Inotuzumab Ozogamicin in Adults With Relapsed or Refractory CD22-positive Acute Lymphoblastic Leukemia: A Phase 1/2 Study
Inotuzumab Ozogamicin in Adults With Relapsed or Refractory CD22-positive Acute Lymphoblastic Leukemia: A Phase 1/2 Study
Inotuzumab Ozogamicin in Adults With Relapsed or Refractory CD22-positive Acute Lymphoblastic Leukemia: A Phase 1/2 Study
Daniel J. DeAngelo,1 Wendy Stock,2 Anthony S. Stein,3 Andrei Shustov,4 Michaela Liedtke,5 Charles A. Schiffer,6 Erik Vandendries,7
Katherine Liau,7 Revathi Ananthakrishnan,7 Joseph Boni,8 A. Douglas Laird,9 Luke Fostvedt,9 Hagop M. Kantarjian,10 and Anjali S. Advani11
1
Dana-Farber Cancer Institute, Boston, MA; 2University of Chicago Comprehensive Cancer Center, Chicago, IL; 3City of Hope, Duarte, CA; 4Division of Hematology, University
of Washington, Seattle, WA; 5Stanford Cancer Institute, Stanford, CA; 6Karmanos Cancer Institute, Detroit, MI; 7Pfizer Inc, Cambridge, MA; 8Pfizer Inc, Collegeville, PA; 9Pfizer
Inc, La Jolla, CA; 10MD Anderson Cancer Center, Houston, TX; and 11Cleveland Clinic, Cleveland, OH
This study evaluated the safety, antitumor activity, pharmacokinetics, and pharmacodynamics
Key Points
of inotuzumab ozogamicin (InO) for CD22-positive relapsed/refractory acute lymphoblastic
• Weekly InO 1.8 mg/m2 leukemia. In phase 1, patients received InO 1.2 (n 5 3), 1.6 (n 5 12), or 1.8 (n 5 9) mg/m2
per cycle is associated per cycle on days 1, 8, and 15 over a 28-day cycle (#6 cycles). The recommended phase 2 dose
with manageable toxic-
(RP2D) was confirmed (expansion cohort; n 5 13); safety and activity of InO were assessed in
ities and encouraging
patients receiving the RP2D in phase 2 (n 5 35) and in all treated patients (n 5 72). The RP2D
activity in patients with
was 1.8 mg/m2 per cycle (0.8 mg/m2 on day 1; 0.5 mg/m2 on days 8 and 15), with reduction to
relapsed/refractory
1.6 mg/m2 per cycle after complete remission (CR) or CR with incomplete marrow recovery (CRi).
ALL.
Treatment-related toxicities were primarily cytopenias. Four patients experienced treatment-
• Achievement of MRD
related venoocclusive disease/sinusoidal obstruction syndrome (VOD/SOS; 1 fatal). Two
negativity and disease
VOD/SOS events occurred during treatment without intervening transplant; of 24 patients
burden was not corre-
proceeding to poststudy transplant, 2 experienced VOD/SOS after transplant. Forty-nine (68%)
lated; InO may thus be
patients had CR/CRi, with 41 (84%) achieving minimal residual disease (MRD) negativity. Median
effective regardless of
baseline disease progression-free survival was 3.9 (95% confidence interval, 2.9-5.4) months; median overall
severity. survival was 7.4 (5.7-9.2) months for all treated patients, with median 23.7 (range, 6.8-29.8)
months of follow-up for all treated patients alive at data cutoff. Achievement of MRD negativity
was associated with higher InO exposure. InO was well tolerated and demonstrated high single-
agent activity and MRD-negativity rates. This trial was registered at www.clinicaltrials.gov as
#NCT01363297.
