9
Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin Moxetumomab Pasudotox Robert J. Kreitman and Ira Pastan Abstract Recombinant immunotoxins are fusion proteins that contain the cytotoxic portion of a protein toxin fused to the Fv portion of an antibody. The Fv binds to an antigen on a target cell and brings the toxin into the cell interior, where it arrests protein synthesis and initiates the apoptotic cascade. Moxetumomab pasudotox, previously called HA22 or CAT-8015, is a recombinant immunotoxin composed of the Fv fragment of an anti- CD22 monoclonal antibody fused to a 38-kDa fragment of Pseudomonas exotoxin A, called PE38. Moxe- tumomab pasudotox is an improved, more active form of a predecessor recombinant immunotoxin, BL22 (also called CAT-3888), which produced complete remission in relapsed/refractory hairy cell leukemia (HCL), but it had a <20% response rate in chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia (ALL), diseases in which the leukemic cells contain much lower numbers of CD22 target sites. Compared with BL22, moxetumomab pasudotox is up to 50-fold more active on lymphoma cell lines and leukemic cells from patients with CLL and HCL. A phase I trial was recently completed in HCL patients, who achieved response rates similar to those obtained with BL22 but without dose-limiting toxicity. In addition to further testing in HCL, moxetumomab pasudotox is being evaluated in phase I trials in patients with CLL, B-cell lymphomas, and childhood ALL. Moreover, protein engineering is being used to increase its activity, decrease nonspecific side effects, and remove B-cell epitopes. Clin Cancer Res; 17(20); 6398–405. Ó2011 AACR. Introduction Targeted toxins Different approaches have been used and are under development to target toxic molecules to cancer cells and kill them more efficiently than would be possible with monoclonal antibodies (mAb) alone. Radioimmunother- apy is the most established of these approaches, repre- sented by the approved drugs ibritumomab tiuxetan and tositumomab (1), and reviewed in this issue by Steiner and Neri (2). Exciting advances have been made with antibody-drug conjugates (3), particularly trastuzumab emtansine (4), SGN-35 (brentuximab vedotin; ref. 5), SAR3419 (6), and calicheamicin conjugates (7). Cyto- kines fused to antibodies are being used to attract immune effector cells to cancer cells (8). Immunotoxins are a type of antibody-conjugate in which a powerful protein toxin instead of a low-molecular-weight drug is attached to an antibody or antibody fragment. Their possible advantages include a very high activity, which enables them to kill cells with relatively few target sites, and a unique mechanism of action that can bypass some types of drug resistance. This review focuses on recom- binant immunotoxins targeted to CD22, particularly moxetumomab pasudotox. Protein toxins Protein toxins produced by bacteria, fungi, and plants are extremely cytotoxic and can kill cells when only one or a few molecules reach the cytosol (9–12). These agents act by inhibiting protein synthesis and inducing apoptosis. The bacterial toxins Pseudomonas exotoxin A (PE) and diphtheria toxin (DT) catalytically inactivate elongation factor 2 by ADP ribosylation (9, 10). Plant toxins inactivate ribosomes by removing adenine 4324 in 28s ribosomal RNA (11, 13). Unlike plant toxins, bacterial toxins are made as single- chain proteins, making them more amenable for construc- tion of recombinant chimeric proteins (14). Recombinant immunotoxins are produced by replacing the binding domain of the toxin with the Fv portion of an antibody that directs the toxin to a cancer cell. Target choice is very important to prevent killing of normal cells. The lineage- restricted differentiation B-cell antigen CD22 is an excellent target because it is absent on normal tissues like liver and skin, and its lack of expression on B-cell precursors allows normal CD22 þ B cells to be rapidly generated after immu- notoxin therapy ceases. CD22, a sialic acid–binding immunoglobulin-like lectin (siglec) that inhibits B-cell receptor calcium signaling, is expressed on many B-cell malignancies, including hairy cell Authors' Afliation: Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, Maryland Corresponding Author: Robert J. Kreitman, Laboratory of Molecular Biology, National Cancer Institute, NIH, 37/5124b, 9000 Rockville Pike, Bethesda, MD 20892. Phone: 301-496-6947; Fax: 800-864-7554; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-0487 Ó2011 American Association for Cancer Research. CCR FOCUS Clin Cancer Res; 17(20) October 15, 2011 6398 on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

Antibody Fusion Proteins: Anti-CD22 RecombinantImmunotoxin Moxetumomab Pasudotox

Robert J. Kreitman and Ira Pastan

AbstractRecombinant immunotoxins are fusion proteins that contain the cytotoxic portion of a protein toxin fused

to the Fv portion of an antibody. The Fv binds to an antigen on a target cell and brings the toxin into the cell

interior, where it arrests protein synthesis and initiates the apoptotic cascade. Moxetumomab pasudotox,

previously calledHA22orCAT-8015, is a recombinant immunotoxin composedof the Fv fragment of an anti-

CD22 monoclonal antibody fused to a 38-kDa fragment of Pseudomonas exotoxin A, called PE38. Moxe-

tumomab pasudotox is an improved, more active form of a predecessor recombinant immunotoxin, BL22

(also calledCAT-3888),whichproducedcomplete remission in relapsed/refractoryhairy cell leukemia (HCL),

but it had a <20% response rate in chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia

(ALL), diseases inwhich the leukemic cells containmuch lower numbers ofCD22 target sites. Comparedwith

BL22,moxetumomabpasudotox is up to 50-foldmore active on lymphoma cell lines and leukemic cells from

patients with CLL and HCL. A phase I trial was recently completed in HCL patients, who achieved response

rates similar to those obtained with BL22 but without dose-limiting toxicity. In addition to further testing in

HCL, moxetumomab pasudotox is being evaluated in phase I trials in patients with CLL, B-cell lymphomas,

and childhood ALL. Moreover, protein engineering is being used to increase its activity, decrease nonspecific

side effects, and remove B-cell epitopes. Clin Cancer Res; 17(20); 6398–405. �2011 AACR.

