• Pomalidomide-cyclophosphamide-prednisone is an active combination in multiple myeloma patients who are relapsed/refractory to lenalidomide.

We performed a phase 1/2 trial to determine the maximum tolerated dose (MTD) of pomalidomide and to explore its efficacy when combined with cyclophosphamide-prednisone in relapsed/refractory myeloma patients. Pomalidomide was given at 1 to 2.5 mg/d, cyclophosphamide at 50 mg every other day, prednisone at 50 mg every other day, for 6 28-day cycles, followed by pomalidomide-prednisone maintenance therapy. Thromboprophylaxis was recommended. Sixty-nine patients were enrolled, 55 received the MTD (2.5 mg/d) and were evaluated. Best responses included complete response in 3 patients (5%), very good partial response in 10 (18%), partial response in 15 (27%), minimal response in 11 (20%), stable disease in 15 (27%), and progressive disease in 1 (3%), for an overall response rate of 51%. The median time-to-response was 1.83 months. After a median follow-up of 14.8 months, median progression-free survival was 10.4 months and 1-year overall survival was 69%. At the MTD, grade 3 to 4 toxicities included anemia (9%), thrombocytopenia (11%), neutropenia (42%), neurologic events (7%), dermatologic events (7%), and thromboembolism (2%). Grade 3 to 5 infections occurred in 5 patients (9%). Five patients (9%) discontinued treatment for toxicity. New grade 3 to 4 adverse events were low during maintenance. Pomalidomide-cyclophosphamide-prednisone is safe and effective in relapsed/refractory myeloma patients. This trial was registered at www.clinicaltrials.gov as #NCT01166113.

Multiple myeloma (MM) is characterized by a clonal proliferation of malignant plasma cells in the bone marrow and osteolytic lesions.1  The introduction of novel agents, such as thalidomide, lenalidomide, and bortezomib, has considerably improved response rates and survival, both at diagnosis and at relapse.2 

However, MM remains incurable, and the majority of patients relapse and become refractory to available therapies. The outcome of these patients is very poor, with a median event-free survival of 5 months and overall survival (OS) of 9 months.3  Newer agents able to overcome drug resistance and achieve a sustained disease control are needed.

Several immunomodulatory derivatives were generated by introducing chemical modifications to the structural backbone of thalidomide. Pomalidomide, a closely related analog of thalidomide, showed potent activity against tumor necrosis factor α secretion and entered clinical studies.4,5  Pomalidomide at 2 to 4 mg, combined with low-dose dexamethasone, has shown significant activity in pretreated patients refractory to lenalidomide and/or bortezomib.6-10  Partial response (PR) rates were 32% to 35%, and median progression-free survival (PFS) was 4.6 to 6.3 months.8-10  Neutropenia and thrombocytopenia were the most frequent adverse events.11 

The addition of alkylating agents to bortezomib and lenalidomide increased the response rates and, in some cases, prolonged disease-free interval.12  So far, no data on the role of cyclophosphamide added to pomalidomide are available.

These observations provided the rationale for this phase 1/2 trial. The primary aim of the study was to identify the most appropriate dose of pomalidomide in combination with cyclophosphamide-prednisone (PCP) and to determine its safety, tolerability, and efficacy in MM patients relapsed and/or refractory to lenalidomide.

Study population

Patients with MM who were 18 years of age or older, were relapsed or relapsed/refractory to lenalidomide, and who had received 1 to 3 prior lines of therapy were eligible. Relapse was defined as the reoccurrence of disease requiring the initiation of a salvage therapy, and refractory disease was defined as relapse while receiving salvage therapy or progression within 60 days of the most recent therapy. Patients were required to have measurable disease, a Karnofsky performance status 60% or higher, a platelet count 50 × 109/L or higher, a neutrophil count of 1.00 × 109/L or higher, a corrected serum calcium 3.5 mmol/L (14 mg/dL) or lower, serum hepatic aminotransferase levels 2.5-fold or less of the upper limit of normal, total bilirubin 1.5-fold or less of the upper limit of normal, and serum creatinine 2 mg/dL or less, and to agree to use contraception. Patients with clinically relevant active comorbid medical or psychiatric conditions or history of malignancy within the last 5 years were excluded. The institutional review board at each participating center approved the study in accordance with the Declaration of Helsinki. All patients provided written informed consent.

Study design

This trial was a phase 1/2, dose-escalating, open-label study. The primary endpoint of the dose-finding phase 1 was to identify the maximum tolerated dose (MTD) of pomalidomide, defined as the dose that achieved a dose-limiting toxicity (DLT) in 25% of patients. DLTs were defined as grade 4 neutropenia lasting more than 3 days, other grade 4 hematologic toxicity, any grade 3 or higher nonhematologic toxicity, febrile neutropenia, and/or infection requiring antibiotics occurring during the first cycle of therapy.

In phase 2, patients received the MTD of pomalidomide established in phase 1. The primary endpoint of phase 2 was the rates of complete response (CR) and very good PR (VGPR). Secondary endpoints were PFS and OS.

All adverse events were assessed during each cycle and graded according to the National Cancer Institute Common Toxicity Criteria (version 3.0).13 

Responses were recorded during every cycle, according to the International Myeloma Working Group criteria.14  Responses among patients refractory to previous novel agents and at high risk were analyzed. High risk was defined as the presence of t(4;14), t(14;16), or del 17p13 at enrolment, detected by fluorescence in situ hybridization (FISH).

Procedures

Oral pomalidomide was administered at doses ranging from 1 to 2.5 mg/d in phase 1 and the MTD in phase 2, in combination with cyclophosphamide at 50 mg every other day and prednisone at 50 mg every other day on days 1 to 28, for 6 cycles of 28 days each (see supplemental Appendix 1 available on the Blood Web site). Maintenance therapy consisted of pomalidomide 1 mg/d and prednisone 25 mg every other day continuously until any sign of relapse or progression.