Introduction
Acute lymphoblastic leukemia (ALL) is a heterogeneous malignancy with an estimated incidence of 6250
newly diagnosed cases in 2015.1,2 Although the majority (58%) of these cases were estimated to be among
pediatric and young adult patients aged ,20 years, 26% were among adults $45 years old.1,2
Current treatments for adults with newly diagnosed B-cell ALL yield complete remission (CR) rates
ranging from 60% to 90%3-12; however, many patients eventually relapse and only ;30% to 50%
maintain extended disease-free survival for $3 years.6-10 Moreover, overall survival (OS) rate at 5 years after
relapse has been reported to be only 7%.13 For B-cell ALL that has relapsed or is refractory to standard
chemotherapy, CR rates decrease with successive salvage treatments (eg, ;30% to 40% in first and
;10% to 25% in second or later salvage).3,7,14 Because CR is required for ensuing allogeneic stem cell
Submitted 6 October 2016; accepted 27 April 2017. DOI 10.1182/ © 2017 by The American Society of Hematology
bloodadvances.2016001925.
The full-text version of this article contains a data supplement.
27 JUNE 2017 x VOLUME 1, NUMBER 15 INOTUZUMAB OZOGAMICIN FOR CD22-POSITIVE ALL 1169
DeANGELO et al
Characteristic 1.2 mg/m2 (n 5 3) 1.6 mg/m2 (n 5 12) 1.8 mg/m2 (n 5 9) Dose expansion 1.8 mg/m2 (n 5 13) Phase 2 part (n 5 35) All treated (n 5 72)
Age, median, y (range) 64.0 (62-65) 43 (23-69) 42 (23-67) 57 (24-75) 34 (20-79) 45 (20-79)
,65, n (%) 2 (67) 10 (83) 8 (89) 10 (77) 31 (89) 61 (85)
4 0 0 0 0 2 (6) 2 (3)
5 0 1 (8) 1 (11) 1 (8) 1 (3) 4 (6)
.5 0 0 0 0 3 (9) 3 (4)
Median (range) duration since primary diagnosis, y 1.5 (0.5-4.9) 2.1 (0.2-6.6) 1.1 (0.4-20.4) 1.6 (0.2-4.9) 2.5 (0.4-16.0) 1.7 (0.2-20.4)
Response to last induction therapy, n (%)
Cytogenetics, n (%)
Median (range) CD221,‖ % blasts 99.6 (96-100) 98.7 (31-100) 98.4 (97-100) 95.1 (52-100) 99.2 (78-100) 99.0 (31-100)
BM blasts, n (%){
,50% 0 4 (33) 1 (11) 1 (8) 5 (14) 11 (15)
Thrombocytopenia* 2 (67) 1 (33) 4 (33) 3 (25) 4 (44) 4 (44) 4 (31) 4 (31) 12 (34) 12 (34) 26 (36) 24 (33)
Neutropenia†,‡ 1 (33) 1 (33) 2 (17) 2 (17) 5 (56) 5 (56) 3 (23) 3 (23) 9 (26) 7 (20) 20 (28) 18 (25)
AST increased 0 0 3 (25) 0 5 (56) 0 3 (23) 0 8 (23) 2 (6) 19 (26) 2 (3)
ALT increased 0 0 1 (8) 1 (8) 1 (11) 1 (11) 1 (8) 0 5 (14) 0 8 (11) 2 (3)
Pyrexia 0 0 0 0 1 (11) 0 2 (15) 0 5 (14) 0 8 (11) 0
In a futility IA of 10 evaluable patients receiving the RP2D in 1 had VOD/SOS ongoing at the time of death due to PD). Of 24
the phase 1 dose-escalation part (see supplemental Materials), $1 patients who proceeded to HSCT, 2 experienced VOD/SOS after
achieved CR/CRi (n 5 7). Per protocol, 13 additional patients were poststudy HSCT. These 2 patients received pretransplant condition-
enrolled in the dose-expansion cohort to confirm the RP2D, among ing with cyclophosphamide plus total body irradiation (TBI) and
whom no DLTs were observed. Six (46% [95% CI, 19% to 75%]) of etoposide plus fractionated TBI.
