Significance
Moxetumomab pasudotox is a fusion protein of an anti-CD22 Fv and Pseudomonas exotoxin. It is highly active against leukemia in vitro but acute lymphoblastic leukemia (ALL) patients often are resistant. Studies with cultured cells showed resistance is caused by reduced diphthamide, the intracellular target of Pseudomonas exotoxin, but diphthamide is not reduced in most cells from most ALL patients. To study how resistance develops in animals, we injected ALL cells into mice and found that resistant cells occur in discrete bone marrow niches and contain major chromosomal and transcriptional changes. Mice pretreated with 5-azacytidine show greatly improved responses, supporting a trial of the combination in leukemia patients.
Keywords: acute lymphoblastic leukemia, CD22, immunotoxin, Moxetumomab pasudotox, 5-azacytidine
Abstract
Moxetumomab pasudotox (Moxe) is a chimeric protein composed of an anti-CD22 Fv fused to a portion of Pseudomonas exotoxin A and kills CD22-expressing leukemia cells. It is very active in hairy-cell leukemia, but many children with relapsed/refractory acute lymphoblastic leukemia (ALL) either respond transiently or are initially resistant. Resistance to Moxe in cultured cells is due to low expression of diphthamide genes (DPH), but only two of six ALL blast samples from resistant patients had low DPH expression. To develop a more clinically relevant model of resistance, we treated NSG mice bearing KOPN-8 or Reh cells with Moxe. More than 99.9% of the cancer cells were killed by Moxe, but relapse occurred from discrete bone marrow sites. The resistant cells would no longer grow in cell culture and showed major chromosomal changes and changes in phenotype with greatly decreased CD22. RNA deep sequencing of resistant KOPN-8 blasts revealed global changes in gene expression, indicating dedifferentiation toward less-mature B cell precursors, and showed an up-regulation of myeloid genes. When Moxe was combined with 5-azacytidine, resistance was prevented and survival increased to over 5 months in the KOPN-8 model and greatly improved in the Reh model. We conclude that Moxe resistance in mice is due to a new mechanism that could not be observed using cultured cells and is prevented by treatment with 5-azacytidine.
Treatment of childhood acute lymphoblastic leukemia (ALL) results in 80% long-term remissions (1). Treating relapsed ALL is a challenge and the likelihood of achieving a long-term remission decreases dramatically with each relapse (2). Consequently, novel therapies are needed to treat relapsed ALL more successfully (3). Emerging new treatment concepts include engineered chimeric antigen receptor (CAR) T cells (4–6), CD19-targeting BiTEs (7–9), B cell receptor (BCR)-signaling inhibitors (10), or antibody drug conjugates (3, 11).
The recombinant immunotoxin Moxetumomab pasudotox (Moxe) consists of a CD22-targeting antibody fragment and the Pseudomonas exotoxin A (12). After binding to CD22, Moxe is internalized, and is transported to the cytosol where it ADP ribosylates elongation factor 2 at a unique diphthamide residue (13). The ADP ribosylation leads to protein synthesis arrest and cell death. The diphthamide moiety (DPH) is generated by seven enzymes (DPH1 through DPH7). The homozygous deletion of the genes for DPH2, -4, or -5 (14) and DPH7/WDR85 (15) or a substantial decrease of DPH1 (16) or DPH4 (17) protein renders cells resistant to immunotoxins in vitro.
Moxe is highly active in 85% of patients with relapsed/refractory hairy-cell leukemia (HCL) (12) and in ∼30% of relapsed/refractory childhood ALL (18). Even-though CD22 is uniformly expressed (19), some HCL and two-thirds of ALL patients do not respond to Moxe. To study resistance to Moxe, three leukemia cell lines were treated in vitro until a resistant clone grew out (15–17). We found that resistant CA46 cells had a small chromosomal deletion containing WDR85; the deletion was preexisting in the population of cells. Resistance in KOPN-8 and HAL-01 was due to promoter silencing of DPH1 and DPH4, respectively. The resistance was associated with methylation of CpG islands in the specific gene promoters, which was partially reversed by treating the cells with the demethylating agent 5-azacytidine (5-AZA).
To identify mechanisms of resistance to Moxe in patients, we studied primary ALL blasts from children with CD22+ pre–B-ALL treated on the pediatric phase I trial (NCT01891981) and found resistance in most patients is not associated with low diphthamide gene expression. To understand Moxe-resistance in vivo, we then developed two ALL mouse models. In mice, we found resistant leukemia cells develop in discrete bone marrow (BM) niches and contain major chromosomal and transcriptional changes. Because these cells do not grow in culture when removed from mice, this mechanism could not have been discovered using cultured cells.
Results
Reduced DPH-Enzyme Expression Correlates with Resistance in Selected Patients.