Introduction

Targeted toxinsDifferent approaches have been used and are under

development to target toxic molecules to cancer cells andkill them more efficiently than would be possible withmonoclonal antibodies (mAb) alone. Radioimmunother-apy is the most established of these approaches, repre-sented by the approved drugs ibritumomab tiuxetan andtositumomab (1), and reviewed in this issue by Steinerand Neri (2). Exciting advances have been made withantibody-drug conjugates (3), particularly trastuzumabemtansine (4), SGN-35 (brentuximab vedotin; ref. 5),SAR3419 (6), and calicheamicin conjugates (7). Cyto-kines fused to antibodies are being used to attractimmune effector cells to cancer cells (8). Immunotoxinsare a type of antibody-conjugate in which a powerfulprotein toxin instead of a low-molecular-weight drug isattached to an antibody or antibody fragment. Theirpossible advantages include a very high activity, which

enables them to kill cells with relatively few target sites,and a unique mechanism of action that can bypass sometypes of drug resistance. This review focuses on recom-binant immunotoxins targeted to CD22, particularlymoxetumomab pasudotox.

Protein toxinsProtein toxins produced by bacteria, fungi, and plants are

extremely cytotoxic and can kill cells when only one or a fewmolecules reach the cytosol (9–12). These agents act byinhibiting protein synthesis and inducing apoptosis. Thebacterial toxinsPseudomonas exotoxinA (PE) anddiphtheriatoxin (DT) catalytically inactivate elongation factor 2 byADP ribosylation (9, 10). Plant toxins inactivate ribosomesby removing adenine4324 in 28s ribosomal RNA (11, 13).Unlike plant toxins, bacterial toxins are made as single-chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinantimmunotoxins are produced by replacing the bindingdomain of the toxin with the Fv portion of an antibodythat directs the toxin to a cancer cell. Target choice is veryimportant to prevent killing of normal cells. The lineage-restricted differentiation B-cell antigen CD22 is an excellenttarget because it is absent on normal tissues like liver andskin, and its lack of expression on B-cell precursors allowsnormal CD22þ B cells to be rapidly generated after immu-notoxin therapy ceases.

CD22, a sialic acid–binding immunoglobulin-like lectin(siglec) that inhibits B-cell receptor calcium signaling, isexpressed onmany B-cell malignancies, including hairy cell

Authors' Affiliation: Laboratory of Molecular Biology, National CancerInstitute, NIH, Bethesda, Maryland

Corresponding Author: Robert J. Kreitman, Laboratory of MolecularBiology, National Cancer Institute, NIH, 37/5124b, 9000 Rockville Pike,Bethesda, MD 20892. Phone: 301-496-6947; Fax: 800-864-7554; E-mail:[email protected]

doi: 10.1158/1078-0432.CCR-11-0487

�2011 American Association for Cancer Research.

CCRFOCUS

Clin Cancer Res; 17(20) October 15, 20116398

on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 2: Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

leukemia (HCL), chronic lymphocytic leukemia (CLL),B-cell non-Hodgkin lymphoma (NHL), and acute lym-phoblastic leukemia (ALL; ref. 15). To kill these cells, weinitially constructed an immunotoxin that reactswithCD22by chemically conjugating a toxin fragment of PE to the LL2antibody (16). In studies with this immunotoxin as well asseveral other chemical conjugates, we found that theseconjugateswere heterogeneous in composition anddifficultto produce. Therefore, we turned to recombinant DNAtechniques to make recombinant immunotoxins, whichare stable,�60- to 65-kDa, homogeneous chimeric proteinsin which the Fv of a mAb is fused to truncated PE. Thechimeric gene encoding the recombinant immunotoxin isintroduced into Escherichia coli, where the recombinantprotein is produced.

Cellular intoxication by recombinant immunotoxinsCrystallographic studies have shown that PE is made up

of 3major structural domains (Fig. 1). Domain Ia is the cell-binding domain, domain II contains a furin site that is

necessary to release domain III from the cell-bindingdomain, and domain III contains the ADP-ribosylatingactivity that inactivates elongation factor 2. Domain Ib,which has no known function, can be partially or totallyremoved without affecting toxin activity. In recombinantimmunotoxins targeting CD22, domain Ia of PE is removedand replaced by the Fv portion of an antibody reacting withCD22 as shown in Fig. 1.

For cancer treatment, the recombinant immunotoxinis administered i.v. so that it can reach all of the cells. Afterinjection, the Fv at the amino terminus binds to CD22present on the B-cell malignancy being targeted (17),and the carboxyl terminal lysine residue at position 613(18) is rapidly removed by plasma carboxypeptidase,generating a protein ending in REDL. Very soon afterbinding, the recombinant immunotoxin-CD22 complex isinternalized through clathrin-coated pits into the endocyticcompartment (19), where 2 steps occur: (i) reduction of thedisulfide bond in domain II, and (ii) a furin-catalyzedcleavage in the middle of domain II, resulting in the

Figure 1. Intoxication of cells by moxetumomab pasudotox. Left, illustration of the structure of PE, PE38, and recombinant immunotoxin moxetumomabpasudotox designed to kill CD22-expressing cells. Right, cartoon showing the steps required for the entry and cell killing by moxetumomab pasudotoxand similar recombinant immunotoxins containing PE38. After internalization, PE undergoes proteolysis and disulfide-bond reduction to separatethe catalytic domain III from the binding domain Ia (20, 60, 61). PE38 undergoes both removal of the carboxyl terminal lysine residue (18) and processingbetween residues 279 and 280, resulting in a 37-kDa carboxyl terminal toxin fragment ending in the residues REDL. This fragment is believed to betransported intracellularly via the KDEL receptor from the Golgi to the endoplasmic reticulum (ER; 21), where it translocates to the cytosol, resulting inADP-ribosylation of elongation factor 2 (9), leading to apoptotic cell death (25).