these patients achieved CR/CRi, of whom 5 (83%) achieved MRD Of all treated patients, 12 (17%) permanently discontinued treatment
negativity. In a futility IA of the first 12 evaluable patients receiving due to TEAEs, including ascites (n 5 2) and liver-related TEAEs
the RP2D in stage 1 of phase 2, $2 achieved CR/CRi; per protocol, (elevated ALT, AST, alkaline phosphatase [AP], GGT, and VOD [n 5 1
23 patients were enrolled in stage 2 without interruption, for a total each]; supplemental Table 2). Seven patients (10%) reduced dose due
of 35 patients in phase 2 of the study. to TEAEs, most commonly thrombocytopenia (n 5 3) and neutropenia
Safety and tolerability (n 5 3); 37 had dose delays due to TEAEs, most commonly thrombo-
Among all treated patients (n 5 72), the most frequent treatment- cytopenia (n 5 10), AST increased (n 5 9), neutropenia (n 5 10), and
related treatment-emergent AEs (TEAEs; $15% of patients) were ALT increased (n 5 6; supplemental Table 2).
thrombocytopenia (any grade, 36%; grade $3, 33%), neutropenia Fifty-four deaths (75%) occurred, including 12 during treatment and
(28%; 25%), aspartate aminotransferase (AST) increased (26%; up to 42 days after last dose and 42 during follow-up. Causes
3%), nausea (21%; 0%), vomiting (17%; 1%), fatigue (15%; 0%), of death during treatment included PD (n 5 9), septic shock,
and febrile neutropenia (15%; 13%; Table 3). The incidence of all- pneumonia, and subdural hematoma (n 5 1 each).
causality TEAEs followed a similar pattern (supplemental Table 1).
The most common serious TEAE was febrile neutropenia (22%). Efficacy
Treatment-related hepatic TEAEs included elevated AST (any Of the 35 patients receiving the RP2D in phase 2, 24 (69%)
grade, 26%; grade $3, 3%), g-glutamyltransferase (GGT: 13%; achieved CR/CRi (1-sided P , .0001 for the null hypothesis: CR/
1%), alanine aminotransferase (ALT; 11%; 3%), and hyperbilirubine- CRi rate #20%); therefore, the primary phase 2 endpoint (CR/CRi)
was met (Table 4). Potential prognostic factors (supplemental
mia (10%; 0%). Four VOD/SOS cases were reported (1 during phase
1 dose expansion [1.8 mg/m2] and 3 during phase 2), 2 confirmed by Table 3) were also assessed.
biopsy; all were considered treatment related and 1 was fatal. One Among all patients treated (n 5 72), 49 (68%) achieved CR/CRi
VOD/SOS event occurred during treatment (patient recovered with (CR, n 5 23; CRi, n 5 26; Table 4), with a median time to CR/CRi
sequelae of ascites and was able to proceed to allogeneic HSCT of 27.0 (range, 15-91) days. The median DOR was 4.6 (95% CI,
without further liver toxicity); another occurred during follow-up 3.8-6.6) months (Figure 1A). Ten patients who initially had CRi later
without intervening HSCT, shortly after starting maintenance therapy achieved CR during treatment. Of the 49 patients with CR/CRi,
with ponatinib and continued until death due to pneumonia. Three of 41 (84%) achieved MRD negativity (median time to MRD negativity,
the 4 patients with VOD/SOS had received defibrotide (2 recovered; 29.0 [range, 21-141] days); 18 (37%) relapsed during treatment or
Hematologic remission,
n (%) (95% CI)*
CR/CRi 2 (67) (9-99) 9 (75) (43-95) 8 (89) (52-100) 6 (46) (19-75) 24 (69) (51-83)† 49 (68) (56-79)