We previously found that CD22-expressing leukemia cell lines that were repeatedly treated with Moxe in vitro develop resistance caused by the loss or a substantial reduction of one of the enzymes required for the diphthamide synthesis (15–17). To study whether primarily Moxe-resistant ALL blasts from patients are resistant due to low DPH expression, we analyzed their expression by RNA deep sequencing in eight primary patient samples (Fig. 1). Compared with the average expression in seven Moxe-sensitive cell lines, two of six nonresponders showed a two- to eightfold reduced expression of DPH4 (Fig. 1A). However, the BM-blasts of Moxe-sensitive patients also showed reduced DPH gene expression. To assess whether the decrease is functionally relevant, we compared the average DPH-expression of the sensitive cell lines to the previously established resistant cell clones (Fig. 1B). A 17-fold reduction of DPH1 in KOPN-8-R (16) and a 10-fold reduction of DPH4 in HAL-01-R (17) are sufficient to cause Moxe-resistance in vitro. The data indicate that for two patients, Moxe-resistance may have been due to reduced DPH4 expression. However, unlike as has been suggested by previous cell line studies, reduced DPH enzymes were likely not involved in the Moxe resistance of the other patients. Due to a lack of patient material, these data were not validated by an independent method.
KOPN-8 Relapse at Distinct BM-Sites Predates Systemic Relapse.
To investigate resistance in an animal setting, we developed a NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mouse model to study resistance in vivo, which we hypothesized would more closely resemble resistance in patients. NSG mice injected intravenously with 5 million KOPN-8 cells die from systemic leukemia after a median of 20 d. When treated with three doses of 0.4 mg/kg Moxe every other day from day 8, mice survived much longer but died from ALL after 40 d (Fig. 2A). The regrowing KOPN-8 cells are detected in the murine BM before they are detected in other organs (20). To assess response to Moxe, we monitored BM-disease burden after treatment (Fig. 2B). BM-infiltration on day 8 was 4%. On day 16, 4 d after the last dose of Moxe, no leukemia was detected in six of eight mice; in the two remaining mice, 1 in 100,000 events was KOPN-8. However, 10 d later, all mice relapsed with an average BM-infiltration rate of 0.05%, which rapidly rose to 43% on day 34.
To visualize where the relapsed cells are located, we generated a KOPN-8 subclone (KOPN-8L) expressing GFP and luciferase (Fig. S1). This clone closely resembles the parental KOPN-8 in growth kinetics and sensitivity to Moxe. Eight days after intravenous injection with KOPN-8L cells, mice showed bioluminescence over the spine, hip, and calvarium (Fig. 2C). Vehicle-treated mice showed a rapid increase in bioluminescence correlating with disease progression. In Moxe-treated mice, there was no bioluminescence detected 4 d after the last dose of Moxe. On day 29, relapse occurred as evidenced by recurrence of the bioluminescence in both mice. The mice were retreated with a second cycle of Moxe. The bioluminescence fell, but persisted over the right tibia in mouse 2, indicating local persistence of ALL cells. Mouse 2 was killed, the leg paraffin-embedded, and stained with a human-specific mitochondrial marker to identify human ALL. Dense ALL infiltration was detected in the distal tibia (Fig. 2 D and E), but no ALL signal was present in the proximal femur or in the surrounding soft tissue (Fig. 2 D and F). Immunohistochemistry confirmed local ALL persistence in the BM of the distal tibia. These results show that the majority of KOPN-8L cells die after initial Moxe treatment, but a few cells persist at an isolated BM site.
We then treated a group of KOPN-8L–bearing mice with two (Fig. 3A) or three (Fig. 3B) cycles of Moxe. The major signal on day 8 was found in close association with bone. Three doses of Moxe abrogated the bioluminescence, but all Moxe-treated mice relapsed. The bioluminescence in relapse appeared over discrete sites in bone, which were randomly distributed over the skeleton. When the bioluminescence after the first cycle reached levels similar to day 8, mice received a second cycle of five doses Moxe every other day. The systemic bioluminescence fell below the detection limit, but the signal over the local sites persisted, and then spread to produce a systemic bioluminescence in relapse. Mice not receiving a third cycle died from progressive disease. Of the three mice receiving a third cycle, two mice responded well but the systemic bioluminescence of the third mouse increased, indicating a generalized Moxe-resistance after the second cycle. These resistant cells were termed KOPN-8L-R.
To test whether localized resistance occurred in a second animal model, we generated a GFP/luficerase-expressing RehL subclone (Fig. S2). The Reh cell line was chosen, because they are equally sensitive to Moxe as KOPN-8 (20), but distinct from the KMT2A-ENL translocated KOPN-8, the Reh cells harbor a TEL-AML1 translocation. Mice bearing RehL where treated with repeated cycles of Moxe (Fig. 3C). Vehicle-treated mice progressed as expected. The bioluminescence of RehL-bearing micetreated with Moxe fell after the first treatment cycle but the signal persisted at isolated spots over bones. If not treated again, the Reh-bearing mice developed hind-limb paralysis and splenomegaly and died of progressive systemic leukemia (Fig. S3). Mice receiving a second cycle of Moxe showed stabilization of the bioluminescence and bioluminescence increased 2 d after the third Moxe cycle. The RehL cells after the third Moxe cycle were considered Moxe-resistant. The two xenografts show that although Moxe is very active initially, a resistant subclone develops and persists at local BM sites, where it survives treatment until a systemically resistant clone emerges after several cycles of treatment.
Leukemia Cells That Acquired Resistance Showed Profound Changes in Phenotype.