Anti-CD22 Recombinant Immunotoxins

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6399

on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 3: Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

separation of the Fv from domain III (Fig. 1; ref. 20). TheREDL sequence at the carboxyl terminus of the protein thenbinds to the KDEL recycling receptor (21), and the ADP-ribosylating domain is transported to the endoplasmicreticulum, from which it translocates to the cytosol (22,23). Once in the cytosol, the toxin catalyzes ADP ribosyla-tion of the diphthamide residue (24) in EF-2 (9). This leadsto a rapid decrease in the levels of the antiapoptotic proteinMcl-1 and,with the assistance of BAK, initiates the apoptoticcascade (25–29).

Development of recombinant immunotoxins for CD22malignancies

The first recombinant immunotoxin that was designed tokill CD22-expressing cells contained a single-chain Fv of theanti-CD22 RFB4 antibody, which had been previouslyisolated by Peter Amlot in England, fused to PE38 (Fig. 1and Fig. 2). This recombinant immunotoxin had relativelymodest cytotoxic activity, with IC50s of 2 to 30 ng/mL,which we attributed to the instability of the 2 variabledomains linked by the (G4S)3 peptide (30). To increasestability, we introduced cysteine residues into the light andheavy chains of the Fv so that a disulfide bond would formand stably anchor the light and heavy chains instead of thepeptide linker that is used in most single-chain Fvs (Fig. 2;refs. 31–36). This protein was named BL22, for B-cellleukemia/lymphoma/CD22 (37).

Development of BL22In preclinical studies, BL22was cytotoxic to awide variety

of CD22-expressing cell lines, being 1.5- to 6.7-fold morecytotoxic than the single-chain recombinant immunotoxin(30, 37). BL22 induced complete remission (CR) in nudemice bearing CD22þ lymphoma xenografts at plasma levels

that could be safely achieved in cynomolgus monkeys (38).Primary ALL, CLL, andNHL cells were also tested ex vivo andfound to be sensitive to BL22 (39). CLL with as few as 350CD22 sites/cell could be killed (39). Thus, the preclinicaldata justified the testing of BL22 in humans. A GoodManufacturing Practices lot of BL22 was produced at theMonoclonal Antibody and Recombinant Protein facility ofthe National Cancer Institute, and clinical trials were initi-ated in 1999.

Phase I–II activity of BL22In the phase I trial, we elected to give BL22 every other day

(QOD), for a total of 3 doses per cycle, and to repeat cyclesevery 21 to 28 days. The QOD dosing was based on animalexperiments that showed that toxicity generally occurswithin 48 hours of each dose. Therefore, keeping the dosinginterval to 2 days in patients may prevent cumulativetoxicity, particularly during dose escalation. In targetingHCL, where high CD22 density represents a sink for thedrug, this dosing interval had an additional advantage inallowing dying cells to clear prior to the next dose.Webeganat 3 mg/kg and determined the maximum tolerated dose(MTD) to be 40 mg/kg � 3. In total, we treated 46 patientswho had experienced failure of standard therapies for theirdisease, including 31 with HCL, 4 with B-NHL, and 11 withB-CLL (40, 41). In the 31 patients with HCL, 19 CRs (61%)and 6 partial remissions (PR; 19%) were achieved, for anoverall response rate (ORR) of 81%. A hemolytic uremicsyndrome composed of thrombocytopenia, hemolytic ane-mia, and renal insufficiencywas observed in 4HCLpatients,and it completely resolved after 6 to 10 days of plasmaphe-resis. Dose-limiting capillary leak syndrome (CLS) was lesscommon, appearing in only 1 HCL patient. Pharmacoki-netic analysis of BL22 showed a strong sink effect of CD22

Figure 2. Recombinantimmunotoxins in use or underdevelopment. BL22 was reported in1997 (37) as RFB4(dsFv)-PE38 andwas later called CAT-3888. VL andVH are disulfide-bonded togetherusing engineered cysteine residuesreplacing Arg44 of VH and Gly100 ofVL. In 2002, BL22 was mutated toHA22 (later called CAT-8015 andmoxetumomab pasudotox) bychanging SSY to THW at positions100, 100a, and 100b of VH(45; horizontal redbars). In 2009, thedeletion mutant HA22-LR wasreported where PE amino acids251–394 were replaced by aminoacids 274–284 containing the Furincleavage site (50). Most recently(in 2011), HA22-LR-8M, a mutant ofHA22-LR, was reported to contain 8mutations: D406A, R432G, R467A,R490A, R513A, E548A, K590S, andQ592A (59).

CCRFOCUS

Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research6400

on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 4: Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

in HCL during the first cycle, caused by disease burden andhigh levels of soluble CD22, both of which decreased withresponse (42). Because 65% of CRs occurred after just 1cycle of BL22, the phase II trial was planned to limit BL22 to1 cycle and re-treat only those patients who had notachieved recovery of cytopenias to the level needed for CR.In 36 phase II patients receiving 1 cycle of BL22 at 40 mg/kgQOD� 3, there were 9 CRs (25%) and 9 PRs (25%), for anORR of 50%. With selective re-treatment of 56% of thepatients, the final ORR increased to 72%, including 47%CRs (43). Themedian time to relapse of cytopenias had notbeen reached after nearly 7 years of follow-up. This indi-cated significant activity in relapsed/refractory HCL with asafety profile supporting continued development. Howev-er, because the activity of BL22wasmuch lower inCLL, ALL,and NHL than in HCL (41, 44) and activity in the more-common malignancies was essential for commercial devel-opment,wehad to switch our clinical efforts to an improvedimmunotoxin.