CR 1 (33) 7 (58) 3 (33) 2 (15) 10 (29) 23 (32)
CR 4.6 NR (0.7-NR) 15.2 (1.2-15.2) 6.1 (2.1-10.0) 2.8 (0.9-5.5) 4.6 (2.2-15.2)
CRi 4.8 NR (NR-NR) 7.0 (1.7-NR) NR (2.8-NR) 4.0 (1.2-6.6) 4.8 (3.8-9.0)
MRD negativity among
responders,{ n (%) (95% CI)
Median (range) time to 98.5 (98-99) 32.0 (22-64) 30.0 (22-141) 25.0 (21-134) 25.5 (21-80) 29.0 (21-141)
MRD negativity, d
HSCT rate
Patients proceeding to 0 9 (75) 4 (44) 3 (23) 8 (23) 24 (33)
poststudy HSCT, n (%)
Median (range) time to — 36 (20-60) 61.5 (41-84) 77 (55-90) 40 (27-148) 45.5 (20-148)
HSCT, d
PFS‡
Probability of PFS at 0 0.46 (0.20-0.70) 0.44 (0.10-0.70) 0.15 (0-0.40) 0.08 (0-0.20) 0.20 (0.10-0.30)
12 mo (95% CI)
OS‡
Events, n (%) 3 (100) 6 (50) 6 (67) 10 (77) 29 (83) 54 (75)
Median (95% CI), mo 9.2 (2.1-11.3) NR (2.6-NR) 16.5 (2.6-NR) 5.8 (3.5-10.8) 6.4 (4.5-7.9) 7.4 (5.7-9.2)
1173
within 30 days of the last dose. Median DOR and MRD-negativity decreased with time on treatment (;50% at C4D1 compared with
rates among patients achieving CRi, CR, and CR/CRi were similar C1D1 [data not shown]); this decrease tended to be less in patients
(Table 4). who had relatively higher proportions of leukemic BM blasts,
suggesting a direct relationship between the extent of InO elimination
For all treated patients, median PFS and OS were 3.9 (95% CI, 2.9-
and BM blast level.
5.4) and 7.4 (95% CI, 5.7-9.2) months; the Kaplan-Meier estimated
probabilities of PFS and OS at 12 months were 0.20 (95% CI, 0.10- BM blasts and MRD negativity
0.30) and 0.30 (95% CI, 0.20-0.40), respectively (Figure 1B-C).
The effect of BM blast count or salvage status on PFS is shown in An analysis of dose-normalized Cmax and Cmin by MRD-negativity
supplemental Table 4. status indicated that patients achieving MRD negativity tended to
have higher peak and trough InO concentrations throughout treatment
In all treated patients, 24 (33%) proceeded to poststudy HSCT vs those who did not (Figure 3A). In contrast, achievement of MRD
(median time to HSCT, 45.5 [range, 20-148] days), nearly all of negativity appeared to be unrelated to baseline disease burden
whom (n 5 22/24 [92%]) had achieved CR/CRi before HSCT. Of (percentage baseline BM blasts). Evidence supporting this is provided
the 2 patients who proceeded to HSCT without achieving CR/CRi, by plots of baseline BM blast percentages determined for individual
1 had a partial response and 1 had disease resistant to InO treatment patients at each protocol-specified BM aspirate collection time
and initiated new (non-InO) induction therapy before HSCT. Before (Figure 3B), which show that line profiles for patients achieving
HSCT, most patients (n 5 15/24 [63%]) had received fludarabine- MRD negativity (green lines) originated across a range of baseline
and/or TBI-based conditioning regimens (n 5 17/24 [71%]); only 1 BM blast percentages. This lack of a relationship between the
had received dual alkylator conditioning (cyclophosphamide, thiotepa, percentage of BM blasts at baseline and achievement of MRD
and fludarabine). Of the 24 patients who had poststudy HSCT, 12 negativity was also apparent from the serum InO concentration time
subsequently died (2 died due to relapse/PD). Seven of the 12 course during cycles 1 through 4 (Figure 3C).
