The treatment of older patients with acute lymphoblastic leukemia (ALL) still represents an unmet medical need. Here we report the results of a chemoimmunotherapy approach combining vincristine/dexamethasone and epratuzumab, a humanized monoclonal therapeutic antibody against CD22, in patients over 55 years of age with relapsed/refractory CD22 B-ALL. This approach shows low toxicity and some activity in this patient population.
The main goal in relapsed ALL patients is to obtain a new complete remission (CR) to subsequently allow allogeneic stem cell transplantation (allo-SCT), which is the best prospect for cure. Undoubtedly, this strategy only applies to younger fit patients who can receive aggressive salvage regimens.1 Due to a higher incidence of poor prognostic factors, older ALL patients (generally considered as over 55–65 years old, depending on trials) have a significantly lower CR rate, much earlier mortality, a higher relapse rate, and a poorer survival compared with younger patients.2 Progress in terms of treatment optimization is limited and there is a lack of clinical trials in this population. Since almost all BALL cells express the CD22 surface antigen, it represents a good target for immunotherapy.43 As such, the anti-CD22 humanized antibody, epratuzumab (Immunomedics, Inc., Morris Plains, NJ, USA), which has been studied extensively in NHL,65 is under active investigation in younger adults and children with B-ALL.87
Here, we have evaluated the addition of epratuzumab (hLL2, 360 mg/m/d i.v. on days 1, 8, 15, and 22) to the combination of vincristine (2 mg i.v. on days 1, 8, 15 and 22) and dexamethasone (40 mg p.o. on days 1, 8, 15, and 22) in patients over 55 years of age with relapsed/refractory CD22 B-ALL. Dexamethasone was administered before epratuzumab administration + paracetamol 1 g as prophylaxis in order to prevent epratuzumab infusion reactions. Also, intrathecal injections of methotrexate 15 mg, aracytine 40 mg, and depomedrol 40 mg were recommended on days 1 and 8. This prospective phase II study was conducted at six French centers and was approved by the Brest ethical committee and the clinical trial department of the “Agence Française de Sécurité Sanitaire des Produits de Santé” (AFSSAPS) (clinicaltrials.gov identifier 01219816). Epratuzumab was supplied by Immunomedics Inc. (Morris Plains, New Jersey, USA). Written informed consent was obtained from each patient. Eligibility criteria were: age over 55 years, B-ALL with 20% or more of blasts in the bone marrow (BM), CD22+ expression on 30% or more of the blast population, refractory B-ALL defined by treatment failure after two successive courses of induction therapy or relapse less than six months after first CR, first relapse or beyond, patients relapsed or refractory to at least one 2nd-generation tyrosine kinase inhibitor for Philadelphia positive (Ph) B-ALL, performance status ECOG 0–2, creatinine clearance 50 mL/min or over, and serum bilirubin 30 μmol/L or under. The main objective of the study was to evaluate the CR rate defined by patients reaching CR + CR without platelet recovery (CRi). CR was defined as less than 5% marrow blasts, neutrophil count 1×10/L or over, platelet count 100×10/L or over, and no evidence of extramedullary disease. CRi was defined similarly but without recovery of platelet counts. Partial response (PR) was defined as a more than 50% decrease in blasts in the bone marrow. Responses were evaluated between four and six weeks post-salvage regimen. Responding patients (CR, CRi or PR) were allowed to receive a second cycle as consolidation. Toxicity was evaluated according to the National Cancer Institute Common Toxicity Criteria (NCI-CTC) version 4. Cytological, phenotypic, karyotype and BCR-ABL1 (for PhB-ALL) molecular analyses were performed on blood samples and/or BM aspirates according to standard methods.
Between November 2010 and December 2013, 26 patients were enrolled. One case was excluded because of progression before receiving the treatment, while 2 younger patients were inappropriately included (a 49-year old female in fourth relapse and a 32-year old female with refractory second relapse). All the 25 treated patients were considered for analysis. Patients’ characteristics are shown in Table 1. The overall response rate was 40%, including 4 CR + one CRi (20%) and 5 PR (20%). Patients obtaining CR/CRi included 2 cases with normal karyotype, 2 with Ph B-ALL (1 in first relapse and 1 in third relapse), and one case with hypodiploidy. Regarding the 6 Ph B-ALL patients, 2 obtained a CR and one a PR. The patient with MLL-rearranged B-ALL obtained a PR. There was no difference between the median percentage of BM blasts at inclusion between responders (CR/PR: 70.5%) versus non-responders (median 72%). The 2 younger patients inappropriately included showed no response. All patients in CR/CRi and one patient in PR received a second cycle as consolidation. None of the responders was consolidated by allo-SCT. Minimal residual disease (MRD) evaluated by cytometry in CR/CRi patients was undetectable in 2, and estimated at 5×10, 7×10 and 1.2% and 1.2% for the 3 other responding patients, respectively (Figure 1). Two patients died during treatment due to progression of the disease. The median overall survival (OS) was four months (range: 0.5–43), with a 1-year OS estimated as 13+7% (Figure 2). Median leukemia-free survival for patients achieving CR/CRi was 3.8 months (range: 1–8). One of the non-responding patients had an unexpected CR after receiving rituximab/6-mercaptopurine/methotrexate therapy,9 and is currently alive at three years after allo-SCT. At time of analysis, all patients have died of progression, except 2 non-responding cases.
