Abstract
Patients with relapsed/refractory (R/R) chronic lymphocytic leukemia (CLL) may benefit from salvage chemoimmunotherapy (CIT). To explore further the use of CIT in the pre-novel agent era, ECOG-ACRIN undertook a phase 2 trial (E2903) for R/R CLL utilizing pentostatin, cyclophosphamide, and rituximab (PCR) followed by a consolidation course of alemtuzumab. This trial enrolled 102 patients with a median age of 64 years. Treatment consisted of PCR for six cycles followed by alemtuzumab for either 4 or 12 weeks depending upon the initial response to PCR. The overall response after PCR (complete remission (CR), nodular partial remission (nPR), partial remission (PR)) was 55%. Major responses (CR, or nPR) were achieved in 6%. The median overall survival (OS) and the median progression- free survival (PFS) were 28 months and 12 months respectively. The most serious non-lethal adverse events were myelosuppression, febrile neutropenia, fatigue, nausea, and hyponatremia. PCR is an effective and well-tolerated nucleoside-based regimen for heavily pretreated CLL patients with R/R disease. The addition of alemtuzumab to CLL patients with a minor response (PR) or stable disease did not result in a significant number of higher responses (CR or nPR) nor an improvement in OS.
Keywords: Pentostatin/cyclophosphamide/rituximab, Chemoimmunotherapy, Chronic lymphocytic leukemia, Relapsed/refractory disease, Alemtuzumab
INTRODUCTION
Treatment for relapsed/refractory (R/R) CLL, prior to the development of BCR signal inhibitors, consisted of purine nucleoside based therapy in combination with cyclophosphamide and an anti-CD20 monoclonal antibody (chemoimmunotherapy-CIT).1–4 The combination of fludarabine, cyclophosphamide and rituximab (FCR) can result in long-term survival for treatment naïve, fit patients with low risk genetics and mutated IGVH.1 The long-term administration of B-cell signal inhibitors alone,5 and in combination6 in treatment naïve and good prognosis patients, has resulted in long-term disease -free survival. Ibrutinib-based therapy versus CIT, in previously untreated patients, resulted in a superior PFS.7,8 However, only marrow ablative approaches 9,10 and CAR-T cell administration 11 have resulted in molecular cure of the disease.
Pentostatin is active alone and in combination in CLL.12–19 An earlier phase 2 trial tested a CIT regimen (PCR) and demonstrated significant efficacy in previously untreated elderly patients with CLL.4 Of particular importance was the ability to administer a full six cycles even in patients with diminished renal function. 20 Encouraged by these results we designed and activated a phase 2 trial, E2903, for R/R CLL that incorporated 6 cycles of PCR, followed by an anti- CD52 antibody, alemtuzumab, as consolidation. The use of alemtuzumab was based on its known effectiveness for the treatment of lymphoid neoplasms, including CLL, 21–30 its activity in patients with 17p- and p53 mutations.25 and activity in fludarabine resistant disease.26
PATIENTS and METHODS
Clinical sites, patient eligibility and trial objectives
This was a phase 2 trial for R/R CLL conducted by the ECOG-ACRIN NCTN Group (ECOG-ACRIN). Eligibility criteria included progressive disease or failure to achieve a meaningful response (< partial response (PR)) or relapse after prior therapy (including fludarabine, pentostatin and rituximab). The definition of progressive disease was based upon the criteria of the National Cancer Institute (NCI) Working Group.31 Patient requirements included an ECOG performance status of 0–3, serum creatinine <2 mg/dl and a creatinine clearance > 30ml/min. Patients were ineligible if they had received prior alemtuzumab. Patients with infections were not eligible for entry onto the study until resolution of the infection. All patients were tested for hepatitis B and those found with evidence of viremia were ineligible. Patients with prior hepatitis B infection and antibodies were eligible. Patients with a history of malignancy, other than squamous/basal cell carcinoma of the skin or in situ carcinoma of the cervix, were not eligible unless the tumor was curatively treated at least two years prior to consideration for the protocol. The protocol was reviewed and approved by the Institutional Review Board at each participating institution. All patients were required to provide written informed consent prior to entry, in accordance with the Declaration of Helsinki.
The primary clinical objectives of the trial were to determine the number of complete remissions (CR), nodular partial remissions (nPR) and partial remissions (PR). Secondary objectives included estimation of the overall survival (OS) and progression-free survival (PFS) and the conversion rate to a higher response category after alemtuzumab. Hematologic toxicity was graded according to the NCI Working Group guidelines 31 and non-hematologic toxicity was assessed according to the NCI Common Toxicity Criteria (Version 3.0). Clinical and laboratory correlates of outcome were analyzed, including the Rai Staging Classification, 32 β2-microglobulin (β2-m) 33,34 and immunoglobulin heavy chain variable region (IGVH) mutation status. Elevated CD 38, and ZAP-70 expression were both defined as >20% in the CLL population. 35 CLL FISH panel status36 was categorized utilizing the following hierarchy: high-risk group included 17p-, 11q-, 6q- defects, intermediate risk patients were trisomy 12 and low risk patients were 13q- or normal by FISH.
