ASH Clinical News ACN_5.7_Digital | Page 46

Drawing First Blood sibling donor, we strongly recommend that approach. However, even if there is a perfectly matched sibling, there remains a 3% to 5% risk of transplant-related mortality. For most parents of young children with beta-thalassemia, the only acceptable number is zero percent. Adequate management with transfusion and iron control gives the patient and his family the time and opportunity to discuss options. Dr. Thompson: Any of these cures has a cost. In addition to the risk of transplant-related mortality, all curative approaches carry the risk of infertility. No matter what kind of transplant – gene therapy, alloHCT, or autologous hematopoietic cell trans- plantation – infertility is still a burden we are not able to alleviate. I am encouraged by some of the progress in fertility cryopreservation in young children, but this is still experimental. In our experience, if fertility is a high priority, parents of children with sibling matches have deferred transplant to ensure that they allow the child time to participate in that decision. Dr. Coates: Another concern about gene therapy are the less-exciting outcomes seen in patients with the ß 0 genotype, compared with those in patients with the ß + genotype. We also have seen that some patients who receive gene therapy are developing thalassemia intermedia instead, when only sporadic or no transfusions are required. These represent some of the most complicated patients to follow, even for physicians with extensive experience in managing thalassemias. If patients undergo gene therapy and their hemoglobin levels are at 11 or 12 g/dL, I anticipate gene therapy will continue to provide long-term clinical benefit; however, if patients undergo gene therapy and their hemoglobin levels linger at 8 or 9 g/dL, they might develop the same long-term complications seen with thalassemia intermedia. In contrast, after a sibling or unrelated donor alloHCT, patients essentially no longer have thalassemia. Dr. Thompson: We have managed a fair number of patients who have thalassemia intermedia, and I am struck by how severely their quality of life worsens over time compared with some of our patients with transfusion-dependent thalassemia – irrespective of transplantation. As we look to improve gene therapy, it must be with the intent to normalize hemoglobin and iron homeostasis. Without achieving those goals, we may see im- provements in terms of scientific measures, but we will fall short in terms of clinical improvement. Dr. Coates: Thalassemia intermedia brings me to another topic: the current enthusiasm for luspa- tercept. 3 This drug is thought to work by inhibit- ing ineffective erythropoiesis. The BELIEVE trial showed that it was efficacious in transfusion- dependent patients, meeting all major transfusion burden–related endpoints. Data from the pre- liminary studies in non–transfusion-dependent patients also are quite exciting, showing that, while the approach did not cure the disease, it markedly improved disease burden – particularly in those with hemoglobin levels of 8 to 9 g/dL. 4 Again, this is a huge problem in the U.S. because patients with non–transfusion-dependent beta- thalassemia are often not recognized as having 44 ASH Clinical News thalassemia by many hematologists or general prac- titioners. Luspatercept will probably be an exciting option for them, but I wonder what effect luspater- cept would have on patients with hemoglobin levels of 8 or 9 g/dL who have received gene therapy. Dr. Thompson: That is an intriguing idea. It could very well be that a treatment like luspatercept could be added post–gene-therapy infusion to increase hemoglobin levels and perhaps eliminate the need for transfusion. It’s hard not to be thrilled with the early out- comes of gene therapy, yet the approach clearly is not for every patient with transfusion-dependent thalassemia. Certain patients feel that there are still too many questions about gene therapy, and they are concerned about the lifestyle changes neces- sary to undergo this procedure. I can imagine that a patient who is finishing college, has a first job, or is raising a young family might not want to put his or her life on hold for even two or three months to undergo a potential curative therapy for which we don’t have long-term data. Luspatercept, understandably, would be more appealing for these types of patients: We could give them an injection when they come in for a transfusion and minimize or eliminate their need for future transfusions, and they can continue with their lives as usual. Dr. Coates: When we talk about potentially curing thalassemia, we also must consider the other option: long-term maintenance. Again, I come back to the issue of the availability of physicians who under- stand how to manage thalassemia because it is such a rare disease. At our center, we are aggressive with transfu- sions, so