Salvage regimen was generally tolerated well, since the large majority of grade 3/4 toxicities were expected pancytopenia. One grade 3 allergic toxicity was related to the first epratuzumab infusion, but the patient received the three other infusions without events. One grade 3 renal toxicity and one grade 4 hypertriglyceridemia of unclear etiology were also documented.
This innovative study reports the results of a chemoimmunotherapy approach with epratuzumab, an anti-CD22 humanized antibody, in the setting of older patients with BALL. The results, although modest, are encouraging because the patients studied here represented a very high-risk refractory/relapsed older patient population. These results also suggest that older patients may be re-treated even after relapse or with refractory disease. This is of particular interest in case of donor availability. Indeed, allo-SCT remains the best consolidation for patients achieving CR2,1 while reduced-intensity conditioning regimens now allow this cellular therapy to be performed in patients up to 70–75 years of age.9 Unfortunately, none of the responders here could be consolidated by an allo-SCT; this was not because of their age (4 out of 5 were under 70 years old), but because they relapsed shortly after the second chemoimmunotherapy cycle. Thus, proceeding straight to transplant as soon as CR2 is achieved might be a better approach to obtain long-term remission in these patients.
Considering epratuzumab, it remains questionable whether this antibody improves the results of the chemotherapy. To our knowledge, there is no series reporting the results of a combination of vincristine/corticosteroids in an older relapsed/refractory B-ALL population, although this remains probably one of the most routinely mild re-induction chemotherapies used in this setting. In first-line therapy, the vincristine/corticosteroid combination provides an up to 53% CR rate that is not inferior to a vincristine/corticosteroid/anthracycline-based regimen.11 Also, the recent use of liposomal vincristine for relapsed/refractory ALL showed results comparable to those we describe here (35% overall response and 20% CR/CRi).12 Thus, it is difficult to conclude whether epratuzumab will increase the therapeutic results of a non-intensive salvage chemotherapy in the particular setting studied here. Nevertheless, more interestingly, epratuzumab may provide an important addition in terms of induction of MRD. Indeed, 2 of our 5 responders were documented with negative MRD, which is currently considered, at least in younger patients, as a marker of long-term disease-free survival.13
In a recent study (submitted for publication) by Raetz et al.,14 the addition of epratuzumab also did not translate into a higher CR rate in pediatric ALL patients in relapse. But among the children in CR, those treated with epratuzumab were significantly more likely to become MRD negative as compared to those treated without epratuzumab. Moreover, in the series reported by Advani et al., in whom a combination of clofarabine/cytarabine/epratuzumab was investigated in younger patients with refractory/relapsed BALL, one patient achieved negative MRD and survived 11 months, much longer than all of the other patients for whom the median OS was five months.8
The question remains as to how to improve the results in older B-ALL patients. Clearly, there is a need for new treatment protocols designed for the elderly ALL patient, as well as a better understanding of the unique biological characteristics of the disease in this age group. Of course, other therapeutic monoclonal antibodies (inotuzumab, blinatumomab, rituximab), 2-generation purine analogs (nelarabine, clofarabine) or chimeric antigen receptor T-cell targeting CD19 may have their place in treatment approaches for older patients and should also be tested in this setting. A radiation approach, using epratuzumab conjugated with a therapeutic radionuclide (Y-epratuzumab tetraxetan radioimmunotherapy) could also be a promising therapeutic option for some CD22 B-ALL patients.15
In conclusion, our results show some activity of epratuzumab combined with vincristine and dexamethasone in the very high-risk, refractory/relapsed, older CD22 B-ALL population studied here. These results pave the way for integrating epratuzumab within first-line chemotherapy approaches in older CD22 B-ALL patients, in order to possibly improve the rate of negative MRD in this setting.