Pomalidomide dose reduction (from 2.5 to 2 to 1.5 to 1 to 0.5 mg/d) was allowed if toxicities occurred. Grade 4 neutropenia, or febrile neutropenia with any other hematologic toxicities, or any other grade 4 hematologic or any grade 3 nonhematologic toxicities required immediate interruption of treatment and subsequent dose reduction at the start of the following cycle. A new cycle could be started if the neutrophil count was 1.00 × 109/L or higher, platelet count was 50 × 109/L or higher, hemoglobin was 8 g/dL or higher, and nonhematologic adverse events were grade 2 or lower. If the start of a new cycle were delayed by 2 or more weeks, dose reductions were required. Aspirin 100 mg/day or low-molecular-weight heparin was recommended as prophylaxis, according to patient risk.15 

Statistical analysis

In phase 1, the continual reassessment method was used as the dose allocation rule in the trial.16,17  It is based on a mathematical modeling of dose–DLT relationship, iteratively updated using Bayes theorem. Before trial onset, prior opinions about DLT probability at each dose level were elicited from expert clinicians and were fixed at 0.15, 0.20, 0.30, and 0.45, respectively. A design with grouped inclusions of 4 patients per dose level was chosen; the starting dose was 1.5 mg. The dose level associated with an updated DLT probability close to 25% was administered to the next cohort. All this process was re-run until the fixed sample size (N = 24) was reached,18  using the BPCT software.19 

In phase 2, we used a Simon optimal 2-stage design for the sample size calculation. A 5% response rate was considered not promising, a 20% rate was promising. The probability of both type I and type II errors was set at 0.05. Accordingly, 24 patients were planned in the first stage, and 31 (total = 55) were planned in the second stage. Our design required VGPR or better in at least 2 patients in the first stage to proceed to the second stage, and at least 5 patients for the treatment to be worth further consideration. Patients enrolled at the MTD in phase 1 were also included in phase 2.

All patients meeting the eligibility criteria who had received at least a single dose of pomalidomide were evaluated for response, toxicity, and survival. For responding patients, we measured the median time to response from the start of treatment to the date of the first response, as well as the duration of response from response to PD or death, censored at the date of last assessment for patients not progressing. We evaluated PFS and OS from the start of treatment until PD or death and until death, respectively.

Survival curves were estimated with the Kaplan-Meier method and compared with log-rank test.20  The individual effects on PFS of age (>75 vs ≤75 years), FISH-defined risk (high vs standard), and achievement of at least PR (treated as a time-dependent variable) were evaluated using a Cox's model. Results are presented as hazard ratios (HRs) and 95% confidence intervals (95% CIs). The analyses were performed using SAS software, version 8.2 (SAS Institute). Data cutoff was October 16, 2012.

Role of the funding source

The pharmaceutical sponsor was not involved in the study design, collection, analysis, or interpretation of the data or the writing of the report. Celgene supplied pomalidomide free of charge. The corresponding author had full access to all the data and had final responsibility for the decision to submit this manuscript for publication.

Patient characteristics

Between August 2010 and May 2012, 69 patients were enrolled at 12 Italian centers. In the phase 1 study, 24 patients were accrued. In the phase 2 study, 12 patients who received the MTD during phase 1 and an additional 45 patients were enrolled. Two patients who rapidly developed PD and died were excluded from the analysis because they failed to start therapy.

Patient characteristics are listed in Table 1. The median age was 69 years (range, 41-84 years). The median number of previous treatments was 3 (range, 1-3 treatments). Nine percent, 29%, and 62% of patients had received 1, 2, and 3 prior regimens, respectively. All patients (100%) had previously received lenalidomide, 84% bortezomib, 20% thalidomide, 33% autologous transplantation, and 14% allogeneic stem-cell transplantation. Twenty-three patients were relapsed and 46 were refractory to lenalidomide; 22 were refractory to both lenalidomide and bortezomib. The median time from diagnosis to study entry was 53 months (range, 11-203 months). Eighteen patients (26%) were classified as high-risk by FISH.