Study design
Patients received pentostatin (4mg/m2 IV) and cyclophosphamide (600mg/m2 IV) on day one. Rituximab was administered for the first cycle at 100 mg on day 1, followed by 375 mg/m2on days 3 and 5. For subsequent cycles (2–6) rituximab was given at a dose of 375 mg/m2 on the same day as the pentostatin and cyclophosphamide (PCR). This PCR regimen was given on a 21-day 6-cycle schedule. Bactrim DS (QOD) and acyclovir (800mg BID) were administered during the entire course of PCR, and alemtuzumab administration. Surveillance for cytomegalovirus (CMV) was performed at each cycle of PCR utilizing either pp65 antigenemia.37 or CMV DNA by real time polymerase chain reaction.38 Discovery of active disease resulted in cessation of therapy and treatment of the CMV. The first cycle of PCR was to be administered at full dose regardless of preexisting cytopenias. During cycle one G-CSF (Neupogen) was administered SQ daily, beginning two days after treatment for 10 consecutive days or until the neutrophil count was greater than 1.0 × 109/L for 2 consecutive days. Pegfilgrastim (6mg/ m2 every three weeks) was an alternative option. After the first cycle the decision to continue administration of growth factors was left up to the treating physician.
Patients achieving a CR or nPR after PCR were then observed during an 8 week treatment- free interval after which a bone marrow study was performed for pathologic confirmation of response. Response was evaluated by sites for treatment decisions and reviewed centrally by data managers. After confirmation of response, alemtuzumab (30mg SQ) was administered three times a week (TIW) for four weeks. The subcutaneous route of administration was utilized because of ease of administration and efficacy.39,40 The four week course for CR or nPR patients as opposed to the 18 week course for a lesser response was based upon the concept that robust responding patients would need less intensive exposure to alemtuzumab and have less infectious complications.41Patients achieving a PR or stable disease (SD) after six cycles or progressive disease (PD) after at least 2 cycles of PCR received alemtuzumab (30mg SQ TIW) for 18 weeks after a treatment-free period determined by the investigator.
Criteria for response
CR and nPR response criteria, determined after completion of PCR and after the 8 week hiatus noted above, was defined by the 1996 CLL Working Group criteria.31 Computerized imaging response criteria were not used to evaluate the responses. PD was characterized by at least one of the following criteria: >50% increase in nodal disease or appearance of new lymph nodes, increase in the size of liver or spleen, >50% increase in blood lymphocytes or appearance of Richter’s syndrome. Patients who did not achieve a CR, nPR or PR but exhibited no findings consistent with PD were considered to have SD.
Statistical analyses
The primary endpoint was the response rate to PCR. Response rates and confidence intervals (CIs) were calculated using the method of Atkinson and Brown.42The distribution of pretreatment characteristics between responders and non-responders was determined using the Fisher’s exact 43 and Wilcoxon rank sum tests.44 A multivariate logistic model was performed on the response rate to examine the effects of potential risk factors. OS was defined as the time from registration until death from any cause or censored at last date known to be alive. PFS was defined as the time from registration to disease progression or any cause of death in the absence of progression. OS and PFS were estimated using the Kaplan-Meier method.45 Log rank tests were used to examine the effects of risk factors on OS and PFS. Univariate and multivariate Cox models were performed on OS and PFS to examine the potential risk factors. Factors with P<0.15 in the univariate model were included in the multivariate model as covariates. If > 10% of patients had a missing value for a particular covariate an indicator for whether or not the covariate was missing was indicated in the multivariate mode. Landmark analysis at 6 months was done when patients had scheduled PCR treatment cycles completed and was utilized to compare the overall survival between responders and non-responders. All p-values reported are for two-sided significance tests with p-values under 0.05 considered significant.
RESULTS
Patient characteristics
A total of 102 patients were enrolled in E2903 beginning in December 2004 (Figure 1). The study was closed to accrual in May 2013. Two patients did not begin assigned therapy (declined to receive therapy; received alternative therapy) and four patients were ineligible for response because of violations of the inclusion criteria. Efficacy analysis included 96 patients and safety analysis (toxicity) included all 100 patients who started treatment. The baseline clinical, laboratory and prognostic factor characteristics are shown in Table 1. The median age was 64 years (range 38–81 years) and the majority of patients were male (79%). An ECOG performance status of 0–1 was present in 96% of patients and 57% of patients were Rai stages III-IV. The number of patients having received ≥ 2 prior treatments was 56%. Among the 91 patients with serum β2-m information, 56(58%) had a level ≤3.5 mgs/ml. Of the 66 patients with ZAP-70 and CD38 studies, 45% and 51%, respectively, were positive based upon previous flow threshold determinations.33 Unmutated IGHV status was found in 68% of available patient material. Pre-therapy FISH data was available in 78 patients and demonstrated high-risk defects in 47 %(17p-, 11q-, 6q), intermediate risk in 15% (trisomy 12) and low risk in 32% (13q-, normal).
Fig 1.
CONSORT diagram showing the analytic population of E2903.