References
- Gökbuget N, Stanze D, Beck J. Outcome of relapsed adult lymphoblastic leukemia depends on response to salvage chemotherapy, prognostic factors, and performance of stem cell transplantation. Blood. 2012; 120(10):2032-2041. PubMedhttps://doi.org/10.1182/blood-2011-12-399287Google Scholar
- Gökbuget N. How I treat older patients with ALL. Blood. 2013; 122(8):1366-1375. PubMedhttps://doi.org/10.1182/blood-2012-07-379016Google Scholar
- Raponi S, De Propris MS, Intoppa S. Flow cytometric study of potential target antigens (CD19, CD20, CD22, CD33) for antibody-based immunotherapy in acute lymphoblastic leukemia: analysis of 552 cases. Leuk Lymphoma. 2011; 52(6):1098-1107. PubMedhttps://doi.org/10.3109/10428194.2011.559668Google Scholar
- Chevallier P, Robillard N, Houille G. Simultaneous study of five candidate target antigens (CD20, CD22, CD33, CD52, HER2) for antibody-based immunotherapy in B-ALL: a monocentric study of 44 cases. Leukemia. 2009; 23(4):806-807. PubMedhttps://doi.org/10.1038/leu.2008.303Google Scholar
- Leonard JP, Coleman M, Ketas JC. Epratuzumab, a humanized anti-CD22 antibody, in aggressive non-Hodgkin’s lymphoma: phase I/II clinical trial results. Clin Cancer Res. 2004; 10(16):5327-5334. PubMedhttps://doi.org/10.1158/1078-0432.CCR-04-0294Google Scholar
- Micallef IN, Maurer MJ, Wiseman GA. Epratuzumab with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy in patients with previously untreated diffuse large B-cell lymphoma. Blood. 2011; 118(15):4053-4061. PubMedhttps://doi.org/10.1182/blood-2011-02-336990Google Scholar
- Raetz EA, Cairo MS, Borowitz MJ. Chemoimmunotherapy reinduction with epratuzumab in children with acute lymphoblastic leukemia in marrow relapse: a Children’s Oncology Group pilot study. J Clin Oncol. 2008; 26(22):3756-3762. PubMedhttps://doi.org/10.1200/JCO.2007.15.3528Google Scholar
- Advani A, McDonough S, Coutre S. SWOG S0910: A phase 2 trial of clofarabine/cytarabine/epratuzumab for relapsed/refractory acute lymphocytic leukemia. Brit J Haematol. 2014; 165:504-509. PubMedhttps://doi.org/10.1111/bjh.12778Google Scholar
- Chevallier P, Pigneux A, Robillard N. Rituximab for the treatment of adult relapsed/refractory CD20 positive B-ALL patients: a pilot series. Leuk Res. 2012; 36(3):311-315. PubMedhttps://doi.org/10.1016/j.leukres.2011.11.010Google Scholar
- Mohty M, Labopin M, Volin L. Reduced-intensity versus conventional myeloablative conditioning allogeneic stem cell transplantation for patients with acute lymphoblastic leukemia: a retrospective study from the European Group for Blood and Marrow Transplantation. Blood. 2010; 116(22):4439-4443. PubMedhttps://doi.org/10.1182/blood-2010-02-266551Google Scholar
- Ferrari A, Annino L, Crescenzi S, Romani C, Mandelli F. Acute lymphoblastic leukemia in the elderly: results of two different treatment approaches in 49 patients during a 25-year period. Leukemia. 1995; 9(10):1643-1647. PubMedGoogle Scholar
- O’Brien M, Schiller G, Lister J. High-dose vincristine sulphate liposome injection for advanced, relapsed, refractory adult Philadelphia chromosome-negative acute lymphoblastic leukemia. J Clin Oncol. 2013; 31(6):676-683. PubMedhttps://doi.org/10.1200/JCO.2012.46.2309Google Scholar
- Beldjord K, Chevret S, Asnafi V. Oncogenetics and minimal residual disease are independent outcome predictors in adult patients with acute lymphoblastic leukemia. Blood. 2014; 123(24):3739-3749. PubMedhttps://doi.org/10.1182/blood-2014-01-547695Google Scholar
- Raetz EA, Cairo MS, Borowitz MJ. Reinduction chemoimmunotherapy with epratuzumab in relapsed acute lymphoblastic leukemia (ALL) in children, adolescents and young adults: Results from Children’s Oncology Group (COG) Study ADVL04P2. Blood. 2011; 118:573. Google Scholar
- Chevallier P, Bodet-Milin C, Robillard N. BCR-ABL1 molecular remission after 90Y-epratuzumab tetraxetan radioimmunotherapy in CD22+ Ph+ B-ALL: proof of principle. Eur J Haematol. 2013; 91(6):552-556. PubMedhttps://doi.org/10.1111/ejh.12183Google Scholar