patients died #100 days after poststudy HSCT (1 died due to
relapse/PD). Other causes of death after poststudy HSCT were Baseline mRNA levels in peripheral whole blood and
sepsis (n 5 4), complications of HSCT, gut graft-versus-host relationship with clinical outcome
disease/liver dysfunction, hepatic failure/SOS, respiratory failure, Baseline levels of several mRNAs were significantly higher in patients
Klebsiella pneumoniae/liver graft-versus-host disease, and un- who subsequently achieved CR/CRi and MRD negativity vs those
known (each n 5 1). During the study, 4 patients had extramedullary who did not achieve CR/CRi, including mRNAs encoded by genes
disease that regressed, and 4 of the patients had extramedullary implicated in stress response/drug resistance (ABCG2, median sev-
disease that remained stable or increased in size. The 27 patients enfold higher [P 5 .0071]; BCL2L1, median 11-fold higher [P 5 .0055];
who achieved CR/CRi and did not have poststudy HSCT received FASLG, median fourfold higher [P 5 .036]). No relationship was
median 4 (range, 1-6) treatment cycles; 5 (19%) of these patients apparent between baseline peripheral blood CD22 mRNA levels
were aged $65 years; 15 (56%) received prestudy SCT; 21 (78%) and achieving CR/CRi and MRD negativity.
received $2 salvage treatments; 16 (59%) experienced PD/relapse
,1 month of discontinuing treatment; 3 (11%) died ,1 month of Peripheral blood CD22 mRNA levels were decreased by day 15
discontinuing treatment (1 due to relapse/PD); and 11 (41%) relative to baseline (median, 97% decrease; P , .0001), consistent
initiated a new induction systemic therapy during follow-up. with selective ablation of CD22-positive leukemic blasts by InO
(supplemental Figure 2). CD22 mRNA levels in patients who
Pharmacokinetics subsequently achieved CR/CRi and MRD negativity were lower by
day 15 vs those who did not achieve CR/CRi (P 5 .01). Comparable
Mean peak serum InO concentrations were observed at or near the
reductions and trends were evident for several other mRNAs
end of infusion; these generally increased with cycle number
encoding B-cell lineage markers, including PAX5, VPREB1, and
(supplemental Table 5). Unconjugated calicheamicin concentra-
EBF1 (data not shown). Conversely, mRNA levels for several genes
tions were below the LLOQ for most patients and time points.
implicated in stress response, drug resistance, and apoptotic
regulation were two- to fivefold higher by day 15 relative to baseline
Peripheral blood B-lymphocyte depletion
(ABCG2, BCL2L1, FASLG; all P , .0001). P values for these PG
and regeneration analyses were not adjusted for the potential for false discovery due
For all treated patients (n 5 72), InO treatment was accompanied to the large number of samples assessed; hence, these findings are
by a consistently rapid depletion followed by a slow regeneration of considered preliminary.
B-lymphocytes in peripheral blood during follow-up, with substantial
interpatient differences in regeneration rate (Figure 2A). A similar Discussion
rapid depletion followed by slow regeneration of B-lymphocytes was In this phase 1/2 trial of single-agent InO for relapsed/refractory
apparent regardless of InO dose, although few patients received the
CD22-positive ALL, the RP2D was 1.8 mg/m2 per cycle (0.8 mg/m2
lower doses (Figure 2B). [day 1]; 0.5 mg/m2 [days 8 and 15]) as a starting dose, with
InO elimination according to percentage of leukemic blasts in BM subsequent dose reduction to 1.6 mg/m2 per cycle upon achievement