Table 1:
Baseline characteristics
| N (%) | |
|---|---|
| Age (yrs) | |
| <=70 | 79 (82.3) |
| >70 | 17 (17.7) |
| Gender | |
| Male | 76 (79.2) |
| Female | 20 (20.8) |
| ECOG PS | |
| 0 | 53 (55.2) |
| 1 | 39 (40.6) |
| 2 | 4 (4.2) |
| Rai CLL Stage | |
| Missing | 2 (2.1) |
| 0-I | 29 (30.2) |
| II | 10 (10.4) |
| III-IV | 55 (57.3) |
| Clinical Status at Therapy | |
| First relapse | 41 (42.7) |
| Second or later relapse | 27 (28.1) |
| Refractory | 6 (6.3) |
| Progressive | 22 (22.9) |
| No. Prior Regimens | |
| Missing | 2 (2.1) |
| 1–2 | 40 (41.7) |
| >2 | 54 (56.3) |
| β-2 Microglobulin (μg/ml) | |
| Missing | 5 (5.2) |
| <=3.5 | 35 (36.5) |
| >3.5 | 56 (58.3) |
| ZAP 70 | |
| Missing | 30 (31.3) |
| Negative | 36 (37.5) |
| Positive | 30 (31.3) |
| CD38 | |
| Missing | 30 (31.3) |
| Negative | 32 (33.3) |
| Positive | 34 (35.4) |
| IGHV | |
| Missing | 45 (46.9) |
| Unmutated | 35 (36.5) |
| Mutated | 16 (16.7) |
| FISH Risk Category | |
| Missing | 18 (18.8) |
| Low | 29 (30.2) |
| Intermediate | 12 (12.5) |
| High | 37 (38.5) |
Responses to therapy and prognostic factor analysis
The median number of PCR cycles received was 5 (range: 1–6) with 46 completing all 6 cycles (47%). There were 11 patients not evaluable for response to PCR, all having received < 2cycles. The reasons included: death(4),thrombocytopenia(2), disease progression(1), myocardial infarction(1), adverse events(1), received one cycle(1), or no measurements of lymph node size (1). The OR was 55%, consisting of 3 CR’s (3%), 3 nPR’s (3%) and 47 PR’s (49%). Three patients (3%) who were classified as having maintained a complete clinical remission (CCR) after PCR at 8 weeks off-therapy, but without bone marrow confirmation for CR status, received alemtuzumab consolidation. A significant number of patients did not receive consolidation (Figure 1). The reasons for this included: adverse events/side effects (32), patient withdrawal/ refusal (7), death on study (6), disease progression/relapse during active treatment (2), or alternative therapy/other (9). As a result only 39 eligible patients subsequently received either alemtuzumab for 4 weeks (9 patients) or for 18 weeks (30 patients). Of the 9 patients on alemtuzumab for 4 weeks, all maintained their response status. Of the 30 patients on alemtuzumab for 18 weeks, 2 patients subsequently achieved a CR (7%). Of these 30, two patients were not evaluable for response. One patient refused further therapy after day 1 and one patient was taken off treatment after week 3 during the first consolidation course.
Patients with a lower (0–2) Rai stage (p=0.04), fewer prior regimens(<2) (p=0.02), hemoglobin >10g/dl (p= 0.03), β2-m ≤ 3.5 (p=0.03), and CD 38 negativity (p=0.01) were more likely to respond to PCR. In a multivariate logistic model only CD 38 <20% was significantly associated with a PCR response (p=0.01). The response rates are 81% and 50% for patients with CD38 negative and positive status respectively. Although baseline IGHV status and FISH studies were largely unfavorable, neither was associated with response (IGHV mutation: p=0.36; FISH risk: p=0.61)
Overall Survival and Progression-Free Survival
The median OS was 27 months (90% CI: 20–42)(Figure 2A) and the median PFS was 12 months (90% CI: 9–14)(Figure 2B). There was no significant difference in OS between CR/nPR responders (21 months) and lesser responders (29.0 months)(p=0.74).
Fig 2.
Kaplan-Meier estimation of (A) overall survival (OS) of all patients, (B)progression-free survival (PFS) of all patients.
In a multivariate model of OS, both β2-m ≤3.5 mg/L (p=0.01) and low FISH risk (p=.02) were significant factors for OS. If β2-m was ≤ 3.5 mg the median survival was 56 months versus 19 months if the β2-m was > 3.5 mg/L (Figure 3A). For low-risk FISH the median survival was 47.2 months versus 15.9 months for high-risk FISH (Figure3B). In a multivariate model, β2-m(≤ 3.5mg/L), IGHV mutation status, and lower FISH risk were not significantly associated with PFS (p=.08).
Fig 3.
Kaplan-Meier curves of OS according to (A) β−2 Microglobulin (B) FISH Risk Category.
Treatment-related toxicity
Treatment-related toxicities during PCR and alemtuzumab were those expected in patients with R/R CLL receiving CIT (Table 2). Anemia, thrombocytopenia and neutropenia were observed in 16%, 34% and 60%, respectively, during PCR treatment. The worst-degree of non-hematologic treatment-related toxicities during PCR treatment were grades 3–4 in 47 patients (47%) and grade 5 in 5 patients (5%). These included fatigue (9%), nausea (8%) and vomiting (7%). Metabolic abnormalities were observed in 18(18%) patients but all were less than grade 4. Tumor-lysis syndrome was documented in six patients, all grade 3. Febrile neutropenia occurred in 10 patients (10%). Most infections, either presumed or proven, were grades 1–3. These included respiratory tract, urinary tract, skin and ocular site infections. Colitis, unrelated to CMV, occurred in two patients (C. difficile, unknown). Septicemia was documented in 2 patients, one of whom died. Pneumonia, attributable to CMV occurred in 2 patients. There were 5 treatment-related deaths during the PCR therapy including multi-organ failure (1), ARDS, non-CMV (1), infection (1), pneumonitis (2).