Table 1

Baseline characteristics

VariablePhase 1 (N = 24)Phase 2 (N = 55)All patients (N = 69)
Age    
 Median-years (range) 71 (50-82) 69 (41-84) 69 (41-84) 
Sex    
  Female 9 (37.5%) 27 (49%) 33 (48%) 
  Male 15 (62.5%) 28 (51%) 36 (52%) 
International Staging System stage    
 I 13 (54%) 28 (51%) 34 (49%) 
 II 10 (42%) 21 (38%) 27 (39%) 
 III 1 (4%) 6 (11%) 8 (12%) 
Myeloma protein class    
 IgG 16 (67%) 34 (62%) 42 (61%) 
 IgA 4 (17%) 13 (24%) 16 (23%) 
 Bence-Jones protein 3 (12%) 8 (14%) 10 (15%) 
 Nonsecretory 1 (4%) 1 (1%) 
Karnofsky performance status, %    
 60-70 3 (12%) 7 (13%) 10 (14%) 
 80 5 (21%) 9 (16%) 13 (19%) 
 90-100 16 (67%) 39 (71%) 46 (67%) 
Serum β2-microglobulin level    
 Median (mg/L) 2.9 (0.03-9) 3 (0.03-9) 3 (1.6-12) 
Months from diagnosis to on study    
 Median (range) 59 (13-203) 53 (11-203) 53 (11-203) 
Prior lines of therapy    
 Median (range) 3 (1-3) 3 (1-3) 3 (1-3) 
Prior therapies    
 Lenalidomide 24 (100%) 55 (100%) 69 (100%) 
 Bortezomib 20 (83%) 46 (84%) 58 (84%) 
 Thalidomide 4 (17%) 11 (20%) 14 (20%) 
 Autologous transplant 7 (29%) 18 (33%) 23 (33%) 
 Allogeneic transplant 3 (12%) 9 (16%) 10 (15%) 
Previous lenalidomide    
 Relapsed 9 (37.5%) 18 (33%) 23 (33%) 
 Refractory 15 (62.5%) 37 (67%) 46 (67%) 
Previous bortezomib    
 Relapsed 4 (17%) 14 (25%) 17 (25%) 
 Refractory 10 (42%) 20 (36%) 27 (39%) 
 Not available 6 (25%) 12 (22%) 14 (20%) 
FISH*    
 High risk 4 (17%) 13 (24%) 18 (26%) 
 Standard risk 9 (37%) 31 (56%) 35 (51%) 
 Not available 11 (46%) 11 (20%) 16 (23%) 
Chromosome abnormalities    
 Del 13 6 (25%) 18 (33%) 24 (35%) 
 t(4;14) 2 (8%) 6 (11%) 8 (12%) 
 t(11;14) 2 (8%) 10 (18%) 12 (17%) 
 t(14;16) 2 (4%) 3 (4%) 
 Del17 2 (8%) 5 (9%) 8 (12%) 
VariablePhase 1 (N = 24)Phase 2 (N = 55)All patients (N = 69)
Age    
 Median-years (range) 71 (50-82) 69 (41-84) 69 (41-84) 
Sex    
  Female 9 (37.5%) 27 (49%) 33 (48%) 
  Male 15 (62.5%) 28 (51%) 36 (52%) 
International Staging System stage    
 I 13 (54%) 28 (51%) 34 (49%) 
 II 10 (42%) 21 (38%) 27 (39%) 
 III 1 (4%) 6 (11%) 8 (12%) 
Myeloma protein class    
 IgG 16 (67%) 34 (62%) 42 (61%) 
 IgA 4 (17%) 13 (24%) 16 (23%) 
 Bence-Jones protein 3 (12%) 8 (14%) 10 (15%) 
 Nonsecretory 1 (4%) 1 (1%) 
Karnofsky performance status, %    
 60-70 3 (12%) 7 (13%) 10 (14%) 
 80 5 (21%) 9 (16%) 13 (19%) 
 90-100 16 (67%) 39 (71%) 46 (67%) 
Serum β2-microglobulin level    
 Median (mg/L) 2.9 (0.03-9) 3 (0.03-9) 3 (1.6-12) 
Months from diagnosis to on study    
 Median (range) 59 (13-203) 53 (11-203) 53 (11-203) 
Prior lines of therapy    
 Median (range) 3 (1-3) 3 (1-3) 3 (1-3) 
Prior therapies    
 Lenalidomide 24 (100%) 55 (100%) 69 (100%) 
 Bortezomib 20 (83%) 46 (84%) 58 (84%) 
 Thalidomide 4 (17%) 11 (20%) 14 (20%) 
 Autologous transplant 7 (29%) 18 (33%) 23 (33%) 
 Allogeneic transplant 3 (12%) 9 (16%) 10 (15%) 
Previous lenalidomide    
 Relapsed 9 (37.5%) 18 (33%) 23 (33%) 
 Refractory 15 (62.5%) 37 (67%) 46 (67%) 
Previous bortezomib    
 Relapsed 4 (17%) 14 (25%) 17 (25%) 
 Refractory 10 (42%) 20 (36%) 27 (39%) 
 Not available 6 (25%) 12 (22%) 14 (20%) 
FISH*    
 High risk 4 (17%) 13 (24%) 18 (26%) 
 Standard risk 9 (37%) 31 (56%) 35 (51%) 
 Not available 11 (46%) 11 (20%) 16 (23%) 
Chromosome abnormalities    
 Del 13 6 (25%) 18 (33%) 24 (35%) 
 t(4;14) 2 (8%) 6 (11%) 8 (12%) 
 t(11;14) 2 (8%) 10 (18%) 12 (17%) 
 t(14;16) 2 (4%) 3 (4%) 
 Del17 2 (8%) 5 (9%) 8 (12%) 
*

High-risk FISH was defined as the presence of at least one of the following abnormalities: t(4;14), t(14;16), or del17.

Phase 1

Table 2 lists the assigned pomalidomide dose levels and the observed DLTs. The dose level 2.5 mg/d was defined as the MTD, with an estimated probability of DLT of 0.258 (95% credibility interval, 0.101-0.468). DLTs were recorded in 4 patients: 1 grade 4 thrombocytopenia with 1.5 mg/d pomalidomide, 1 grade 3 peripheral neuropathy, 1 grade 3 hepatic toxicity, and 1 grade 4 thrombocytopenia, with 2.5 mg/d pomalidomide. The dose of pomalidomide maintenance was increased to the identified MTD.