We attempted to grow the resistant KOPN-8L-R cells from mice in cell culture to facilitate their study. But the KOPN-8L-R cells would not grow, indicating that they had undergone major changes in the mice and suggesting that they had become dependent on the BM environment. Because in vitro generated Moxe-resistant cells show decreased expression of DPH enzymes, we assessed changes in mRNA levels by real-time PCR (Fig. 4A). The KOPN-8L-R had no decrease in any of the DPH-enzyme mRNAs; however, the CD22 mRNA was more than fourfold reduced. Flow cytometry confirmed the reduction of cell surface CD22 and of other B cell markers (Fig. 4B). The surface CD22 was reduced 2.5-fold (P < 0.0001), CD19 was reduced 2.1-fold (P = 0.0001), and CD10 was reduced 80-fold (P < 0.0001); surface CD28, however, increased 3.7-fold (P = 0.001). No CD22− population was detected (Fig. S4). The total CD22 protein by Western blot in FACS-sorted cells from five resistant and sensitive mice was reduced sevenfold (Fig. 4C). Because surface CD22 was reduced only twofold, these data suggest a profound depletion of intracellular CD22 needed for replenishing cell surface CD22 (21).
We also examined the resistant RehL-R cells that emerged after mice were treated with three cycles of Moxe and found they had acquired similar changes in surface markers as KOPN-8L-R (Fig. 4D). We found the resistant cells had a 1.6-fold decrease of CD22, a 1.6-fold decrease in CD19, a 1.8-fold decrease in CD10, and a 3.6-fold increase in CD33.
KOPN-8L-R Acquired Partial Moxe Resistance.
Because the resistant KOPN-8L-R cells alone cannot grow in culture, we hypothesized that they need BM support to survive. We investigated whether BM stromal cell lines would allow the resistant cells to grow and demonstrated that the murine calvarium-derived mesenchymal stromal cell line OP-9 rescued cell growth of KOPN-8L-R cells in vitro. Comparing KOPN-8L-R and KOPN-8L cells growing with OP-9 cell support, we found KOPN-8L-R had an IC50 of 3.0 ng/mL, which was 10-fold higher than the IC50 of the sensitive KOPN-8L cells with or without OP-9 cells (Fig. 4E). These data show that KOPN-8L-R cells need stromal cell support to survive and that the OP-9 support itself does not change the Moxe activity against sensitive KOPN-8L cells. This suggests that the KOPN-8L-R cells acquired a partial and nonreversible resistance to Moxe.
Moxe-Resistant ALL Showed Major Chromosomal Changes.
Because it is characteristic for ALL, we investigated whether the resistant cells had developed additional chromosomal alterations and found many changes (Fig. 4F). The parental KOPN-8L cells showed the expected karyotype of 44,XX, t(11;19), t(13;14), −13, and −14. The KOPN-8L-R cells showed additional changes including trisomy of chromosomes 6, 7, 8, and 20 and two additional t(5;8)-chromosomes, resulting in a 50,XX karyotype. These chromosomal changes could be responsible for the changes in phenotype of KOPN-8L-R. In addition, we found that the Moxe-resistant RehL-R cells from three individual mice also showed major changes in their karyotype by G-band staining (Fig. S5). Compared with the Moxe-sensitive RehL, the resistant subclone of one mouse had loss of chromosomes 2 and 18, while the subclone of another had gained chromosomes 2 and 16, and a subclone of a third mouse had gain of chromosomes 2 and 3 and loss of chromosomes 18, 19, 21, and 22. The predominant resistant subclone of each mouse presented distinct karyotype changes, which were different from the untreated cells growing in mice.
Because the KMT2A-rearrangement distinguishes the KOPN-8 cell line from other ALL subtypes (22), we determined if expression of the KMT2A-fusion gene changed significantly in resistant KOPN-8 cells. Analyzing the fusion transcript expression from the RNA deep-sequencing data, we found no difference between sensitive and resistant cells (Fig. 4G). On average, there were 13 uniquely aligned reads for KOPN-8L cells and 12.8 reads for KOPN-8L-R cells (P = 0.74). The major changes in karyotype did not alter the expression level of the KMT2A-fusion mRNA.
RNA Deep Sequencing Identifies Down-Regulation of B Cell-Specific Gene Sets.
To study effects of the chromosomal and other changes in the resistant ALL cells, we analyzed our RNA deep-sequencing data on flow-sorted ALL cells from five mice bearing KOPN-8L and KOPN-8L-R. The dot-plot in Fig. 5A shows the overall distribution of the average log2 (counts per million) for the 16,561 genes expressed in KOPN-8L and KOPN-8L-R. In accord with the flow data, there is a substantial decrease in mRNA for MME (=CD10, 16-fold) and MS4A1 (=CD20, 28-fold) and a less than threefold but significant (P < 0.001) decrease for CD19 (1.7-fold), CD22 (2.2-fold), CD79A (2.2-fold), and CD79B (2.8-fold). In addition, there was an increase in the monocyte-specific genes TLR7 (5.2-fold), TNFRSF21 (=DR6, 14.4-fold), PLEK (8.8-fold), CSF2RB (=GM-CSF-R, 1.9-fold), and FLT3 (1.6-fold), and the activation marker CD28 (>50-fold).