Development of moxetumomab pasudotox, anaffinity-matured version of BL22An important variable in the activity of immunotoxins is

their affinity for the target antigen, which determines howmuch immunotoxin will bind to a target cell at any givenconcentration and how long the cell-bound immunotoxinwill remainon the cell surface. The longer it stays attached tothe target protein (CD22), the more likely it is to enter andkill the target cell. BL22 has a moderate affinity for CD22(Kd �10 nmol/L). This affinity is sufficient to kill enoughHCL cells in patients to achieveCR.HCL cells have amedianof �40,000 CD22 sites per cell, ranging from 10,000 to>100,000. However, BL22 was much less effective in chil-drenwith ALL (4,500 sites/cell) or CLL (1,200 sites/cell). Toimprove the affinity and activity of BL22, we carried outmutagenesis studies and selected an Fv with a higher bind-ing affinity by antibody phage display. Mutation of 3residues inCDR3of theheavy chainof theBL22Fv (residues100, 100a, and100b) fromSSY to THW increased affinity by�15-fold and cytotoxicity toward HCL and CLL cells by up

to 50-fold (45). The improvement in binding affinity wasdue to a slowing of the off rate, which led to increasedcytotoxicity and antitumor activity despite similar pharma-cokinetics and animal toxicity (46). Thenew, higher-affinityversion of BL22 was initially named HA22 (high-activityBL22). Later, after it was licensed to Cambridge AntibodyTechnologies, it was named CAT-8015, and it is now beingdeveloped by MedImmune as moxetumomab pasudotox.

Phase I testing of moxetumomab pasudotoxAt the 2010 American Society of Hematology Annual

Meeting (Orlando, FL; December 4–7, 2010), interimresults were reported for clinical trials with moxetumomabpasudotox in HCL and ALL. Clinical trials are listed inTable 1. Several striking differences between moxetumo-mab pasudotox and BL22 have been noted. One is that inthe HCL trial, dose-limiting toxicity was not observed anddose escalation was terminated at 50 mg/kg, a dose levelabove the MTD for BL22, because response rates were highat all dose levels. In fact, theORR froman interimanalysis ofmoxetumomab pasudotox, 79% (47), was similar to the72% ORR reported from the phase II study of BL22 (43).Another striking finding is that 3 responses (all CRs) wereobserved in 12 patients with pediatric ALL (48), represent-ing the first time responses of thismagnitude were observedwith this type of agent in this aggressive disease.

Interim phase I results in HCLAt the 2010 American Society of Hematology Annual

Meeting (49), investigators presented findings obtainedfrom 32 patients with refractory HCL, all of whom hadreceived 2 ormore prior courses of purine analogue therapyand needed treatment based on established criteria for HCL(i.e., neutrophils < 1000/mm3, platelets < 100,000/mm3,hemoglobin < 10 g/dL, or symptomatic splenomegaly).Patients received dose levels of 5 to 50 mg/kg QOD � 3 ina standard dose-escalation phase, with cycles repeated every4 weeks. Patients were re-treated until they reached 2 cyclespast CR; however, treatment was stopped earlier if progres-sive disease or immunogenicity was detected. Surprisingly,

Table 1. Ongoing phase I trials with moxetumomab pasudotox

Diseases Prior therapy needed for eligibility Current location

HCL �2 prior therapies, �2 purine analogue courses, unlessresponse to first course was <2 years

NIH

CLLa �2 prior therapies, �1 with rituximab NIH, SCRI, IU, MUSC, CCCN, CSMCDLBCL, MCL �1 prior therapy with rituximabFL �2 prior therapies, �1 with rituximabALL (pediatric) �2 prior therapies, prior HSCT allowed NIH, SJ, DF

Abbreviations: CCCN, Comprehensive Cancer Center of Nevada; CSMC, Cedars Sinai Medical Center; DF, Dana-Farber CancerInstitute; DLBCL, diffuse large B-cell lymphoma; HSCT, hematopoietic stem cell transplant; IU, Indiana University; MCL, mantlecell lymphoma; MUSC, Medical University of South Carolina; SCRI, Sarah Cannon Research Institute; SJ, Saint Jude Children'sResearch Hospital.aListed as a phase I–II trial.

Anti-CD22 Recombinant Immunotoxins

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6401

on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 5: Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

no dose-limiting toxicity was observed in the 32 patients,despite the fact that half of the patients enrolled (n ¼ 16)were at the 50 mg/kg � 3 dose level, which was higherthan the MTD of BL22 (41). The most common adverseevents, seen in 20% to 50% of patients, were expected fromprior experience with immunotoxins. These included mildCLS causing weight gain, hypoalbuminemia, peripheraledema, fever, elevated transaminases, fatigue, and head-ache. The only evidence of possible hemolytic uremic

syndrome was asymptomatic laboratory abnormalitiesin 2 patients, with peak creatinine of 1.53 to 1.66 mg/dLand platelet nadir of 111,000 to 120,000/mm3. Majorresponses were observed at all dose levels in the 32 patientsreported, with CRs observed at all dose levels from 10to 50 mg/kg QOD � 3, and the CR rate at all doses was31% at the time of interim analysis. The median time toresponse was 2.8 months (49). Only 1 of the 14 patientswho achieved CR relapsed in less than 1 year, indicating

Moxetumomab pasudotox

HA22-LR

R490

R432

R467R513

E548

Q592

D406K590

HA22-LR-8M

Figure 3. Structure of HA22-LR-8M.Ribbon structures show thelocations of the mutations inHA22-LR-8M and the deletion fromHA22 used in HA22-LR andHA22-LR-8M. Adapted from Ondaet al. (59).