Table 2.
Treatment-related toxicities (Grades 3–5)
| Step 1 | Step 2 | ||||
|---|---|---|---|---|---|
| PCR Treatment -6 weeks (Arm A) | N(%) | Alemtuzumab treatment- 4 weeks (Arm B) | N(%) | Alemtuzumab treatment- 18weeks (Arm C) | N(%) |
| Anemia | 16 (16) | Lymphopenia | 8 (89) | Anemia | 7 (23) |
| Lymphopenia | 60 (60) | Neutropenia | 4 (44) | Lymphopenia | 28 (90) |
| Neutropenia | 38 (38) | Platelets | 3 (33) | Neutropenia | 17 (55) |
| Platelets | 34 (34) | Platelets | 13 (42) | ||
| Fatigue | 9 (9) | F. Neutropenia | 5 (16) | ||
| Diarrhea | 5 (5) | Fatigue | 4 (13) | ||
| Nausea | 8 (8) | Pneumonitis | 2 (6) | ||
| Vomiting | 7 (7) | Lung infection | 5 (16)** | ||
| F. Neutropenia | 10 (10) | M-organ failure | 1 (3)* | ||
| Hyponatremia | 6 (6) | ||||
| Renal failure | 6 (6) | ||||
| T. Lysis Syndrome | 6 (6) | ||||
| Pneumonitis | 3 (3)* | ||||
| M-organ failure | 1 (1)* | ||||
| Respiratory disorder | 2 (2)* | ||||
| Septicemia | 2 (2)* | ||||
| ARDS | 1(1)* | ||||
1 of those patients had Grade 5
2 of those patients had Grade 5
During the alemtuzumab treatment arms, pain at the injection site, fever, and myalgias were grades I or 2. One patient with a grade 4 local injection site reaction was removed from study after 2 doses of alemtuzumab. Myelosuppression was as common as during the PCR. One patient developed CMV positive serology during the 4-week alemtuzumab treatment and two patients in subsequent follow-up. Two patients developed CMV positive serology during the 18-week alemtuzumab treatment, two of whom developed pneumonia.
The autoimmune disorders found in this study are those previously described in CLL patients. 46 Although autoimmune cytopenia occurs in CLL, occasionally related to purine nucleoside or alemtuzumab administration,47–49 there were no cases in the present study. Second malignancy in patients with CLL has been well-documented.50–53 The spectrum of second malignancies documented in this trial included non-Hodgkin lymphoma (NHL) (5), including three cases of Richter’s syndrome, myelodysplastic syndrome. (2), non-melanoma skin tumors (7), breast cancer (1), colorectal cancer (1), esophageal cancer (1), bladder cancer (1), and metastatic undifferentiated adenocarcinoma (1). No cases of melanoma were observed.54
DISCUSSION
In this study, 55% of R/R CLL patients achieved CR, nPR or PR during PCR treatment. However, only 3% achieved a CR and 3% an nPR,. CD 38 <20% was the only prognostic factor associated with a major response. β2-m was the only significant risk factor for OS, consistent with previous reports.33,34 No differences were found in OS and PFS between patients who achieved CR or nPR versus others, which may be due to the subsequent salvage therapies that the non-major responders received off-study, such as high dose chemotherapy, immunotherapy and stem-cell transplantation. These results were inferior to those of a previous PCR study without FISH data. 55Our results of this salvage trial of PCR are similar to the results of FCR in a series of patients having failed multiple prior regimens. 56 In a recent analysis of pentostatin-based therapies for upfront therapy of CLL an overall response rate across all trials of over 90% with a 41% CR rate was reported.57 The toxicity of PCR CIT appears tolerable, although, during PCR 40 patients (40%) developed grade 3 non-hematologic related toxicities and 12 (12%) patients had 4–5 non-hematologic treatment-related toxicities. The rationale for PCR CIT therapy is based on ease of administration and side effect profile particularly when administering to older patients with renal insufficiency.4
Numerous publications have documented the use of post-CIT consolidation therapy with alemtuzumab in previously untreated patients, reporting successful eradication of MRD and clonal mutations, improved survival and progression-free survival. 58–66 The present study in previously treated patients demonstrated the inability of alemtuzumab to convert a significant number of lesser clinical responses (i.e. PR), to CRs after PCR induction. CMV viremia occured when alemtuzumab was used alone in heavily pre-treated patients67 or combined with other agents in frontline therapy. 68 In the present study four patients (4%) developed clinical CMV disease.
Therapeutic options for patients with CLL with a poor prognosis have been recently expanded to include ibrutinib,5, idelalisib with rituximab6 and venetoclax.69 Although highly active, they may be accompanied by significant side effects such as atrial fibrillation, 70 bleeding, 71,72 infectious complications (ibrutinib),73 immune-related events such as rash, pneumonitis and/or colitis.74 and tumor lysis syndrome(venetoclax). Drug resistance and progression over time are observed with these agents. 75 Our data with PCR support the use of CIT in CLL patients particularly if they choose to have short-term rather than long-term therapy 76 or those in whom administration of ibrutinib may be contraindicated or difficult to administer ( i.e. patients on anticoagulants or antiplatelet agents).