Table 2

Phase 1: DLT for each cohort of enrolled patients

CohortDose (mg/day)DLTs, nType of DLTsUpdated estimated probability of DLT per dose level
1 mg1.5 mg2 mg2.5 mg
1.5 Grade 4 thrombocytopenia 0.237 0.298 0.409 0.553 
1.0 — 0.104 0.145 0.232 0.376 
2.0 — 0.051 0.076 0.136 0.255 
2.5 Grade 3 neuropathy 0.052 0.078 0.139 0.259 
2.5 Grade 3 hepatic 0.052 0.078 0.139 0.259 
2.5 Grade 4 thrombocytopenia 0.052 0.077 0.138 0.258 
CohortDose (mg/day)DLTs, nType of DLTsUpdated estimated probability of DLT per dose level
1 mg1.5 mg2 mg2.5 mg
1.5 Grade 4 thrombocytopenia 0.237 0.298 0.409 0.553 
1.0 — 0.104 0.145 0.232 0.376 
2.0 — 0.051 0.076 0.136 0.255 
2.5 Grade 3 neuropathy 0.052 0.078 0.139 0.259 
2.5 Grade 3 hepatic 0.052 0.078 0.139 0.259 
2.5 Grade 4 thrombocytopenia 0.052 0.077 0.138 0.258 

The dose level closest to the toxicity target (0.25) is in bold.

Responses and time to event analysis during phase 1 are reported in Table 3 and Table 4.

Table 3

Best responses to combination treatment

Relapsed after lenalidomideRefractory to lenalidomideRefractory to both lenalidomide and bortezomib
1 mg (N = 4)1.5 mg (N = 4)2 mg (N = 4)2.5 mg (N = 55)2.5 mg (N = 18)2.5 mg (N = 37)2.5 mg (N = 16)
Response        
 Complete or partial 1 (25%) 2 (50%) 2 (50%) 28 (51%) 11 (61%) 17 (46%) 8 (50%) 
  Complete response — — — 
  Very good partial response — — — 10 
  Partial response 15 11 
 Minimal response — 11 
 Stable disease 15 10 
 Progressive disease — — 
Relapsed after lenalidomideRefractory to lenalidomideRefractory to both lenalidomide and bortezomib
1 mg (N = 4)1.5 mg (N = 4)2 mg (N = 4)2.5 mg (N = 55)2.5 mg (N = 18)2.5 mg (N = 37)2.5 mg (N = 16)
Response        
 Complete or partial 1 (25%) 2 (50%) 2 (50%) 28 (51%) 11 (61%) 17 (46%) 8 (50%) 
  Complete response — — — 
  Very good partial response — — — 10 
  Partial response 15 11 
 Minimal response — 11 
 Stable disease 15 10 
 Progressive disease — — 
Table 4

Time to event analysis

Median follow-up (months, range)Median PFS (months; 95% CI)Median OS (months; 95% CI)12-mo OS (95% CI)
All patients (N = 67) 15.0 (3.7-26.4) 8.6 (7.5-13.9) Not reached 65% (51-76%) 
Dose level 1, 1.5, 2 mg (N = 12) 24.1 (3.7-26.4) 4.6 (3.3-8.0) 9 (5.2-not reached) 44% (15-70%) 
Dose level 2.5 mg (N = 55) 14.8 (6.1-21.4) 10.4 (7.8-15.8) Not reached 69% (54-81%) 
 Relapsed after lenalidomide (N = 18) 12.7 (7.2-21.4) 15.7 (12.8-20.7) Not reached 88% (60-97%) 
 Refractory to lenalidomide (N = 37) 15.3 (6.1-21.4) 8.6 (7.5-13.9) Not reached 60% (41-75%) 
 Refractory to lenalidomide and bortezomib (N = 16) 15.8 (6.6-21.4) 8.6 (4.8-not reached) Not reached 67% (37-85%) 
Median follow-up (months, range)Median PFS (months; 95% CI)Median OS (months; 95% CI)12-mo OS (95% CI)
All patients (N = 67) 15.0 (3.7-26.4) 8.6 (7.5-13.9) Not reached 65% (51-76%) 
Dose level 1, 1.5, 2 mg (N = 12) 24.1 (3.7-26.4) 4.6 (3.3-8.0) 9 (5.2-not reached) 44% (15-70%) 
Dose level 2.5 mg (N = 55) 14.8 (6.1-21.4) 10.4 (7.8-15.8) Not reached 69% (54-81%) 
 Relapsed after lenalidomide (N = 18) 12.7 (7.2-21.4) 15.7 (12.8-20.7) Not reached 88% (60-97%) 
 Refractory to lenalidomide (N = 37) 15.3 (6.1-21.4) 8.6 (7.5-13.9) Not reached 60% (41-75%) 
 Refractory to lenalidomide and bortezomib (N = 16) 15.8 (6.6-21.4) 8.6 (4.8-not reached) Not reached 67% (37-85%) 

Phase 2

Fifty-five patients treated at the MTD (2.5 mg/d) were evaluated. Patients received a median of 6 cycles (range, 1-6 cycles). Five patients did not complete the assigned 6 cycles for toxicity: grade 3 cutaneous rash and grade 2 pancreatitis (1 patient), grade 2 bradycardia and dyspnea (1 patient), grade 5 sepsis (1 patient), grade 4 deep vein thrombosis (1 patient), and grade 3 liver failure (1 patient). Eight patients did not complete salvage therapy for PD, 1 patient for a concomitant mesothelioma, and 1 patient skipped the last cycle and started maintenance for medical reasons. Thirty-four patients started maintenance treatment; 13 of them discontinued therapy for PD and 2 for toxicity, including pulmonary embolism and limbic encephalitis (1 patient each) (Figure 1).

Figure 1

Flow diagram.

At the end of the first stage of the phase 2 portion, 2 CRs and 3 VGPRs were observed, allowing us to proceed with the second stage.

At least PR was achieved in 28/55 patients (51%), at least VGPR was achieved in 13/55 patients (24%), and immunofixation-negative CR was achieved in 3/55 patients (5%). A high proportion of patients achieved a clinical benefit, with at least a minimal response (MR) in 39/55 patients (71%) and at least stable disease in 54/55 patients (98%) (Table 3). The median time to at least PR was 1.83 months (range, 0.65-6.4 months); at least PR was achieved in 19 patients after 2 cycles, in 20 patients after 4 cycles, and in 23 patients after 6 cycles. The median duration of response for the 28 responding patients has not been achieved yet.