CCRFOCUS

Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research6402

on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 6: Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

that responses were durable. In summary, moxetumomabpasudotox showed high and durable antitumor activity inpatients with relapsed/refractory HCL, statistically similartoBL22butwithout dose-limiting toxicity, justifying furtherclinical development toward the goal of approval for thisdisease. Moxetumomab pasudotox is also being evaluatedin CLL and B-cell lymphomas, but data are not yet availablefrom those trials.

Interim phase I results in ALLBecause pediatric ALL is a much more rapidly growing

and aggressive disease than HCL, patients were treated withmoxetumomab pasudotox QOD for 6 doses, and the cycleswere repeated every 21 days (48). A rapid dose-escalationschemewas used for the lowdose levels (5, 10, and 20mg/kgQOD � 6), and then a standard dose escalation began at30 mg/kg QOD � 6. Fourteen patients 5 to 22 years of age(median, 11 years) were enrolled in the trial. All of thesepatients had been heavily pretreated with 2 to 8 (median 4)prior regimens, and 7 had previously undergone stemcell transplantation. Common toxicities included elevatedbilirubin, transaminases, and creatinine, and CLS in-cluding hypoalbuminemia, proteinuria, hypoxia, and pleu-ral effusion. Grade 3/4 CLS was observed in 2 of the first7 patients (both treated at 30 mg/kg QOD � 3) but not in7 subsequent patients once dexamethasone prophylaxiswas instituted. As mentioned above, of 12 patients whowere evaluable for response, 3 (25%) achieved CR after 1to 2 cycles, and 5 (42%) had hematologic improvementwith reduction in circulating blasts. As in the HCL trial,CRs were observed at dose levels as low as 10 mg/kg. Thus,moxetumomab pasudotox has achieved major responses,including CRs, in patients with ALL. Due to the aggressivenature of the disease and the young age of these patients,even a transient CR can be lifesaving as a bridge to trans-plantation. Further clinical development ofmoxetumomabpasudotox is proceeding in ALL to determine whetherhigher dose levels and more frequent dosing can improveresponses.

A Protease-Resistant Moxetumomab Pasudotox

One impediment to the cytotoxic activity of immuno-toxins is that many of the immunotoxin molecules thatenter target cells do not reach the endoplasmic reti-culum and cytosol; instead, they are transferred to lyso-somes and other compartments containing lysosomalenzymes, where they are inactivated by proteolysis (Fig. 1).To identify potential lysosomal protease cleavage sites,investigators treated recombinant immunotoxins with lyso-somal enzymes and identified the cleavage sites by aminoacid analysis (50). These studies revealed that all of themajor protease sites were clustered in domain II and couldbe removed, leading to a new lysosomal protease-resistantimmunotoxin named HA22-LR (Fig. 2 and Fig. 3). InHA22-LR, all of domain II is deleted except for the 11 aminoacids that make up the essential furin-processing site.HA22-LR had several useful properties, including increased

activity, particularly on CLL cells, where HA22-LR had amedian 16-fold improved cytotoxic activity comparedwith HA22 and more than 10-fold reduced nonspecifictoxicity to mice (50). The latter finding may be due to lossof residues that interact with endothelium or macrophagesand produce liver damage in mice and possibly CLS inother animals, including humans (50–53). It is possiblethat the smaller size of HA22-LR (51.0 kDa versus63.3 kDa for HA22) may allow improved tumor penetra-tion. Finally, because we could safely give much higherdoses to mice, we were able to achieve much better anti-tumor activity (50).

A Moxetumomab Pasudotox Variant with NoImmunogenicity in Mice

Although the incidence of antibody formation inpatients treated with moxetumomab pasudotox is lowand did not prevent CRs in HCL and ALL, furtherdecreases in toxin immunogenicity remain an importantgoal. For patients with solid tumors and normal immunesystems, immunogenicity rates of 80% to 90% wereobserved after 1 cycle of 3 doses (54–56). To deimmunizethe toxin, efforts to identify and remove both T-cell (57)and B-cell (58) epitopes are underway. To date, moreprogress has been made with the B-cell epitope approach(53, 59). On the basis of the hypothesis that human andmouse B-cell epitopes are similar, we used mice for thesestudies and identified 7 major epitopes in PE38: 3 indomain II, and 4 in domain III. The epitopes in domain IIare removed in HA22-LR (50), which has a deletion ofmost of domain II, and the epitopes in domain III weredestroyed by mutating 8 different bulky amino acids toalanine, serine, or glycine to produce HA22-LR-8M (59).We measured the reactivity of HA22-LR-8M with serumfrom patients who made antibodies to HA22, and wefound that reactivity with antisera was greatly decreasedbut not abolished. We are now engaged in studies toidentify and remove the remaining human-specific epi-topes. Our data suggest that new immunotoxins withlow immunogenicity will not be needed for the success-ful treatment of B-cell malignancies, because the immunesystem is very suppressed. However, they will be usefulfor the treatment of solid tumors, where neutralizingantibodies develop in the vast majority of patients after1 cycle of 3 doses. For HCL in particular, we believethe clinical results (both efficacy and safety profile) formoxetumomab pasudotox justify further clinical devel-opment, including a pivotal phase III trial currentlyunder design, with the goal of achieving U.S. Food andDrug Administration approval for the use of moxetumo-mab pasudotox for this disease.