Acknowledgments
The authors wish to acknowledge the important role of the patients for participating and donating samples to this study. This study was coordinated by the ECOG- ACRIN Cancer Research Group (Peter O’Dwyer, M.D. and Mitchell D. Schnall, M.D., Ph.D, Group Co-Chairs) and supported by the National Cancer Institute of the National Institutes of Health under the following award numbers: CA180820, CA180794, CA180790, CA180791, CA180799, CA180853, CA189825, CA189859, CA189956, CA189971. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S government. The authors also wish to acknowledge Henry Baptista and Dr. Mark Litzow for coordinating the clinical data and reviewing the manuscript respectively.
Footnotes
Conflict of interest disclosure: The authors declare no competing financial interests.
References
- 1.Fischer K, Bahlo J, Fink AM, et al. Long-term remissions after FCR chemoimmunotherapy in previously untreated patients with CLL: updated results of the CLL8 trial. Blood 2016;127(2):208–215. [DOI] [PubMed] [Google Scholar]
- 2.Thompson PA, Tam CS, O’Brien SM, et al. Fludarabine, cyclophosphamide, and rituxan treatment achieves long-term disease-free survival in IGHV-mutated chronic lymphocytic leukemia. Blood 2016;127(3):303–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Chai-Adisaksopha C, Brown JR. FCR achieves long-term durable remissions in patients with IGHV-mutated CLL. Blood 2017;130(21): 2278–2282. [DOI] [PubMed] [Google Scholar]
- 4.Kay N, Geyer S, Call TG, et al. Combination chemotherapy with pentostatin, cyclophosphamide, and rituximab shows significant clinical activity with low accompanying toxicity in previously untreated B chronic lymphocytic leukemia. Blood 2007;109:405–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.O’Brien SO, Furman RR, Coutre S, et al. Single-agent ibrutinib in treatment-naïve and relapse/refractory chronic lymphocytic leukemia: a 5-year experience. Blood 2018;131(17):1910–1919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Furman RR, Sharman JP, Coutre SE, et al. Idelalisib and Rituximab in Relapsed Chronic Lymphocytic Leukemia. N Eng J Med 2014; 370: 997–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Woyach JA, Ruppert AS, Heerema NA, et al. Ibrutinib Regimens versus Chemoimmunotherapy in Older Patients with Untreated CLL. N Engl J Med 2018;379:2517–2528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Shanafelt T, Wang V, Kay NE, et al. Ibrutinib-based Therapy vs Chemimmunotherapy for Chronic Lymphocytic Leukemia. Blood 2018; 132:LBA–4. [Google Scholar]
- 9.Dreger P, Dohner H, Ritgen M, et al. Allogeneic stem cell transplantation provides durable disease control in poor-risk chronic lymphocytic leukemia: long-term clinical and MRD results of the German CLL Study Group CLL3X trial. Blood 2010;116(14): 2438–2447. [DOI] [PubMed] [Google Scholar]
- 10.Dreger P, Dohner H, McClanahan F, et al. Early autologous stem cell transplantation for chronic lymphocytic leukemia: long-term follow-up of the German CLL Study Group CLL3 trial. Blood 2012;119(21): 4851–4859. [DOI] [PubMed] [Google Scholar]
- 11.Turtle CJ, Hay KA, Hanafi LA, et al. Durable molecular remissions in chronic lymphocytic leukemia treated with CD19 specific chimeric antigen receptor-modified T cells after failure of ibrutinib. J Clin Oncol 2017;35:3010–3020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Grever M, Siaw MP, Jacob WF, et al. The biochemical and clinical consequences of 2’-deoxycoformycin in refractory lymphoproliferative malignancy. Blood 1981;57:406–417. [PubMed] [Google Scholar]
- 13.Smyth JF, Grant Prentice H, Proctor S, Victor Hoffbrand A. Deoxycoformycin in the Treatment of Leukemias and Lymphomas. Ann NY Acad Sciences 1985;451:123–128.. [DOI] [PubMed] [Google Scholar]
- 14.Grever MR, Leiby JM, Kraut EH, et al. Low-Dose Deoxycoformycin in Lymphoid Malignancy. J Clin Oncol 1985;3:1196–1201. [DOI] [PubMed] [Google Scholar]
- 15.O’Dwyer PJ, Wagner B, Leyland-Jones B, et al. 2-’Deoxycoformycin (Pentostatin) for Lymphoid Malignancies. Rational Development of an Active Drug. Ann Int Med 1988;108:733–743. [DOI] [PubMed] [Google Scholar]
- 16.Ho A, Thaler J, Stryckmans P, et al. Pentostatin in refractory chronic lymphocytic leukemia: a phase II trial of the European Organization for Treatment of Cancer. J Natl Cancer Inst 1990;82(17):1416–1420. [DOI] [PubMed] [Google Scholar]
- 17.Dillman RO, Mack R, McIntyre RO. Pentostatin in Chronic Lymphocytic Leukemia. A Phase II Trial of Cancer and Leukemia Group B. J Clin Oncol 1989;7:433–438. [DOI] [PubMed] [Google Scholar]