The median follow-up from study entry was 14.8 months (range, 6-21 months). At the time of the analysis, 40 patients were alive, 26 had progressed, and 15 had died from PD (10 patients), pneumonia and respiratory failure (1 patient), sudden death (1 patient), sepsis (1 patient), liver failure (1 patient), and mesothelioma (1 patient). The 1-year PFS was 48% (95% CI, 33%-62%), with a median of 10.4 months (95% CI, 7.9-15.8 months; Figure 2A). The 1-year OS was 69% (95% CI, 54%-81%), and the median value was not reached (Figure 2B).

Figure 2

Time-to-event analysis. (A) PFS in phase 2 patients. (B) OS in phase 2 patients.

Figure 2

Time-to-event analysis. (A) PFS in phase 2 patients. (B) OS in phase 2 patients.

Close modal

The 1-year PFS was 72% for patients relapsed after lenalidomide and 37% for those refractory to lenalidomide (P = .22; Figure 3A). The 1-year PFS was 68% in patients who achieved at least PR and 26% in those who achieved less than PR (P = .02; Figure 3B). The 1-year PFS was 47% in standard-risk and 35% in high-risk patients (P = .21; Figure 3C).

Figure 3

Progression-free survival. (A) PFS in phase 2 patients refractory (N = 18) or relapsed (N = 37) after lenalidomide. (B) PFS in phase 2 patients according to best response (treated as time-dependent covariate). (C) PFS in phase 2 patients with standard-risk or high-risk FISH abnormalities.

Figure 3

Progression-free survival. (A) PFS in phase 2 patients refractory (N = 18) or relapsed (N = 37) after lenalidomide. (B) PFS in phase 2 patients according to best response (treated as time-dependent covariate). (C) PFS in phase 2 patients with standard-risk or high-risk FISH abnormalities.

Close modal

In a multivariable analysis, older age negatively affected PFS (HR, 2.65; P = .035), and the achievement of PR confirmed its positive effect on PFS (HR, 0.38; P = .059) (Figure 3B). No differences according to FISH-defined risk were noted (HR, 1.36; P = .48).

At the MTD, the most frequent grade 3 to 4 adverse events were neutropenia (42%), thrombocytopenia (11%), anemia (9%), neurologic (7%), dermatologic reactions (7%), and infections (5%). Two grade 5 infections were reported (Table 5). Grade 3 peripheral neuropathy was detected in 2 patients. Dermatologic events were mild to moderate and were manageable with pomalidomide dose reduction and corticosteroids. Grade 4 deep vein thrombosis was reported in 1 patient, despite low-molecular-weight heparin prophylaxis. Pomalidomide dose was reduced in 17 patients for grade 4 hematologic toxicity (4 patients), grade 1 to 2 nonhematologic toxicity (2 patients), grade 3 to 4 nonhematologic toxicity (7 patients), and unknown causes (4 patients). During maintenance treatment, the frequency of new grade 3 to 4 adverse events was low and included grade 4 neutropenia (2 patients), pulmonary embolism (1 patient, while receiving aspirin), and grade 3 neurologic toxicity (vertigo, peripheral neuropathy, and limbic encephalitis, 1 patient each).

Table 5

Treatment-related adverse events during salvage therapy

EventsPhase 2 (N = 55)
Grade 1Grade 2Grade 3Grade 4Grade 5Total
Hematologic       
 Neutropenia 10 14 — 33 
 Thrombocytopenia 15 — 26 
 Anemia 10 22 — — 37 
Nonhematologic       
 Cardiologic — — — 
  Ischemia — — — — 
  Arrhythmia — — — — 
 Neurologic 11 — 21 
  Sensory neuropathy — — — 
  Neuralgia — — 
  Motor neuropathy — — — — 
  Tremor — — — — 
  Confusion — — — 
  Mood depression — — — — 
  Other — — — 
 Infective 14 24 
  Upper respiratory — — — 
  Pneumonia — 11 
  Sepsis — — — — 
  Other — — — 
 Gastrointestinal — 11 
  Diarrhea — — — — 
  Constipation — — — — 
  Nausea/Vomiting — — — — 
  Other — — 
 Hepatic/pancreatic — 
  Increased transaminase — — — 
  Liver failure — — — — 
  Pancreatitis — — — — 
 Vascular — 
  Deep-vein thrombosis — — — 
  Phlebitis — — — — 
 Systemic — 18 
  Fatigue — — 14 
  Fever — — — — 
  Drowsiness — — — — 
  Weight gain — — — — 
 Dermatologic — — 
  Rash — — — 
  Other — — — — 
Other — — 16 
EventsPhase 2 (N = 55)
Grade 1Grade 2Grade 3Grade 4Grade 5Total
Hematologic       
 Neutropenia 10 14 — 33 
 Thrombocytopenia 15 — 26 
 Anemia 10 22 — — 37 
Nonhematologic       
 Cardiologic — — — 
  Ischemia — — — — 
  Arrhythmia — — — — 
 Neurologic 11 — 21 
  Sensory neuropathy — — — 
  Neuralgia — — 
  Motor neuropathy — — — — 
  Tremor — — — — 
  Confusion — — — 
  Mood depression — — — — 
  Other — — — 
 Infective 14 24 
  Upper respiratory — — — 
  Pneumonia — 11 
  Sepsis — — — — 
  Other — — — 
 Gastrointestinal — 11 
  Diarrhea — — — — 
  Constipation — — — — 
  Nausea/Vomiting — — — — 
  Other — — 
 Hepatic/pancreatic — 
  Increased transaminase — — — 
  Liver failure — — — — 
  Pancreatitis — — — — 
 Vascular — 
  Deep-vein thrombosis — — — 
  Phlebitis — — — — 
 Systemic — 18 
  Fatigue — — 14 
  Fever — — — — 
  Drowsiness — — — — 
  Weight gain — — — — 
 Dermatologic — — 
  Rash — — — 
  Other — — — — 
Other — — 16 

