release. In many cases, it is present at birth, but the diagnosis is made only later, when the cancer has already spread to other parts of the body, and the child begins to show symptoms of the disease.

Dinutuximab is a monoclonal antibody designed to recognize and attach to disialoganglioside (GD2), an antigen that is present in high amounts on the surface of neuroblastoma cells but in lower amounts in normal cells. When the product attaches to the neuroblastoma cells, it marks them as targets for the body’s immune system, which is then expected to attack the cancer cells and thereby reverse or slow down the progression of the disease, the EMA explained.

The recommendation for approval was based on data from a clinical trial in children with high-risk neuroblastoma who had already responded to chemotherapy (showing at least a partial response) and were further treated with myeloablative therapy and autologous stem-cell transplant. The study randomly assigned 230 patients to receive dinutuximab combined with GM-CSF and IL-2 and oral isotretinoin, or isotretinoin alone.

After 2 years, 66% of patients receiving the dinutuximab combination were alive and free from recurrence or tumor growth, compared with 48% of patients treated with isotretinoin alone.

Dinutuximab “provides a much-needed treatment option to prolong survival” of patients who are considered to have high-risk neuroblastoma, the agency commented.

The most common side effects of dinutuximab are pain, allergic reactions, and hypotension. Because the protein targeted by the product is also present on normal nerve cells, its use may cause irritation and severe pain of the nerve cells, so pain relief is recommended both before and during treatment. Despite prophylaxis, two thirds of children experience pain, and about 40% experience severe pain.

The CHMP recommended that the safety profile be further assessed post authorization, and the agency required the company to include this in their risk management plan for dinutuximab.

Ibrutinib for Rare Lymphoma

An indication extension was recommended for ibutinib (Imbruvica, Pharmacyclics, Inc, Janssen Biotech, Inc). This drug is already approved for use in chronic lymphocytic leukemia and mantle cell lymphoma. The new indication covers its use in Waldenstrӧm’s macroglobulinemia (also known as lymphoplasmacytic lymphoma), a type of non-Hodgkin’s lymphoma. Ibrutinib will be the first drug for the treatment of this rare blood cancer. It has orphan drug designation for this indication.

Waldenstrӧm’s macroglobulinemia is characterized by an excess of abnormal B lymphocytes and plasma cells in the bone marrow and sometimes in other organs, the EMA explains. These abnormal cells produce large amounts of an immunoglobulin M (IgM), which can make the blood thicker than normal. This cancer usually begins in people older than 60 years. Five years after diagnosis, between 36% and 87% of patients are still alive, depending on their individual risk factors.

Ibrutinib offers a novel strategy in the treatment of malignancies involving B lymphocytes. It acts as an inhibitor of Bruton’s tyrosine kinase (Btk), which has a key role in the survival of B lymphocytes and their migration to the organs where these cells normally divide. By blocking Btk, ibrutinib decreases survival and migration of B lymphocytes, thereby delaying the progression of the cancer, the agency explains.

The recommendation for approval is based on the results of a phase 2 study in 63 patients with previously treated Waldenstrӧm’s macroglobulinemia. Around 90% of the patients treated with ibrutinib responded positively to the treatment, and approximately 80% of patients were alive without disease progression after 18 months.

The adverse events reported during the clinical trial were similar to those observed in the already approved indications of ibrutinib, the agency noted. They include neutropenia and thrombocytopenia.

Generic Bortezomib for Multiple Myeloma

In addition, the EMA recommended for approval a generic version of bortezomib (Bortezomib Accord, Accord Healthcare) for the treatment of multiple myeloma and mantle cell lymphoma.

Bortezomib Accord is a generic version of Velcade (Takeda/Millennium), which has been authorized for use in the European Union since April 2004, the agency noted. “Studies have demonstrated the satisfactory quality,” it added, stating that because Bortezomib Accord is administered intravenously and is 100% bioavailable, a bioequivalence study vs the reference product Velcade was not required.