Disclosure of Potential Conflicts of Interest

I. Pastan is an inventor on patents on moxetumomab pasudotoxheld by the NIH. The authors disclose no other potential conflicts ofinterest.

Anti-CD22 Recombinant Immunotoxins

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6403

on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 7: Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

Acknowledgments

The authors thank the members of our current clinical team, Rita Min-cemoyer, Elizabeth Maestri, Natasha Kormanik, Barbara Debrah, and SonyaDuke. We also thank our research team and collaborators Inger Margulies,Hong Zhou, Evgeny Arons, David FitzGerald, Alan Wayne, Richard Beers,JohnWeldon, Laiman Xiang, Byungkook Lee, andMasanori Onda. Principalinvestigators for the ALL trial included Drs. Alan Wayne, Deepa Bhojwani,and Lewis Silverman, and principal investigators for the HCL trial includedDrs. Martin Tallman, Steven Coutre, and Tadeusz Robak.

Grant Support

National Cancer Institute Intramural Program; development ofBL22 and moxetumomab was supported in part by MedImmune,LLC.

Received April 12, 2011; revised July 28, 2011; accepted July 29, 2011;published online October 14, 2011.

References1. Witzig TE. Radioimmunotherapy for B-cell non-Hodgkin lymphoma.

Best Pract Res Clin Haematol 2006;19:655–68.2. Steiner M, Neri D. Antibody-radionuclide conjugates for cancer ther-

apy: historical considerations and new trends. Clin Cancer Res2011;17:6406–16.

3. Teicher BA, Chari RVJ. Antibody conjugate therapeutics: challengesand potential. Clin Cancer Res 2011;17:6389–97.

4. LoRusso PM, Weiss D, Guardino E, Girish S, Sliwkowski MX. Trastu-zumab emtansine: a unique antibody-drug conjugate in developmentfor human epidermal growth factor receptor 2–positive cancer. ClinCancer Res 2011;17:6437–47.

5. Katz J, Janik JE, Younes A. Brentuximab vedotin (SGN-35). ClinCancer Res 2011;17:6428–36.

6. Blanc V, Bousseau A, Caron A, Carrez C, Lutz RJ, Lambert JM.SAR3419: an anti-CD19-maytansinoid immunoconjugate for the treat-ment of B-cell malignancies. Clin Cancer Res 2011;17:6448–58.

7. Ricart AD. Antibody-drug conjugates of calicheamicin derivative:gemtuzumab ozogamicin and inotuzumab ozogamicin. Clin CancerRes 2011;17:6417–27.

8. Williams P, Galipeau J. GM-CSF-based fusion cytokines as ligands forimmune modulation. J Immunol 2011;186:5527–32.

9. Carroll SF, Collier RJ. Active site ofPseudomonas aeruginosa exotoxinA. Glutamic acid 553 is photolabeled by NAD and shows functionalhomology with glutamic acid 148 of diphtheria toxin. J Biol Chem1987;262:8707–11.

10. Van Ness BG, Howard JB, Bodley JW. ADP-ribosylation of elongationfactor 2 by diphtheria toxin. Isolation and properties of the novelribosyl-amino acid and its hydrolysis products. J Biol Chem1980;255:10717–20.

11. Endo Y, Mitsui K, Motizuki M, Tsurugi K. The mechanism of action ofricin and related toxic lectins on eukaryotic ribosomes. The site and thecharacteristics of the modification in 28 S ribosomal RNA caused bythe toxins. J Biol Chem 1987;262:5908–12.

12. Yamaizumi M, Mekada E, Uchida T, Okada Y. One molecule ofdiphtheria toxin fragment A introduced into a cell can kill the cell. Cell1978;15:245–50.

13. Zamboni M, Brigotti M, Rambelli F, Montanaro L, Sperti S. High-pressure-liquid-chromatographic and fluorimetric methods for thedeterminationof adenine released from ribosomesby ricin andgelonin.Biochem J 1989;259:639–43.

14. Kreitman RJ. Getting plant toxins to fuse. Leuk Res 1997;21:997–9.15. Nitschke L.CD22andSiglec-G:B-cell inhibitory receptorswith distinct

functions. Immunol Rev 2009;230:128–43.16. Kreitman RJ, Hansen HJ, Jones AL, FitzGerald DJP, Goldenberg DM,

Pastan I. Pseudomonas exotoxin-based immunotoxins containing theantibody LL2 or LL2-Fab0 induce regression of subcutaneous humanB-cell lymphoma in mice. Cancer Res 1993;53:819–25.

17. KounnasMZ,Morris RE, ThompsonMR, FitzGerald DJ, StricklandDK,Saelinger CB. The a2-macroglobulin receptor/low density lipoproteinreceptor-relatedprotein binds and internalizesPseudomonasexotoxinA. J Biol Chem 1992;267:12420–3.

18. Hessler JL, Kreitman RJ. An early step in Pseudomonas exotoxinaction is removal of the terminal lysine residue, which allows bindingto the KDEL receptor. Biochemistry 1997;36:14577–82.

19. Du X, Beers R, Fitzgerald DJ, Pastan I. Differential cellular internaliza-tion of anti-CD19 and -CD22 immunotoxins results in different cyto-toxic activity. Cancer Res 2008;68:6300–5.

20. Ogata M, Fryling CM, Pastan I, FitzGerald DJ. Cell-mediated cleavageof Pseudomonas exotoxin between Arg279 and Gly280 generates theenzymatically active fragment which translocates to the cytosol. J BiolChem 1992;267:25396–401.