Because smaller but concordant changes in several genes of one pathway can be biologically relevant, the changes were analyzed using gene-set enrichment analyses (GSEA) (23). The changes were ranked from most up-regulated to most down-regulated and compared with the “c2-curated pathway” gene sets and the “c7-immunological” gene sets provided by the Molecular Signatures Database (MSigDb). GSEA generates the q-value describing the chance that differences between genes in the experimental list and a curated list are false. A low q-value indicates a high probability that the GSEA result is not found by chance. In addition, GSEA provides the enrichment score, which is generated by an algorithm that increases when a gene is present in both the experimentally determined list and the MSigDb list, or decreases when it is not. The enrichment score (ES) is then normalized (NES) (23).
Fig. 5B shows four gene sets that closely resemble the differences between KOPN-8L-R and KOPN-8L. Genes that are down-regulated in large prelymphocytes compared with mature B-lymphocytes (24) are down-regulated in KOPN-8L-R (q < 0.0009, NES = −2.33). Genes defined as “BCR pathway” (25) (q = 0.004, NES = −2.07) are also down-regulated. Both sets of data suggest a change of KOPN-8L-R to less mature B-cells. Additionally, genes that are down-regulated in B-cells compared with plasmacytoid dendritic cells (pDC) (26) were up-regulated in KOPN-8L-R (q < 0.0009, NES = 2.15), indicating a change toward pDCs. In addition, genes up-regulated in B cells compared with monocytes (26) were down-regulated in KOPN-8L-R cells, indicating a change toward a monocytic expression pattern (q = 0.003, NES = −2.04). Taken together, GSEA analysis supports a dedifferentiation of KOPN-8L-R cells from pre-B cells toward less-mature B cells and cells of non-B hematological lineages.
The ingenuity pathway analysis (IPA) further supported a change toward less-mature B cells, as indicated by the decreased expression of BCR-pathway genes. IPA identified the BCR-pathway as the second-most altered canonical pathway in KOPN-8L-R compared with KOPN-8L (Fig. S6).
The transcription factors (TFs) determining early and intermediate B cell fates also changed significantly in agreement with the pathway changes (Fig. 5C) (27). We found the B cell-specific TFs TCF3 (−1.5×, P < 0.0001), FOXO1 (−3.2×, P < 0.0001), EBF1 (−1.5×, P < 0.0001), and BCL6 (−6.3×, P < 0.0001) decreased, while DC-determining TFs ID2 (2.4×, P < 0.0001), IRF4 (2.1×, P < 0.0001), and GFI1 (>50-fold, P < 0.0001) increased (Fig. 5 A and C) (28). Together, these data indicate that a significant down-regulation of several key TFs in B cell development lead to a marked down-regulation of B cell-specific genes (27), whereas the up-regulation of ID2, IRF4, and GLI1 likely result in the up-regulation of genes expressed in monocytic cells.
The Combination of 5-AZA and Moxe Prevented Local Resistance.
Moxe-resistance in vitro can be caused by methylation of promoter regions, which can be prevented by 5-AZA (16, 17). Consequently, we hypothesized that the resistance observed in mice might also be due to DNA methylation and we tested 5-AZA in our model. Considering that the methylation changes might occur early after cell implantation, we treated mice with daily doses of 5-AZA from day 3 (29) followed by five doses of 0.3 mg/kg Moxe intravenously from day 8. Untreated mice progressed rapidly (Fig. 6 A and B, Upper). The bioluminescence for mice treated with Moxe alone fell and rose again in relapse as expected. Daily treatment with 5-AZA alone slowed disease progression, as indicated by the lower bioluminescence on day 8 (P = 0.003). The bioluminescence then increased between days 8 and 19 (P = 0.06). The combination of Moxe and 5-AZA resulted in a total abrogation of the bioluminescence and none of the mice showed any luciferase signal thereafter. The changes in bioluminescence correlated with animal survival (Fig. 6B, Lower). Animal survival was marginally prolonged by 5-AZA alone, and significantly increased by Moxe alone. However, mice that received the combination treatment were healthy and disease-free on day 120, when the experiment was terminated. Pathological examination and BM flow at day 120 showed no evidence of leukemia. It appeared that these mice were cured.
5-AZA Did Not Reverse Established Moxe-Resistance.
To determine whether 5-AZA could overcome the established resistance of KOPN-8L-R in mice, we harvested ALL cells from mouse 5, reinjected the cells in new mice, and tested the effect of Moxe, 5-AZA, or the combination (Fig. 6C, Upper). Untreated mice showed a rapid increase in bioluminescence and died on day 25. Mice treated with 5-AZA from day 3 showed a slow, but steady increase in bioluminescence (P < 0.001). The combination of 5-AZA and Moxe slowed the increase of bioluminescence, but all mice progressed after the treatment ended. The mice treated with 5-AZA survived on average 4 d longer, Moxe-treated mice 11 d longer, and mice treated with the combination 16.5 d longer than untreated mice (Fig. 6C, Lower). None of the mice showed long-term disease-free survival, as seen in the mice bearing KOPN-8L cells. These results indicate that once resistance is established it cannot be reversed with 5-AZA.
5-AZA Enhanced the Activity of Moxe in RehL Xenografts.