(relative to total leukocytes) separated by dosing events is shown of CR/CRi. Toxicities associated with treatment were primarily
for individual patients in Figure 2C. As shown by the color gradient cytopenias and liver-related toxicities. Encouraging clinical activity
of the points (darker 5 higher percent blasts; lighter 5 lower was observed, with 68% of all treated patients achieving CR/CRi and
percent blasts), the percentage of leukemic blasts in BM decreased 33% of patients able to proceed to poststudy HSCT. Nevertheless,
over time and with each cycle, with very low percentages observed at remission was often transitory with 18 (37%) patients relapsing during
later time points. In general, the InO serum elimination rate constant treatment or within 30 days of the last dose; consolidation with HSCT
DOR probability
0.6
0.5
0.4
0.3
0.2
0.1
0.0
0 3 6 9 12 15 18 21 24 27 30
Months
Patients at risk, n
All Treated 49 31 18 13 10 9 7 7 1 1 0
Phase 2 part 24 13 5 2 0
B
1.0
n Median PFS (95% CI), mo
0.9
All treated 72 3.9 (2.9–5.4)
0.8 Phase 2 part 35 3.7 (2.6–4.7)
0.7
Probability of PFS
0.6
0.5
0.4
0.3
0.2
0.1
0.0
0 3 6 9 12 15 18 21 24 27 30
Months
Patients at risk, n
All Treated 72 43 24 17 11 10 7 7 3 1 0
Phase 2 part 35 21 9 5 1 0
C
1.0 n Median OS (95% CI), mo
0.9
All treated 72 7.4 (5.7–9.2)
0.8 Phase 2 part 35 6.4 (4.5–7.9)
0.7
Probability of OS
0.6
0.5
0.4
0.3
0.2
0.1
0.0
0 3 6 9 12 15 18 21 24 27 30
Months
Patients at risk, n
All Treated 72 58 42 28 19 14 12 12 7 2 0
Phase 2 part 35 28 19 9 4 0
27 JUNE 2017 x VOLUME 1, NUMBER 15 INOTUZUMAB OZOGAMICIN FOR CD22-POSITIVE ALL 1175
B 20
1.2 mg/m2
15
10
Lymphocytes in peripheral blood, 109/L
5
0
20
1.6 mg/m2
15
10
5
0
20
1.8 mg/m2
15
10
5
0
0 250 500 750
Days
InO treatment (n=72) Follow–up (n=24)
C 7.5
Cycle 1
Day 1
5.0
2.5
0.0
7.5
Cycle 1
Day 15
Log [InO], ng/mL
5.0
2.5
0.0
7.5
Cycle 2
Day 1
5.0
2.5
0.0
7.5
Cycle 2
Day 15
5.0
2.5
0.0
10 1000
Time postdose, h
Percent leukemic
BM aspirate at Screening Cycle1 Day 28 Cycle 2 Day 28 BM blasts 0 25 50 75
B
100
75
Baseline BM blasts, %
50
25
0
Screening Cycle 1 Cycle 2
Day 28 Day 28
Achievement of MRD Negativity Yes No
C
Cycle 1 Day 1 Cycle 1 Day 15 Cycle 2 Day 1 Cycle 2 Day 15 Cycle 4 Day 1
n=59 n=55 n=56 n=52 n=23
1000
Log [InO], ng/mL
10
e
e
st
st
st
st
st
h
h
Pr
Pr
Pr
Pr
Pr
Po
Po
Po
Po
Po
+2
+2
+2
+2
+2
27 JUNE 2017 x VOLUME 1, NUMBER 15 INOTUZUMAB OZOGAMICIN FOR CD22-POSITIVE ALL 1177
References
1. National Cancer Institute. The Surveillance, Epidemiology, and End Results (SEER) Program: Acute Lymphocytic Leukemia. http://seer.cancer.gov/
statfacts/html/alyl.html. Accessed March 7, 2016.
2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5-29.
3. Gökbuget N, Stanze D, Beck J, et al; German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia. 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.
4. Bassan R, Hoelzer D. Modern therapy of acute lymphoblastic leukemia. J Clin Oncol. 2011;29(5):532-543.
5. Rowe JM, Buck G, Burnett AK, et al; ECOG; MRC/NCRI Adult Leukemia Working Party. Induction therapy for adults with acute lymphoblastic leukemia:
results of more than 1500 patients from the international ALL trial: MRC UKALL XII/ECOG E2993. Blood. 2005;106(12):3760-3767.
6. Kantarjian H, Thomas D, O’Brien S, et al. Long-term follow-up results of hyperfractionated cyclophosphamide, vincristine, doxorubicin, and
dexamethasone (Hyper-CVAD), a dose-intensive regimen, in adult acute lymphocytic leukemia. Cancer. 2004;101(12):2788-2801.
7. Thomas X, Boiron JM, Huguet F, et al. Outcome of treatment in adults with acute lymphoblastic leukemia: analysis of the LALA-94 trial. J Clin Oncol.