21. Kreitman RJ, Pastan I. Importance of the glutamate residue of KDEL inincreasing the cytotoxicity of Pseudomonas exotoxin derivatives andfor increased binding to the KDEL receptor. Biochem J 1995;307:29–37.

22. Theuer C, Kasturi S, Pastan I. Domain II of Pseudomonas exotoxin Aarrests the transfer of translocating nascent chains into mammalianmicrosomes. Biochemistry 1994;33:5894–900.

23. Theuer CP, Buchner J, FitzGerald D, Pastan I. The N-terminalregion of the 37-kDa translocated fragment of Pseudomonas exo-toxin A aborts translocation by promoting its own export aftermicrosomal membrane insertion. Proc Natl Acad Sci U S A 1993;90:7774–8.

24. Webb TR, Cross SH, McKie L, Edgar R, Vizor L, Harrison J, et al.Diphthamide modification of eEF2 requires a J-domain protein and isessential for normal development. J Cell Sci 2008;121:3140–5.

25. Brinkmann U, Brinkmann E, Gallo M, Pastan I. Cloning and charac-terization of a cellular apoptosis susceptibility gene, the human homo-logue to the yeast chromosome segregation gene CSE1. Proc NatlAcad Sci U S A 1995;92:10427–31.

26. Keppler-Hafkemeyer A, Kreitman RJ, Pastan I. Apoptosis induced byimmunotoxins used in the treatment of hematologicmalignancies. Int JCancer 2000;87:86–94.

27. Decker T, Oelsner M, Kreitman RJ, Salvatore G, Wang QC, Pastan I,et al. Induction of caspase-dependent programmed cell death in B-cellchronic lymphocytic leukemia by anti-CD22 immunotoxins. Blood2004;103:2718–26.

28. Bogner C, Dechow T, Ringshausen I, Wagner M, Oelsner M, Lutzny G,et al. Immunotoxin BL22 induces apoptosis in mantle cell lymphoma(MCL) cells dependent on Bcl-2 expression. Br J Haematol 2010;148:99–109.

29. Du X, Youle RJ, FitzGerald DJ, Pastan I. Pseudomonas exotoxinA-mediated apoptosis is Bak dependent and preceded by the degra-dation of Mcl-1. Mol Cell Biol 2010;30:3444–52.

30. Mansfield E, Chiron MF, Amlot P, Pastan I, FitzGerald DJ. Recombi-nant RFB4 single-chain immunotoxin that is cytotoxic towards CD22-positive cells. Biochem Soc Trans 1997;25:709–14.

31. Chaudhary VK, Queen C, Junghans RP, Waldmann TA, FitzGerald DJ,Pastan I. A recombinant immunotoxin consisting of two antibodyvariable domains fused to Pseudomonas exotoxin. Nature 1989;339:394–7.

32. Kreitman RJ, Batra JK, Seetharam S, Chaudhary VK, FitzGerald DJ,Pastan I. Single-chain immunotoxin fusions between anti-Tac andPseudomonas exotoxin: relative importance of the two toxin disulfidebonds. Bioconjug Chem 1993;4:112–20.

33. Brinkmann U, Pai LH, FitzGerald DJ, Willingham M, Pastan I. B3(Fv)-PE38KDEL, a single-chain immunotoxin that causes complete regres-sion of a human carcinoma in mice. Proc Natl Acad Sci U S A1991;88:8616–20.

34. Brinkmann U, Reiter Y, Jung SH, Lee B, Pastan I. A recombinantimmunotoxin containing a disulfide-stabilized Fv fragment. Proc NatlAcad Sci U S A 1993;90:7538–42.

35. Reiter Y, Brinkmann U, Kreitman RJ, Jung S-H, Lee B, Pastan I.Stabilization of the Fv fragments in recombinant immunotoxins by

CCRFOCUS

Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research6404

on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 8: Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

disulfide bonds engineered into conserved framework regions. Bio-chemistry 1994;33:5451–9.

36. Reiter Y, Kreitman RJ, Brinkmann U, Pastan I. Cytotoxic and antitumoractivity of a recombinant immunotoxin composed of disulfide-stabi-lized anti-Tac Fv fragment and truncatedPseudomonas exotoxin. Int JCancer 1994;58:142–9.

37. Mansfield E, Amlot P, Pastan I, FitzGerald DJ. Recombinant RFB4immunotoxins exhibit potent cytotoxic activity for CD22-bearing cellsand tumors. Blood 1997;90:2020–6.

38. KreitmanRJ,WangQC,FitzGeraldDJP, Pastan I.Complete regressionof human B-cell lymphoma xenografts in mice treated with recombi-nant anti-CD22 immunotoxin RFB4(dsFv)-PE38 at doses tolerated bycynomolgus monkeys. Int J Cancer 1999;81:148–55.

39. Kreitman RJ, Margulies I, Stetler-Stevenson M, Wang QC, FitzGeraldDJP, Pastan I. Cytotoxic activity of disulfide-stabilized recombinantimmunotoxin RFB4(dsFv)-PE38 (BL22) toward fresh malignant cellsfrom patients with B-cell leukemias. Clin Cancer Res 2000;6:1476–87.

40. KreitmanRJ,WilsonWH, BergeronK, RaggioM, Stetler-StevensonM,FitzGerald DJ, et al. Efficacy of the anti-CD22 recombinant immuno-toxin BL22 in chemotherapy-resistant hairy-cell leukemia. N Engl JMed 2001;345:241–7.

41. Kreitman RJ, Squires DR, Stetler-Stevenson M, Noel P, FitzGerald DJ,Wilson WH, et al. Phase I trial of recombinant immunotoxin RFB4(dsFv)-PE38 (BL22) in patients with B-cell malignancies. J Clin Oncol2005;23:6719–29.