Mice bearing the RehL cells were treated as the mice bearing KOPN-8L. Vehicle-treated mice showed an increase in bioluminescence (Fig. 7A). In mice treated with 5-AZA, bioluminescence increased between days 8 and 20 (P = 0.02). In mice treated with Moxe alone, bioluminescence on day 20 was stable compared with day 8 (P = 0.94). Only the Moxe/5-AZA combination greatly reduced the signal intensity on day 20 in all 10 mice (P = 0.001). The signal fell below the detection limit in four mice and the mice stayed free of bioluminescence up to 50 d. Nevertheless, all mice relapsed from discrete sites closely associated with bones. Untreated mice showed a median survival of 37 d, mice treated with Moxe of 45 d, 5-AZA treated mice of 51 d, and mice receiving the combination of 67 d (Fig. 7B).
Discussion
We have developed a mouse model of resistance of ALL cells to Moxe, to understand the patho-biologic basis for reduced clinical responsiveness. We find that the ALL cells growing in murine BM become resistant to Moxe and that the resistant cells have developed major chromosomal changes and changes in gene expression, indicating dedifferentiation toward less mature B cell precursors. These changes were not observed when Moxe-resistant cells were selected in cell culture, probably because the resistant cells from mice cannot grow in vitro.
An unexpected feature of the resistant cells growing in mice, which is more likely to recapitulate resistance in patients in comparison with in vitro cell line studies, is that resistant cells first appear at discrete locations in various bones and then spread to other locations. Treatment initially clears the disseminated cells, which then regrow, originating from discrete BM-sites and gradually become resistant. This behavior indicates that resistance develops in several steps. It is also consistent with a protective BM-niche providing local supportive signals (10, 30, 31) and inducing a therapy-resistant state (32, 33). BM niche-induced Moxe resistance is in line with findings in pediatric ALL patients treated with anti-CD22 immunotoxin, where clearance of peripheral blood blasts was observed despite persistence of BM disease (34). Because the development of resistance is prevented by treatment with 5-AZA, but 5-AZA is not effective after resistance has developed, we believe that early changes in gene-expression patterns contribute to the Moxe-resistance and possibly to the major chromosomal changes observed in cells isolated after several cycles of treatment.
Relapse of ALL after chemotherapy commonly presents with chromosomal changes resulting in substantial phenotype changes (35, 36). Relapse of ALL after targeted therapy with CD19-targeting BiTEs (37, 38) or CD19-targeting CAR-T cells (39, 40) similarly present with the loss of CD19, which is accompanied by a lineage switch to a myeloid leukemia not expressing B cell markers with additional chromosomal changes (37–40). For aggressive B cell malignancies, such chromosomal changes in relapse have been described as linear evolution out of the predominant clone at first diagnosis (41–43). Our model suggests that these chromosomal changes develop step-wise and by chance, and take place in cells that are protected by discrete BM-niches. Therapeutic pressure by repeated cycles of CD22-targeted immunotoxin gives rise to a linear subclone with substantial chromosomal and phenotypic changes.
The therapy-resistant subclones of both, the KMT2A-ENL translocated ALL cell line KOPN-8 and the TEL-AML1 translocated cell line Reh, have a marked down-regulation of B cell-specific markers, including CD22 the target of Moxe. An only twofold reduction of the surface CD22 is paralleled by a profound reduction of intracellular CD22. The surface CD22 is internalized after receptor binding and then replenished by the intracellular pool (21). Thus, the reduction of the intracellular pool of CD22 may affect the internalization of Moxe over time. In accord with our previous data showing that CD22-targeted immunotoxins are more active the longer the cells are exposed (20, 44), the reduction of intracellular CD22 suggests that the resistant cells must be exposed to Moxe longer for them to die. Because Moxe has a short 20-min half-life in mice, the drug concentration after a bolus dose falls rapidly to low levels not capable of killing the resistant cells (20, 44). Even though suggested by our data, CD22 expression levels and Moxe activity does not necessarily correlate (45) and CD22-targeted immunotoxin can be highly active against ALL that expresses little CD22 (34).
In summary, we showed in two mouse models that Moxe resistance is due to major structural changes in chromosomes and in gene expression. Systemic resistance develops after several cycles of treatment. By combining Moxe with 5-AZA, resistance and subsequent relapse is completely prevented in KOPN-8 xenografts and delayed in Reh xenografts. The marked increase of Moxe efficacy by 5-AZA makes this combination a promising approach for future clinical testing in children and adults with ALL.
Materials and Methods
Cell Lines.
KOPN-8 and Reh cells were described previously (20). Cellular identity was established by short tandem-repeat analysis. Cells were grown in RPMI-1640 with 10% FBS, 100 U penicillin, and 100 mg streptomycin. Generation of luciferase+ KOPN-8L and RehL cells is described in Figs. S1 and S2, respectively. The Moxe-resistant derivate KOPN-8L-Res emerged spontaneously in vivo after repeated cycles of treatment, as described in Results. The stromal cell line OP-9 (CRL-2749) was purchased from ATCC.
For coculture experiments, OP-9 cells were seeded at 3,000 cells per well in a 96-well plate on day 0. On day 1, 50,000 GL-Res or GL cells were added and 6 h later treated with Moxe at indicated concentrations. The cells were stained 48 h later with Annexin V/7-AAD and viability determined by flow cytometry.