2004;22(20):4075-4086.
8. Takeuchi J, Kyo T, Naito K, et al. Induction therapy by frequent administration of doxorubicin with four other drugs, followed by intensive consolidation and
maintenance therapy for adult acute lymphoblastic leukemia: the JALSG-ALL93 study. Leukemia. 2002;16(7):1259-1266.
9. Annino L, Vegna ML, Camera A, et al; GIMEMA Group. Treatment of adult acute lymphoblastic leukemia (ALL): long-term follow-up of the GIMEMA ALL
0288 randomized study. Blood. 2002;99(3):863-871.
10. Larson RA, Dodge RK, Burns CP, et al. A five-drug remission induction regimen with intensive consolidation for adults with acute lymphoblastic leukemia:
cancer and leukemia group B study 8811. Blood. 1995;85(8):2025-2037.
11. Goldstone AH, Richards SM, Lazarus HM, et al. In adults with standard-risk acute lymphoblastic leukemia, the greatest benefit is achieved from a
matched sibling allogeneic transplantation in first complete remission, and an autologous transplantation is less effective than conventional consolidation/
maintenance chemotherapy in all patients: final results of the International ALL Trial (MRC UKALL XII/ECOG E2993). Blood. 2008;111(4):1827-1833.
12. Petersdorf SH, Kopecky KJ, Head DR, et al. Comparison of the L10M consolidation regimen to an alternative regimen including escalating methotrexate/
L-asparaginase for adult acute lymphoblastic leukemia: a Southwest Oncology Group Study. Leukemia. 2001;15(2):208-216.
13. Fielding AK, Richards SM, Chopra R, et al; Medical Research Council of the United Kingdom Adult ALL Working Party; Eastern Cooperative Oncology
Group. Outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an MRC UKALL12/ECOG 2993 study. Blood. 2007;109(3):
944-950.
14. Tavernier E, Boiron JM, Huguet F, et al; GET-LALA Group; Swiss Group for Clinical Cancer Research SAKK; Australasian Leukaemia and Lymphoma
Group. Outcome of treatment after first relapse in adults with acute lymphoblastic leukemia initially treated by the LALA-94 trial. Leukemia. 2007;21(9):
1907-1914.
15. Thomas DA, Kantarjian H, Smith TL, et al. Primary refractory and relapsed adult acute lymphoblastic leukemia: characteristics, treatment results, and
prognosis with salvage therapy. Cancer. 1999;86(7):1216-1230.
16. Kebriaei P, Wilhelm K, Ravandi F, et al. Feasibility of allografting in patients with advanced acute lymphoblastic leukemia after salvage therapy with
inotuzumab ozogamicin. Clin Lymphoma Myeloma Leuk. 2013;13(3):296-301.
17. Alvarnas JC, Brown PA, Aoun P, et al. Acute Lymphoblastic Leukemia, Version 2.2015. J Natl Compr Canc Netw. 2015;13(10):1240-1279.
18. Kantarjian H, Thomas D, Jorgensen J, et al. Results of inotuzumab ozogamicin, a CD22 monoclonal antibody, in refractory and relapsed acute lymphocytic
leukemia. Cancer. 2013;119(15):2728-2736.
19. Thomas X. Profile of inotuzumab ozogamicin and its potential in the treatment of acute lymphoblastic leukemia. Blood Lymphat Cancer. 2014;4:1-8.
20. Yilmaz M, Richard S, Jabbour E. The clinical potential of inotuzumab ozogamicin in relapsed and refractory acute lymphocytic leukemia. Ther Adv
Hematol. 2015;6(5):253-261.
21. Poe JC, Tedder TF. CD22 and Siglec-G in B cell function and tolerance. Trends Immunol. 2012;33(8):413-420.
22. Haso W, Lee DW, Shah NN, et al. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood. 2013;121(7):
1165-1174.
27 JUNE 2017 x VOLUME 1, NUMBER 15 INOTUZUMAB OZOGAMICIN FOR CD22-POSITIVE ALL 1179