In this study, we evaluated dosing, the safety profile, and the efficacy of the combination of PCP in patients who were relapsed/refractory to multiple lines of treatment, including lenalidomide. At the MTD (2.5 mg/d pomalidomide), the at least PR rate was 51%, and the median PFS was 10.4 months. Adverse events were mainly hematologic: the rate of grade 4 neutropenia was 16%, and the rate of grade 4 thrombocytopenia was 5%. A Bayesian adaptive design for dose finding was implemented. This approach is expected to replace classic dose-finding schemes because it enables more patients be treated at near-optimal doses while controlling excessive toxicities.

Patients who experience multiple relapses and become refractory to current salvage treatments have virtually no treatment options. Responses after relapse are generally short-lived, and outcomes can be affected by comorbidities, adverse chromosomal abnormalities, and the toxicity of the previous treatments.21-23  A recent survey on 286 relapsed myeloma patients who were refractory to bortezomib and relapsed/refractory to an immunomodulatory drug reported at least MR in 44% of patients, including 32% PR, and median event-free survival and OS of 5 and 9 months, respectively.3 

Initial reports demonstrated encouraging activity with pomalidomide alone or with low-dose dexamethasone in relapsed/refractory myeloma.6-12,24,25  In a phase 3 study comparing pomalidomide (4 mg/d for 21 days in a 28-day cycle) plus low-dose dexamethasone (160 mg monthly) with high-dose dexamethasone (480 mg monthly) in multirelapsed/refractory myeloma, median PFS was 3.9 vs 2 months (P < .001), and median OS was not reached vs 8.5 months (P < .001), in the pomalidomide and the high-dose dexamethasone groups, respectively.26 

In a previous study, pomalidomide-dexamethasone induced at least PR rate of 32%, at least MR rate of 47%, and median PFS of 4.8 months in patients refractory to lenalidomide.8  In another phase 2 trial, the at least PR rate was 40% in patients refractory to lenalidomide and 60% in those refractory to bortezomib.6  In our trial, the PCP regimen induced an at least PR rate of 51% and a clinical benefit, with an at least MR rate of 71%. These results are clinically meaningful in patients who had received most of the available therapies. Furthermore, patients were mostly refractory to lenalidomide (67%) or bortezomib (39%), and a subgroup was refractory to both these agents (29%). At least PR was reported in 46% of patients refractory to lenalidomide and in 50% of those refractory to bortezomib-lenalidomide. The addition of an alkylating agent to novel drugs has demonstrated an additive positive effect.12,27,28  In our study, median PFS was 10.4 months and median OS was not reached, supporting the hypothesis that cyclophosphamide increased the clinical efficacy of pomalidomide.

The results obtained with PCP are impressive considering the dose and schedule used compared with the most recent trials.9,10,12,29  In our study, the monthly dose of cyclophosphamide was 700 mg, which is inferior to the most commonly used doses. The monthly dose of pomalidomide (2.5 mg daily, 70 mg in a 28-day cycle) is slightly inferior to the standard schedule (4 mg/d for 21 days in a 28-day cycle, 84 mg monthly). This suggests that the positive results obtained with PCP are mainly related to the synergistic activity of the combination. Because PCP was well-tolerated, increasing dose intensity of pomalidomide to 4 mg for 21 days might further improve its efficacy without a major increase in toxicity.9,10,12,29  We planned 6 cycles of PCP, but prolonging the treatment up to 9 cycles might also improve response rate and outcome. Future phase 3 trials should investigate higher doses of pomalidomide (4 mg/day) in a less-intensive 21-day schedule to minimize both the acute and the cumulative toxicity.

A dose-response relationship of pomalidomide is difficult to establish because the trial was not powered to evaluate it. Although the number of patients enrolled at different dose levels was quite limited, 2.5 mg/d pomalidomide induced better responses and a significant prolonged PFS (P = .05; data not shown) compared with lower doses.

The quality of response and the amount of cytoreduction seem to be predictive factors of longer remission duration. In our study, patients with at least PR showed a significantly prolonged PFS compared with patients achieving less than PR, suggesting a potential clinical benefit of a more intense cytoreduction in fit patients. Older age negatively affects PFS, demonstrating that dose adjustments are needed in vulnerable patients. Chromosomal abnormalities are the major prognostic factors for MM. A quarter of the patients in this study were classified as high risk at enrolment. PFS was not significantly different between standard- and high-risk patients, but the numbers are too limited and larger series are needed to confirm these preliminary findings.

The most common toxicities were hematologic, with grade 3 to 4 toxicities occurring in 25 patients (45%) at the MTD. Neutropenia was seen mainly in the first cycles, suggesting a concomitant role of the disease and the toxicity of the regimen. Hematologic toxicities were consistent with previous studies in which pomalidomide-dexamethasone induced a rate of grade 3 to 4 adverse events ranging from 38% to 53%.6.8 Similarly, hematologic toxicity reported with PCP was comparable to the rates reported with lenalidomide-dexamethasone (52%).30  These data suggest that cyclophosphamide at 50 mg every other day does not significantly increase hematologic toxicity.