- 18.Oken MM, Lee S, Kay NE, Knospe W, Cassileth PA. Pentostatin, chlorambucil and prednisone therapy for B-chronic lymphocytic leukemia: a phase I/II study by the Eastern Cooperative Oncology Study Group study E1488. Leuk Lymphoma 2004;45:79–84. [DOI] [PubMed] [Google Scholar]
- 19.Weiss MA, Maslak PG, Jurcic J et al. Pentoststin and cyclophosphamide: an effective new regimen in previously treated patients with chronic lymphocytic leukemia. J Clin Oncol 2003;21:1278–1284. [DOI] [PubMed] [Google Scholar]
- 20.Shanafelt TD, Lin T, Geyer SM, et al. Pentostatin, cyclophosphamide, and rituximab regimen in older patients with chronic lymphocytic leukemia. Cancer 2007; 109: 2291–2298. [DOI] [PubMed] [Google Scholar]
- 21.Dyer MJS, Hale G, Hayhoe FG, Waldmann H. Effects of CAMPATH −1 Antibodies In Vivo in Patients With Lymphoid Malignancies: Influence of Antibody Isotype. Blood 1989;73:1431–1439. [PubMed] [Google Scholar]
- 22.Dyer MJS, Hale G, Marcus R, Waldmann H. Remission Induction in Patients With Lymphoid Malignancies Using Unconjugated CAMPATH-1 Monoclonal Antibodies. Leukemia and Lymphoma 1990;2: 179–193. [DOI] [PubMed] [Google Scholar]
- 23.Hillmen P, Skotnicki AB, Robak T et al. Alemtuzumab Compared With Chlorambucil As First-Line Therapy for Chronic Lymphocytic Leukemia. J Clin Oncol 2007;25:5616–5623. [DOI] [PubMed] [Google Scholar]
- 24.Osterborg A, Dyer MJ, Bunjes D, et al. A phase II multi-center study of human CD 52 antibody in previously treated chronic lymphocytic leukemia. European Study Group of CAMPATH-1H Treatment in chronic lymphocytic leukemia. J Clin Oncol 1997;15:1567–1574. [DOI] [PubMed] [Google Scholar]
- 25.Lozanski G, Heerema NA, Flinn IW, et al. Alemtuzumab is an effective therapy for chronic lymphocytic leukemia with p53 mutations and deletions. Blood 2004;103:3278–3281. [DOI] [PubMed] [Google Scholar]
- 26.Keating MJ, Flinn I, Jain V, et al. Therapeutic role of alemtuzumab (CAMPATH-1H) in patients who have failed fludarabine: results of a large international study. Blood 2002;99:3554–3561. [DOI] [PubMed] [Google Scholar]
- 27.Montillo T, Tedeschi A, Petrizzi VB, et al. An open-label, pilot study of fludarabine, cyclophosphamide, and alemtuzumab in relapsed/refractory patients with B-cell chronic lymphocytic leukemia. Blood 2011;118:4079–4085. [DOI] [PubMed] [Google Scholar]
- 28.Badoux XC, Keating MJ, Wang X, et al. Cyclophosphamide, fludarabine, alemtuzumab, and rituximab as salvage therapy for heavily pretreated patients with chronic lymphocytic leukemia. Blood 2011;118:2085–2093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Geisler CH, van’t Veer MB, Jurlander J, et al. Frontline low-dose alemtuzumab with fludarabine and cyclophosphamide prolonged progression-free survival in high–risk CLL. Blood 2014;123(21):3255–3262. [DOI] [PubMed] [Google Scholar]
- 30.Parikh SA, Keating MJ, O’Brien S, et al. Frontline chemoimmunotherapy with fludarabine, cyclophosphamide, alemtuzumab, and rituximab for high-risk chronic lymphocytic leukemia. Blood 2011;118:2062–2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Cheson BD, Bennett JM, Grever M, et al. National Cancer Institute -sponsored Working Group guidelines for chronic lymphocytic leukemia: revised guidelines for diagnosis and treatment. Blood 1996;87(12):4990–4997. [PubMed] [Google Scholar]
- 32.Rai KR, Sawitsky A, Cronkite EP, Chanana AD, Levy RN. Clinical staging of chronic lymphocytic leukemia. Blood 1975;46(2):219–234 [DOI] [PubMed] [Google Scholar]
- 33.Tsimberidou AM, Tam C, Wierda W, O’Brien S, Lerner S, Keating MJ. β−2 microglobulin (B2M) is an independent prognostic factor for clinical outcome in patients with CLL treated with frontline fludarabine, cyclophosphamide, and rituximab (FCR) regardless of age, creatinine clearance (CrCL). J Clin Oncol 2007;25(18S),Abstract 7034 [Google Scholar]
- 34.Gentile M, Cutrona G, Neri A, et al. Predictive value of β2-microglobulin (B2-m) levels in chronic lymphocytic leukemia since Binet A stages. Haematologica 2009;94(6): 887–888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Rassenti LZ, Jain S, Keating MJ, et al. Relative value of ZAP-70, CD38, and immunoglobulin mutation status in predicting aggressive disease in chronic lymphocytic leukemia. Blood 2008;112(5):1923–1930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Dewald GW, Brockman SR, Paternoster SF, et al. Chromosome anomalies detected by interphase fluorescence in situ hybridization: correlation with significant biological features of B-cell chronic lymphocytic leukaemia. Br J Haematol 2003;121:287–295. [DOI] [PubMed] [Google Scholar]