Reagents.
Moxe was supplied by Medimmune. 5-AZA (Sigma) was dissolved in 0.9% NaCl at 2 mg/mL, sterile-filtered, and stored at −80 °C. Panobinostat (Selleck) was dissolved in 2% DMSO, 2% Tween80, 48% PEG-300, and 48% water at 5 mg/mL.
Secondary Western blot antibodies were purchased from Santa Cruz, polyclonal α-CD22 was produced in rabbits. Antibodies for flow cytometry (α-CD10-Cy5.5, α-CD19-PE, α-CD22-PE, α-CD28-APC, α-CD33-PerCP, and α-CD34-PE) were purchased from Becton Dickinson.
Animal Studies.
Animals were handled according to NIH guidelines; studies were approved by the National Cancer Institute Animal Care and Use Committee.
Five million KOPN-8L (20), 1 million KOPN-8L-Res, or 5 million RehL cells (34) were injected on day 1 via tail vein into 6- to 8-wk-old NSG mice. Moxe was given intravenously, as indicated. 5-AZA was given at 2 mg/kg and panobinostat at 5 mg/kg intraperitoneally daily from day 3 until the last dose of Moxe (29). To assess BM response, mice were killed on days indicated. For survival studies mice were followed until they showed disease progression and were killed. Disease progression for KOPN-8L-bearing mice was defined as >10% loss of highest body weight, for RehL-bearing mice as development of hind-limb paralysis or weight loss >10% of highest body weight.
BM was extracted with mortar and pestle, mesh-filtered, washed, and either viably frozen or sorted with a FACS Aria (for FSC/SSC, FSC-H/FSC-W, and GFPpos). Six million sorted cells per mouse were split in half and lysed for extraction of total RNA using the RNEasy kit (Qiagen) or lysed in modified RIPA buffer [50 mM Tris⋅HCl pH 7.5, 0.25% Na-Deoxycholate, 1% Nonidet P-40, 150 mM NaCl, HALT protease inhibitor (Pierce)]. Total RNA was quality controlled and samples with a registrant identification number of 10 sent to the National Cancer Institute RNA deep-sequencing facility; 1 μg was reverse-transcribed and amplified with the QuantiFast SYBR Green PCR kit (Qiagen) for qRT-PCR using primers, as indicated in Table S1. The data were analyzed using the comparative CT method (ddCT) with SDS manager (Applied Biosystems) and normalized to the average CT values of three housekeeping genes.
For immune-phenotyping, cells were Fc-receptor blocked, stained in PBS/1% BSA/2 mM EDTA/0.1% Na-Azide, analyzed on a FACS Calibur, and quantified using FlowJo (TreeStar).
For immunohistochemistry, murine tissue was formalin-fixed for 24 h, stored in 70% ethanol for 48 h, and paraffin-embedded; 4-μm sections were stained with anti-human Mito A/B.
RNA Deep Sequencing.
RNA libraries were prepared using the TruSeq stranded RNA protocol and sequenced on an Illumina HiSeq2500 sequencer using TruSeq V4.0. Quality was checked using FastQC, reads were trimmed using Trimmomatic, and mapped to the human genome (hg19) by STAR in “2-pass” mode (46). The reads at gene level were quantified using subread feature counts (47) and normalized to library size as counts per million. Quantile normalization and differential expression was carried out using limma-voom (48). For GSEA, the genes were preranked by t-statistic and compared with curated gene lists in “pre-ranked mode” (23). To generate the reads corresponding to the KMT2A-ENL fusion, the BOWTIE aligner for gene mapping (49) was combined with the BLAT and STAR aligner for mapping the genes to the fusion junction (50).
Patient Studies.
Primary ALL blast samples from patients with CD22+ pre–B-ALL treated on a phase I trial of Moxe (NCT01891981) were collected with informed consent under protocol 04-C-0102 and approved by the National Cancer Institute Institutional Review Board. Total RNA was extracted and RNA deep sequencing performed as described above.
Luciferase Imaging.
d-luciferin (75 mg/kg, VivoGlo; Promega) was injected intraperitoneally, mice anesthetized with Isoflurane, and images were taken 5 min after injection using a Xenogen IVIS-100 (Caliper).
Statistics.
Statistical analyses were performed with Graph Pad Prism v7.00 as unpaired t tests (two group comparison), as ANOVA (multiple comparison analyses), or as log-rank test (animal survival).
Supplementary Material
Acknowledgments
The authors thank the Building 37 Animal Facility for their support. This work utilized the computational resources of the NIH High-Performance Computing Biowulf cluster (https://hpc.nih.gov/). This research was supported in part by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research. F.M. was supported in part by the German Research Foundation, Award MU 3619/1-1 and the Interdisciplinary Center for Clinical Research, Erlangen, Award IZKF-J59 and IZKF-P002. A.S.W. was supported in part by National Cancer Institute Award P30CA014089. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute or the National Institutes of Health.
Footnotes
Conflict of interest statement: A.S.W. and I.P. are coinventors on patents assigned to the NIH for the investigational products. A.S.W. has received research support, honorarium, and travel support from Medimmune; and honorarium and travel support from Pfizer, Kite Pharma, and Spectrum Pharmaceuticals.