The most frequent grade 3 to 4 nonhematologic toxicities were neurologic events, infections, and dermatologic reactions. The incidence of treatment-related peripheral neuropathy was low (4%), particularly when compared with bortezomib or thalidomide. In our study, grade 3 to 5 infections were noted in 9% of patients. In these patients, antibiotic prophylaxis may be recommended. A careful management of fever and neutropenia with the prompt institution of antibiotics is also suggested to reduce the incidence of infections. The incidence of grade 3 to 4 thromboembolic events was low (4%), supporting the need for anticoagulant prophylaxis.

This is the first study to establish that a novel combination, PCP, induces encouraging responses and outcomes in refractory MM. Data from this phase 1/2 study justify further exploration of this combination.

The online version of this article contains a data supplement.

There is an Inside Blood commentary on this article in this issue.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank the patients who took part in the study; the nurses, D’Ambrosio Luisella and Mastropasqua Gianmauro; the data managers, Antonella Fiorillo and Jessica Mastrovito; and the editorial assistant, Giorgio Schirripa.

The study was supported by Fondazione Neoplasie Sangue Onlus and Celgene.

Contribution: A.P., M.B., and P.C. designed the study, supervised its conduct and data analysis; A.L. and A.P. wrote the manuscript; A.L., V.M., S.B., D.R., C.C., R.M., M.G., M.M., G.L.V., N.G., V.M., T.G., D.R-S., P.O., A.S., A.M.C., and P.C. provided study material or recruited patients; I.B. performed statistical analysis; and all the authors had access to, commented on, and approved the final manuscript.

Conflict-of-interest disclosure: A.L. has received honoraria from Celgene and Janssen-Cilag. S.B. has received honoraria from Celgene, Janssen-Cilag, and Novartis and served on the advisory committee for Merck Sharp & Dohme. M.B. has received research support from and served on the scientific advisory board for Celgene and Janssen-Cilag. A.P. has received honoraria and consultancy fees from Celgene, Janssen-Cilag, Bristol-Myers Squibb, Millennium, Amgen, and Onyx. The remaining authors declare no competing financial interests.

Correspondence: Antonio Palumbo, Myeloma Unit, Division of Hematology, Azienda Ospedaliera Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Torino, Italy; e-mail: appalumbo@yahoo.com.

1
Palumbo
 
A
Anderson
 
K
Multiple myeloma.
N Engl J Med
2011
, vol. 
364
 
11
(pg. 
1046
-
1060
)
2
Kumar
 
SK
Rajkumar
 
SV
Dispenzieri
 
A
, et al. 
Improved survival in multiple myeloma and the impact of novel therapies.
Blood
2008
, vol. 
111
 
5
(pg. 
2516
-
2520
)
3
Kumar
 
SK
Lee
 
JH
Lahuerta
 
JJ
, et al. 
International Myeloma Working Group
Risk of progression and survival in multiple myeloma relapsing after therapy with IMiDs and bortezomib: a multicenter international myeloma working group study.
Leukemia
2012
, vol. 
26
 
1
(pg. 
149
-
157
)
4
Bartlett
 
JB
Dredge
 
K
Dalgleish
 
AG
The evolution of thalidomide and its IMiD derivatives as anticancer agents.
Nat Rev Cancer
2004
, vol. 
4
 
4
(pg. 
314
-
322
)
5
Schey
 
S
Ramasamy
 
K
Pomalidomide therapy for myeloma.
Expert Opin Investig Drugs
2011
, vol. 
20
 
5
(pg. 
691
-
700
)
6
Lacy
 
MQ
Hayman
 
SR
Gertz
 
MA
, et al. 
Pomalidomide (CC4047) plus low-dose dexamethasone as therapy for relapsed multiple myeloma.
J Clin Oncol
2009
, vol. 
27
 
30
(pg. 
5008
-
5014
)
7
Lacy
 
MQ
Allred
 
JB
Gertz
 
MA
, et al. 
Pomalidomide plus low-dose dexamethasone in myeloma refractory to both bortezomib and lenalidomide: comparison of 2 dosing strategies in dual-refractory disease.
Blood
2011
, vol. 
118
 
11
(pg. 
2970
-
2975
)
8
Lacy
 
MQ
Hayman
 
SR
Gertz
 
MA
, et al. 
Pomalidomide (CC4047) plus low dose dexamethasone (Pom/dex) is active and well tolerated in lenalidomide refractory multiple myeloma (MM)
Leukemia
2010
, vol. 
24
 
11
(pg. 
1934
-
1939
)
9
Leleu
 
X
Attal
 
M
Arnulf
 
B
, et al. 
 
High Response Rates to Pomalidomide and Dexamethasone in Patients with Refractory Myeloma, Final Analysis of IFM 2009-02. In: 53rd ASH Annual Meeting and Exposition; December 10-13, 2011; San Diego, CA. Abstract 812
10
Richardson
 
PG
Siegel
 
D
Vij
 
R
, et al. 
 
Randomized, Open Label Phase 1/2 Study of Pomalidomide (POM) Alone or in Combination with Low-Dose Dexamethasone (LoDex) in Patients (Pts) with Relapsed and Refractory Multiple Myeloma Who Have Received Prior Treatment That Includes Lenalidomide (LEN) and Bortezomib (BORT): Phase 2 Results. In: 53rd ASH Annual Meeting and Exposition; December 10-13, 2011; San Diego, CA. Abstract 634
11
McCarthy
 
DA
Macey
 
MG
Streetly
 
M
Schey
 
SA
Brown
 
KA
The neutropenia induced by the thalidomide analogue CC-4047 in patients with multiple myeloma is associated with an increased percentage of neutrophils bearing CD64.
Int Immunopharmacol
2006
, vol. 
6
 