- 37.Ho SKN, Lo CY, Cheng IKP, Chan TM. Rapid Cytomegalovirus pp65 Antigenemia Assay by Direct Erythrocyte Lysis and Immunoflourescence Staining. J Clin Microbiol 1998;36(3):638–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Jebbink J, Bai X, Rogers BB, Dawson DB, Scheuermann RH, Domiati-Saad R. Development of Real-Time PCR Assays for the Quantitative Detection of Epstein-Barr Virus and Cytomegalovirus, Comparison of TaqMan Probes and Molecular Beacons. J Mol Diagn 2003;5(1): 15–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bowen AL, Zomas A, Emmett E, Matutes E, Dyer MJ, Catovsky D. Subcutaneous CAMPATH-1H in fludarabine-resistant/relapsed chronic lymphocytic and B-prolymphocytic leukaemia. Br J Haematol 1997;96:617–619. [DOI] [PubMed] [Google Scholar]
- 40.Stilgenbauer S, Zenz T, Winkler D, et al. Subcutaneous Alemtuzumab in Fludarabine-Refractory Chronic Lymphocytic Leukemia: Clinical Results and Prognostic Marker Analyses From the CLL2H Study of the German Chronic Lymphocytic Leukemia Study Group. J Clin Oncol 2009;27:3994–4001. [DOI] [PubMed] [Google Scholar]
- 41.Elter T, Vehreschild JJ, Gribben J, Cornely OA, Engert A, Hallek M. Management of Infections in Patients with Chronic Lymphocytic Leukemia Treated with Alemtuzumab. Ann Hematol 2009;88:121–132. [DOI] [PubMed] [Google Scholar]
- 42.Atkinson EN, Brown BW. Confidence limits for probability of response in multistage phase II clinical trials. Biometrics 1985;41(3): 741–744. [PubMed] [Google Scholar]
- 43.Fisher RA. On the interpretation of X2 from contingency tables and the calculation of P. J Royal Statistical Soc 1922;85(1)87–94 [Google Scholar]
- 44.Wilcoxon F Comparisons by ranking methods. Biometrics Bull 1945;1(6):80–83 [Google Scholar]
- 45.Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Statist Assoc 1958;53:457–481. [Google Scholar]
- 46.Jung M, Rice I. Unusual autoimmune non-hematologic complications in chronic lymphocytic leukemia. Clin Lymphoma, Myeloma and Leukemia. 2011;11:510–513. [DOI] [PubMed] [Google Scholar]
- 47.Hamblin TJ. Autoimmune complications of chronic lymphocytic leukemia. Semin Oncol 2006;33:230–239. [DOI] [PubMed] [Google Scholar]
- 48.Zent CS, Kay NE. Autoimmune Complications in Chronic Lymphocytic Leukemia (CLL). Best Pract Res Clin Haematol 2010;23(1): 47–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Otton SH, Turner DL, Frewin R, Davies SV, Johnson SA. Autoimmune thrombocytopenia after treatment with Campath −1H in a patient with chronic lymphocytic leukaemia. Br J Haematol 1999;106(1):261–262. [DOI] [PubMed] [Google Scholar]
- 50.Schollkopf C, Rosendahl D, Rostgaard K, Pipper C, Hjalgrim H. Risk of Second cancer after chronic lymphocytic leukemia. Int J Cancer 2007;121:151–156. [DOI] [PubMed] [Google Scholar]
- 51.Morton LM, Curtis RE, Linet MS, et al. Second Malignancy Risks After Non-Hodgkin’s Lymphoma and Chronic Lymphocytic Leukemia Subtype. J Clin Oncol 2010;28:4935–4944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Smith MR, Neuberg D, Flinn IW, et al. Incidence of therapy-related myeloid neoplasia after initial therapy for chronic lymphocytic leukemia with fludarabine-cyclophosphamide versus fludarabine: long-term follow-up of US Intergroup Study E2997. Blood 2011; 118(13): 3525–3527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cheson BD, Vena DA, Barrett J, Freidlin B. Second Malignancies as a Consequence of Nucleoside Analog Therapy for Chronic Lymphoid Leukemias. J Clin Oncol 1999;17:2454–2460. [DOI] [PubMed] [Google Scholar]
- 54.Lam CJ, Curtis RE, Dores GM, et al. Risk Factors for Melanoma Among Survivors of Non-Hodgkin Lymphoma. J Clin Oncol 2015;33(28):3096–3104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lamanna N, Kalaycio M, Maslak P, et al. Pentostatin, Cyclophosphamide, and Rituximab is an Active, Well-Tolerated Regimen for Patients with Previously Treated Chronic Lymphocytic Leukemia. J Clin Oncol 2006;24:1575–1581. [DOI] [PubMed] [Google Scholar]
- 56.Badoux XC, Keating MJH, Wang X, et al. Fludarabine, cyclophosphamide, and rituximab chemoimmunotherapy is highly effective treatment for relapsed patients with CLL. Blood 2011;117:3016–3024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kay NE, LaPlant BR, Pettinger AM, et al. Cumulative experience and long term follow-up of pentostatin based chemoimmunotherapy trials for patients with chronic lymphocytic leukemia. Expert Rev Hematol 2018;11(4):337–349 [DOI] [PubMed] [Google Scholar]