many patients receiving transfusions actually have beta-thalassemia intermedia and would be considered by other centers to have non–transfusion-dependent disease. We see few complications when patients’ iron levels are controlled. We also have to consider that, as patients with beta-thalassemia intermedia age, they do not tolerate their anemia as well and later become transfusion dependent. Luspatercept will prob- ably help a substantial portion of these “borderline transfusion-dependent” patients, without the need for transfusion. Therefore, luspatercept is going to make the prospect of a non-transplant option even more attractive to certain patients. In clinical trials, the drug was shown to decrease RBC transfusion requirements by approximately 30%, including among patients with thalassemia intermedia and the β 0 genotype. My hope is that luspatercept will allow us to achieve the same degree of hemoglobin increase with less transfusion, which would make the non-transplant approach even more palatable for patients with thalassemia intermedia. Dr. Thompson: This is a remarkable time in the treatment of thalassemias. We have made it over the big hurdles of providing safe transfusions, monitoring iron noninvasively, and developing more convenient chelators, but I can’t help but think there is room for other advances for patients with thalassemia. For example, the newer oral che- lators have represented such an improvement over the onerous medications like deferoxamine, which required nightly subcutaneous infusion. Given the dosing schedules and side-effect profiles of the oral options, though, compliance and adherence are still issues. Unfortunately, we do not have an ideal chelator for some patients. Similarly, we don’t yet know whether there is a role for drugs like hepci- din to help with balancing iron load in patients. The trials of gene-editing in thalassemia also are just preparing to launch. Dr. Coates: The major challenge to overcome with chelation therapy is finding a chelator that can be administered in a less-frequent schedule. Of course, all these medications have the same fatal flaw: They don’t work if you don’t take them. The beauty of transplant is that, once a patient gets through the process, the daily drug therapy falls away. Dr. Thompson: With gene therapy, at least in the current research setting, patients must adhere to a certain regimen postinfusion, but for a fairly fixed period. Many patients will need to resume iron chelation to fully unload residual tissue iron after gene therapy, but most will not need long-term medications. They will require close follow-up at a specialized center postinfusion, then less frequent monitoring after a few years. Compared with the burdens of transfusion, chelation, and management of other thalassemia complications, gene therapy should give most patients remarkable freedom. Dr. Coates: That freedom is also one of the advan- tages of luspatercept; it is an injection administered approximately once every three weeks, so adher- ence should be a bit higher. Iron is the factor I am always at the pulpit about. Pediatric patients who undergo transplant are now expected to survive for decades, so my big- gest concern is the long-term consequences of ma- lignant transformation. Research has shown that patients receiving transfusions have an elevated risk of developing leukemia and lymphoma later in life, compared with those who were not transfused. Dr. Thompson: We can both agree that we need long-term follow-up to evaluate late effects of gene therapy, as well as the consequences of pre-existing organ injury from thalassemia. It is easy to talk about the intent of curative therapies, but the question we need to answer is, “Does this person still have thalassemia?” Patients want to consider themselves disease-free, and it is incumbent upon us to remind them that, even after one of these curative approaches, they will need surveillance for a long time. ● REFERENCES 1. Thompson AA, Walters MC, Kwiatkowski J, et al. Gene therapy in patients with transfusion-dependent β-thalassemia. N Engl J Med. 2018;378:1479-93. 2. Locatelli F, Walters MC, Kwiatkowski JL, et al. Lentiglobin gene therapy for patients with transfusion-dependent β-thalassemia (TDT): results from the phase 3 Northstar-2 and Northstar-3 studies. Abstract #1025. Presented at the 2018 ASH Annual Meeting, December 1, 2018; San Diego, CA. 3. Cappellini MD, Viprakasit V, Taher A, et al. The BELIEVE trial: results of a phase 3, randomized, double-blind, placebo-controlled study of luspatercept in adult beta thalassemia patients who require regular red blood cell (RBC) transfusions. Abstract #163. Presented at the 2018 ASH Annual Meeting, December 1, 2018; San Diego, CA. 4. Piga A, Perrotta S, Gamberini MR, et al. Luspatercept improves hemoglobin levels and blood transfusion requirements in a study of patients with β-thalassemia. Blood. 2019;133:1279-89. June 2019