This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1714512115/-/DCSupplemental.
References
- 1.Hunger SP, et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: A report from the children’s oncology group. J Clin Oncol. 2012;30:1663–1669. doi: 10.1200/JCO.2011.37.8018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ko RH, et al. Outcome of patients treated for relapsed or refractory acute lymphoblastic leukemia: A Therapeutic Advances in Childhood Leukemia Consortium study. J Clin Oncol. 2010;28:648–654. doi: 10.1200/JCO.2009.22.2950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wayne AS, Fitzgerald DJ, Kreitman RJ, Pastan I. Immunotoxins for leukemia. Blood. 2014;123:2470–2477. doi: 10.1182/blood-2014-01-492256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Maude SL, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Lee DW, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet. 2015;385:517–528. doi: 10.1016/S0140-6736(14)61403-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Haso W, et al. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood. 2013;121:1165–1174. doi: 10.1182/blood-2012-06-438002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Klinger M, et al. Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood. 2012;119:6226–6233. doi: 10.1182/blood-2012-01-400515. [DOI] [PubMed] [Google Scholar]
- 8.Topp MS, et al. Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood. 2012;120:5185–5187. doi: 10.1182/blood-2012-07-441030. [DOI] [PubMed] [Google Scholar]
- 9.Topp MS, et al. Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia. J Clin Oncol. 2014;32:4134–4140. doi: 10.1200/JCO.2014.56.3247. [DOI] [PubMed] [Google Scholar]
- 10.Park E, et al. Targeting survivin overcomes drug resistance in acute lymphoblastic leukemia. Blood. 2011;118:2191–2199. doi: 10.1182/blood-2011-04-351239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kantarjian H, et al. Inotuzumab ozogamicin, an anti-CD22-calecheamicin conjugate, for refractory and relapsed acute lymphocytic leukaemia: A phase 2 study. Lancet Oncol. 2012;13:403–411. doi: 10.1016/S1470-2045(11)70386-2. [DOI] [PubMed] [Google Scholar]
- 12.Kreitman RJ, et al. Phase I trial of anti-CD22 recombinant immunotoxin moxetumomab pasudotox (CAT-8015 or HA22) in patients with hairy cell leukemia. J Clin Oncol. 2012;30:1822–1828. doi: 10.1200/JCO.2011.38.1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Pastan I, Hassan R, Fitzgerald DJ, Kreitman RJ. Immunotoxin therapy of cancer. Nat Rev Cancer. 2006;6:559–565. doi: 10.1038/nrc1891. [DOI] [PubMed] [Google Scholar]
- 14.Stahl S, et al. Loss of diphthamide pre-activates NF-κB and death receptor pathways and renders MCF7 cells hypersensitive to tumor necrosis factor. Proc Natl Acad Sci USA. 2015;112:10732–10737. doi: 10.1073/pnas.1512863112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wei H, et al. A modified form of diphthamide causes immunotoxin resistance in a lymphoma cell line with a deletion of the WDR85 gene. J Biol Chem. 2013;288:12305–12312. doi: 10.1074/jbc.M113.461343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hu X, et al. Methylation of the DPH1 promoter causes immunotoxin resistance in acute lymphoblastic leukemia cell line KOPN-8. Leuk Res. 2013;37:1551–1556. doi: 10.1016/j.leukres.2013.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wei H, et al. Immunotoxin resistance via reversible methylation of the DPH4 promoter is a unique survival strategy. Proc Natl Acad Sci USA. 2012;109:6898–6903. doi: 10.1073/pnas.1204523109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wayne AS, et al. Pediatric phase 1 trial of moxetumomab pasudotox: Activity in chemotherapy refractory acute lymphoblastic leukemia (ALL) Cancer Res. 2014;74(19 Suppl):CT230 (abstr). [Google Scholar]
- 19.Shah NN, et al. Characterization of CD22 expression in acute lymphoblastic leukemia. Pediatr Blood Cancer. 2015;62:964–969. doi: 10.1002/pbc.25410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Sharma SV, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141:69–80. doi: 10.1016/j.cell.2010.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Du X, Beers R, Fitzgerald DJ, Pastan I. Differential cellular internalization of anti-CD19 and -CD22 immunotoxins results in different cytotoxic activity. Cancer Res. 2008;68:6300–6305. doi: 10.1158/0008-5472.CAN-08-0461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Roberts KG, Mullighan CG. Genomics in acute lymphoblastic leukaemia: Insights and treatment implications. Nat Rev Clin Oncol. 2015;12:344–357. doi: 10.1038/nrclinonc.2015.38. [DOI] [PubMed] [Google Scholar]