- 58.Lepretre S, Aurran T, Mahe B, et al. Excess mortality after treatment with fludarabine and cyclophosphamide in combination with alemtuzumab in previously untreated patients with chronic lymphocytic leukemia in a randomized phase 3 trial. Blood 2012;119(22):5104–5110. [DOI] [PubMed] [Google Scholar]
- 59.Dyer MJ, Kelsey SM, MacKay HJ, et al. In vivo “purging” of residual disease in CLL with CAMPATH −1H. Br J Haematol 1997;97:669–672. [DOI] [PubMed] [Google Scholar]
- 60.Montillo M, Cafro AM, Tedeschi A, et al. Safety and efficacy of subcutaneous CAMPATH-1H for treating residual disease in patients with chronic lymphocytic leukemia responding to fludarabine. Hematologica 2002;87: 695–700. [PubMed] [Google Scholar]
- 61.O’Brien SM, Kantarjian HM, Thomas DA, et al. Alemtuzumab as treatment for residual disease after chemotherapy in patients with chronic lymphocytic leukemia. Cancer 2003;98:2657–2663. [DOI] [PubMed] [Google Scholar]
- 62.Wendtner CM, Ritgen M, Schweighofer CD, et al. Consolidation with alemtuzumab in patients with chronic lymphocytic leukemia(CLL) in first remission- an experience on safety and efficacy within a randomized multicenter phase III trial of the German CLL Study Group (GCLLSG). Leukemia 2004;18:1093–1101. [DOI] [PubMed] [Google Scholar]
- 63.Schweighofer CD, Ritgen M, Eichhorst BF, et al. Consolidation with alemtuzumab improves progression-free survival in patients with chronic lymphocytic leukemia (CLL) in first remission-long-term follow-up of a randomized phase III trial of the German CLL Study Group (GCLLSG). Br J Haematol 2008;144:95–98. [DOI] [PubMed] [Google Scholar]
- 64.Montillo M, Tedeschi A, Miqueleiz S, et al. Alemtuzumab As Consolidation After a Response to Fludarabine Is Effective in Purging Residual Disease in Patients With Chronic Lymphocytic Leukemia. J Clin Oncol 2006;24: 2337–2342. [DOI] [PubMed] [Google Scholar]
- 65.Hainsworth JD, Vazquez ER, Spigel DR, et al. Combination therapy with fludarabine and rituximab followed by alemtuzumab in the first-line treatment of patients with chronic lymphocytic leukemia or small lymphocytic lymphoma. Cancer 2008;112(6):1288–1295. [DOI] [PubMed] [Google Scholar]
- 66.Moreton P, Kennedy B, Lucas G, et al. Eradication of Minimal Residual Disease in B-Cell Chronic Lymphocytic Leukemia After Alemtuzumab Therapy Is Associated With Prolonged Survival. J Clin Oncol 2005;23:2971–2979. [DOI] [PubMed] [Google Scholar]
- 67.Nguyen DD, Cao TM, Dugan K, Starcher SA, Fechter RL, Coutre SE. Cytomegalovirus viremia during Campath-1H therapy for relapsed and refractory chronic lymphocytic leukemia. Clin Lymphoma 2002;3(2):105–110. [DOI] [PubMed] [Google Scholar]
- 68.Lin TS, Donohue KA, Byrd JC, et al. Consolidation Therapy With Subcutaneous Alemtuzumab After Fludarabine and Rituximab Induction Therapy for Previously Untreated Chronic Lymphocytic Leukemia: Final Analysis of CALGB 10101. J Clin Oncol 2010;28:4500–4506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Seymour JF, Kipps TJ, Eichorst B, et al. Venetoclax-Rituximab in Relapsed or Refractory Chronic Lymphocytic Leukemia. N Engl J Med 2018; 378:1107–1120. [DOI] [PubMed] [Google Scholar]
- 70.Leong DP, Caron F, Hills C, et al. The risk of atrial fibrillation with ibrutinib use: a systematic review and meta-analysis. Blood 2016;138: 138–140. [DOI] [PubMed] [Google Scholar]
- 71.Caron F, Leong DP, Hills C, Fraser G, Siegel D. Current understanding of bleeding with ibrutiunib use: a systematic review and meta-analysis. Blood Adv 2017;1(12): 772–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Shatzel JJ, Olson SR, Tao DL, et al. Ibrutinib-associated bleeding; pathogenesis, management, and risk reduction strategies. J Thromb Haemost 2017;15(5):835–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ghez D, Calleja A, Protin C, et al. Early-onset invasive aspergillosis and other fungal infections in patients treated with ibrutinib. Blood 2018;131(17):1955–1959. [DOI] [PubMed] [Google Scholar]
- 74.Weidner AS, Panarelli NC, Geyer JT. Idelalisib-associated Colitis: Histologic Findings in 14 Patients. Am J Surg Pathol 2015; 39(12): 1661–1667. [DOI] [PubMed] [Google Scholar]
- 75.Ahn IE, Underbayev C, Albitar A, et al. Clonal evolution leading to ibrutinib resistance in chronic lymphocytic leukemia. Blood 2017;129(11):1469–1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Brown JR, Kay NE. Chemoimmunotherapy Is Not Dead Yet in Chronic Lymphocytic Leukemia. J Clin Oncol 2017; 35 :1–4. [DOI] [PubMed] [Google Scholar]