7
(pg. 
1194
-
1203
)
12
Schey
 
SA
Morgan
 
GJ
Ramasamy
 
K
, et al. 
The addition of cyclophosphamide to lenalidomide and dexamethasone in multiply relapsed/refractory myeloma patients; a phase I/II study.
Br J Haematol
2010
, vol. 
150
 
3
(pg. 
326
-
333
)
13
Cancer Therapy Evaluation Program
 
Common terminology criteria for adverse events, version 3.0, DCTD, NCI, NIH, DHHS March 31, 2003.http://ctep.cancer.gov.
14
Kyle
 
RA
Rajkumar
 
SV
Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma.
Leukemia
2009
, vol. 
23
 
1
(pg. 
3
-
9
)
15
Palumbo
 
A
Rajkumar
 
SV
Dimopoulos
 
MA
, et al. 
International Myeloma Working Group
Prevention of thalidomide- and lenalidomide-associated thrombosis in myeloma.
Leukemia
2008
, vol. 
22
 
2
(pg. 
414
-
423
)
16
O’Quigley
 
J
Pepe
 
M
Fisher
 
L
Continual reassessment method: a practical design for phase 1 clinical trials in cancer.
Biometrics
1990
, vol. 
46
 
1
(pg. 
33
-
48
)
17
O’Quigley
 
J
Zohar
 
S
Experimental designs for phase I and phase I/II dose-finding studies.
Br J Cancer
2006
, vol. 
94
 
5
(pg. 
609
-
613
)
18
Zohar
 
S
Chevret
 
S
The continual reassessment method: comparison of Bayesian stopping rules for dose-ranging studies.
Stat Med
2001
, vol. 
20
 
19
(pg. 
2827
-
2843
)
19
Zohar
 
S
Latouche
 
A
Taconnet
 
M
Chevret
 
S
Software to compute and conduct sequential Bayesian phase I or II dose-ranging clinical trials with stopping rules.
Comput Methods Programs Biomed
2003
, vol. 
72
 
2
(pg. 
117
-
125
)
20
Kaplan
 
EL
Meier
 
P
Nonparametric estimation from incomplete observations.
J Am Stat Assoc
1958
, vol. 
53
 
282
(pg. 
457
-
481
)
21
Augustson
 
BM
Begum
 
G
Dunn
 
JA
, et al. 
Early mortality after diagnosis of multiple myeloma: analysis of patients entered onto the United kingdom Medical Research Council trials between 1980 and 2002—Medical Research Council Adult Leukaemia Working Party.
J Clin Oncol
2005
, vol. 
23
 
36
(pg. 
9219
-
9226
)
22
Quach
 
H
Mileshkin
 
L
Seymour
 
JF
, et al. 
Predicting durable remissions following thalidomide therapy for relapsed myeloma.
Leuk Lymphoma
2009
, vol. 
50
 
2
(pg. 
223
-
229
)
23
Gertz
 
MA
Kumar
 
S
Lacy
 
MQ
, et al. 
Stem cell transplantation in multiple myeloma: impact of response failure with thalidomide or lenalidomide induction.
Blood
2010
, vol. 
115
 
12
(pg. 
2348
-
2353, quiz 2560
)
24
Schey
 
SA
Fields
 
P
Bartlett
 
JB
, et al. 
Phase I study of an immunomodulatory thalidomide analog, CC-4047, in relapsed or refractory multiple myeloma.
J Clin Oncol
2004
, vol. 
22
 
16
(pg. 
3269
-
3276
)
25
Streetly
 
MJ
Gyertson
 
K
Daniel
 
Y
Zeldis
 
JB
Kazmi
 
M
Schey
 
SA
Alternate day pomalidomide retains anti-myeloma effect with reduced adverse events and evidence of in vivo immunomodulation.
Br J Haematol
2008
, vol. 
141
 
1
(pg. 
41
-
51
)
26
Dimopoulos
 
MA
Lacy
 
MQ
Moreau
 
P
, et al. 
 
Pomalidomide in Combination with Low-Dose Dexamethasone: Demonstrates a Significant Progression Free Survival and Overall Survival Advantage, in Relapsed/Refractory MM: A Phase 3, Multicenter, Randomized, Open-Label Study. In: 54th ASH Annual Meeting and Exposition; December 8-11, 2012; Atlanta, GA. Abstract LBA-6
27
Reece
 
D
Masih-Khan
 
E
Khan
 
A
, et al. 
 
Phase I-II Trial of Oral Cyclophosphamide, Prednisone and Lenalidomide (Revlimid®) (CPR) for the Treatment of Patients with Relapsed and Refractory Multiple Myeloma. In: 51st ASH Annual Meeting and Exposition; December 5-8, 2009; New Orleans, LA. Abstract 1874
28
Sidra
 
G
Williams
 
CD
Russell
 
NH
Zaman
 
S
Myers
 
B
Byrne
 
JL
Combination chemotherapy with cyclophosphamide, thalidomide and dexamethasone for patients with refractory, newly diagnosed or relapsed myeloma.
Haematologica
2006
, vol. 
91
 
6
(pg. 
862
-
863
)
29
Baz
 
R
Shain
 
KH
Alsina
 
M
, et al. 
 
Oral Weekly Cyclophosphamide in Combination with Pomalidomide and Dexamethasone for Relapsed and Refractory Myeloma: Report of the Dose Escalation Cohort. In: 54th ASH Annual Meeting and Exposition; December 8-11, 2012; Atlanta, GA. Abstract 4062
30
Weber
 
DM
Chen
 
C
Niesvizky
 
R
, et al. 
Multiple Myeloma (009) Study Investigators
Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America.
N Engl J Med
2007
, vol. 
357
 
21
(pg. 
2133
-
2142
)

Author notes

P.C. and A.P. contributed equally to this study.

Sign in via your Institution