- 23.Subramanian A, et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Somasundaram R, Prasad MA, Ungerbäck J, Sigvardsson M. Transcription factor networks in B-cell differentiation link development to acute lymphoid leukemia. Blood. 2015;126:144–152. doi: 10.1182/blood-2014-12-575688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mori S, et al. Utilization of pathway signatures to reveal distinct types of B lymphoma in the Emicro-myc model and human diffuse large B-cell lymphoma. Cancer Res. 2008;68:8525–8534. doi: 10.1158/0008-5472.CAN-08-1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Schaefer CF, et al. PID: The pathway interaction database. Nucleic Acids Res. 2009;37:D674–D679. doi: 10.1093/nar/gkn653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Nakaya HI, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol. 2011;12:786–795. doi: 10.1038/ni.2067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Lin Q, et al. Epigenetic program and transcription factor circuitry of dendritic cell development. Nucleic Acids Res. 2015;43:9680–9693. doi: 10.1093/nar/gkv1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Borodovsky A, et al. 5-azacytidine reduces methylation, promotes differentiation and induces tumor regression in a patient-derived IDH1 mutant glioma xenograft. Oncotarget. 2013;4:1737–1747. doi: 10.18632/oncotarget.1408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Konopleva MY, Jordan CT. Leukemia stem cells and microenvironment: Biology and therapeutic targeting. J Clin Oncol. 2011;29:591–599. doi: 10.1200/JCO.2010.31.0904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Reagan MR, Rosen CJ. Navigating the bone marrow niche: Translational insights and cancer-driven dysfunction. Nat Rev Rheumatol. 2016;12:154–168. doi: 10.1038/nrrheum.2015.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Ebinger S, et al. Characterization of rare, dormant, and therapy-resistant cells in acute lymphoblastic leukemia. Cancer Cell. 2016;30:849–862. doi: 10.1016/j.ccell.2016.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Boyerinas B, et al. Adhesion to osteopontin in the bone marrow niche regulates lymphoblastic leukemia cell dormancy. Blood. 2013;121:4821–4831. doi: 10.1182/blood-2012-12-475483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wayne AS, et al. Anti-CD22 immunotoxin RFB4(dsFv)-PE38 (BL22) for CD22-positive hematologic malignancies of childhood: Preclinical studies and phase I clinical trial. Clin Cancer Res. 2010;16:1894–1903. doi: 10.1158/1078-0432.CCR-09-2980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Duijf PH, Benezra R. The cancer biology of whole-chromosome instability. Oncogene. 2013;32:4727–4736. doi: 10.1038/onc.2012.616. [DOI] [PubMed] [Google Scholar]
- 36.Santaguida S, Amon A. Short- and long-term effects of chromosome mis-segregation and aneuploidy. Nat Rev Mol Cell Biol. 2015;16:473–485. doi: 10.1038/nrm4025. [DOI] [PubMed] [Google Scholar]
- 37.Rayes A, McMasters RL, O’Brien MM. Lineage switch in MLL-rearranged infant leukemia following cd19-directed therapy. Pediatr Blood Cancer. 2016;63:1113–1115. doi: 10.1002/pbc.25953. [DOI] [PubMed] [Google Scholar]
- 38.Zoghbi A, Zur Stadt U, Winkler B, Müller I, Escherich G. Lineage switch under blinatumomab treatment of relapsed common acute lymphoblastic leukemia without MLL rearrangement. Pediatr Blood Cancer. 2017;64 doi: 10.1002/pbc.26594. [DOI] [PubMed] [Google Scholar]
- 39.Jacoby E, et al. CD19 CAR immune pressure induces B-precursor acute lymphoblastic leukaemia lineage switch exposing inherent leukaemic plasticity. Nat Commun. 2016;7:12320. doi: 10.1038/ncomms12320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gardner R, et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood. 2016;127:2406–2410. doi: 10.1182/blood-2015-08-665547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Juskevicius D, et al. Distinct genetic evolution patterns of relapsing diffuse large B-cell lymphoma revealed by genome-wide copy number aberration and targeted sequencing analysis. Leukemia. 2016;30:2385–2395. doi: 10.1038/leu.2016.135. [DOI] [PubMed] [Google Scholar]
- 42.Aukema SM, et al. Sequential karyotyping in Burkitt lymphoma reveals a linear clonal evolution with increase in karyotype complexity and a high frequency of recurrent secondary aberrations. Br J Haematol. 2015;170:814–825. doi: 10.1111/bjh.13501. [DOI] [PubMed] [Google Scholar]
- 43.Hogan LE, et al. Integrated genomic analysis of relapsed childhood acute lymphoblastic leukemia reveals therapeutic strategies. Blood. 2011;118:5218–5226. doi: 10.1182/blood-2011-04-345595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Müller F, Stookey S, Cunningham T, Pastan I. Paclitaxel synergizes with exposure time adjusted CD22-targeting immunotoxins against B-cell malignancies. Oncotarget. 2017;8:30644–30655. doi: 10.18632/oncotarget.16141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Mussai F, et al. Cytotoxicity of the anti-CD22 immunotoxin HA22 (CAT-8015) against paediatric acute lymphoblastic leukaemia. Br J Haematol. 2010;150:352–358. doi: 10.1111/j.1365-2141.2010.08251.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dobin A, et al. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Liao Y, Smyth GK, Shi W. The subread aligner: Fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 2013;41:e108. doi: 10.1093/nar/gkt214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Law CW, Chen Y, Shi W, Smyth GK. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15:R29. doi: 10.1186/gb-2014-15-2-r29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9:357–359. doi: 10.1038/nmeth.1923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kent WJ. BLAT–The BLAST-like alignment tool. Genome Res. 2002;12:656–664. doi: 10.1101/gr.229202. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.