42. Matsushita K, Margulies I, Onda M, Nagata S, Stetler-Stevenson M,Kreitman RJ. Soluble CD22 as a tumor marker for hairy cell leukemia.Blood 2008;112:2272–7.

43. KreitmanRJ, Stetler-StevensonM,Margulies I, Noel P, FitzgeraldDJP,Wilson WH, et al. Phase II trial of recombinant immunotoxin RFB4(dsFv)-PE38 (BL22) in patients with hairy cell leukemia. J Clin Oncol2009;27:2983–90.

44. Wayne AS, Kreitman RJ, Findley HW, Lew G, Delbrook C, SteinbergSM, et al. Anti-CD22 immunotoxin RFB4(dsFv)-PE38 (BL22) for CD22-positive hematologic malignancies of childhood: preclinical studiesand phase I clinical trial. Clin Cancer Res 2010;16:1894–903.

45. Salvatore G, Beers R, Margulies I, Kreitman RJ, Pastan I. Improvedcytotoxic activity toward cell lines and fresh leukemia cells of a mutantanti-CD22 immunotoxin obtained by antibody phage display. ClinCancer Res 2002;8:995–1002.

46. Alderson RF, Kreitman RJ, Chen T, Yeung P, Herbst R, Fox JA, et al.CAT-8015: a second-generation pseudomonas exotoxin A-basedimmunotherapy targeting CD22-expressing hematologic malignan-cies. Clin Cancer Res 2009;15:832–9.

47. Kreitman RJ, Tallman MS, Coutre SE, Robak T, Wilson WH, Stetler-Stevenson M, et al. Phase 1 trial of recombinant immunotoxin CAT-8015 (HA22) in multiply relapsed hairy cell leukemia. J Clin Oncol2010;28(May 20 Suppl):6523.

48. Wayne A, Bhojwani D, Richards K, Stetler-Stevenson M, SilvermanLB, Jeha S, et al. Complete remissions in 3 of 12 patients withpediatric acute lymphoblastic leukemia (ALL) during phase I testing

of the anti-CD22 immunotoxin moxetumomab pasudotox. Proceed-ings of the 52nd ASH Annual Meeting; 2010 Dec 4–7; Orlando, FL.3246.

49. Kreitman RJ, Tallman MS, Coutre SE, Robak T, Wilson WH, Stetler-Stevenson M, et al. A phase 1 study of moxetumomab pasudotox, ananti-CD22 recombinant immunotoxin. In: Relapsed/refractory hairycell leukemia (HCL). Updated Results. Proceedings of the AmericanSociety of Hematology Annual Meeting. Orlando, FL; December 4–7,2010. p. 2516.

50. Weldon JE, Xiang L, Chertov O, Margulies I, Kreitman RJ, FitzGeraldDJ, et al. A protease-resistant immunotoxin against CD22 with greatlyincreased activity against CLL and diminished animal toxicity. Blood2009;113:3792–800.

51. Baluna R, Rizo J, Gordon BE, Ghetie V, Vitetta ES. Evidence for astructural motif in toxins and interleukin-2 that may be responsible forbinding to endothelial cells and initiating vascular leak syndrome. ProcNatl Acad Sci U S A 1999;96:3957–62.

52. SmallshawJE,Ghetie V, Rizo J, Fulmer JR, Trahan LL,GhetieMA, et al.Genetic engineering of an immunotoxin to eliminate pulmonary vas-cular leak in mice. Nat Biotechnol 2003;21:387–91.

53. Onda M, Beers R, Xiang L, Nagata S, Wang QC, Pastan I. An immu-notoxin with greatly reduced immunogenicity by identification andremoval of B cell epitopes. Proc Natl Acad Sci U S A 2008;105:11311–6.

54. Pai LH, Wittes R, Setser A, Willingham MC, Pastan I. Treatment ofadvanced solid tumorswith immunotoxin LMB-1: an antibody linked toPseudomonas exotoxin. Nat Med 1996;2:350–3.

55. Hassan R, Bullock S, Premkumar A, Kreitman RJ, Kindler H, Will-inghamMC, et al. Phase I study of SS1P, a recombinant anti-mesothe-lin immunotoxin given as a bolus I.V. infusion to patientswithmesothe-lin-expressing mesothelioma, ovarian, and pancreatic cancers. ClinCancer Res 2007;13:5144–9.

56. Kreitman RJ, Hassan R, Fitzgerald DJ, Pastan I. Phase I trial ofcontinuous infusion anti-mesothelin recombinant immunotoxin SS1P.Clin Cancer Res 2009;15:5274–9.

57. Harding FA, SticklerMM,Razo J, DuBridgeRB. The immunogenicity ofhumanized and fully human antibodies: residual immunogenicityresides in the CDR regions. MAbs 2010;2:256–65.

58. Nagata S, Pastan I. Removal of B cell epitopes as a practical approachfor reducing the immunogenicity of foreign protein-based therapeu-tics. Adv Drug Deliv Rev 2009;61:977–85.

59. Onda M, Beers R, Xiang L, Lee B, Weldon JE, Kreitman RJ, et al. Arecombinant immunotoxin against B cell malignancies with no immu-nogenicity in mice by removal of B-cell epitopes. Proc Natl Acad SciU S A 2011;108:5742–7.

60. Chiron MF, Fryling CM, FitzGerald DJ. Cleavage of Pseudomonasexotoxin and diphtheria toxin by a furin-like enzyme prepared frombeef liver. J Biol Chem 1994;269:18167–76.

61. Fryling C, Ogata M, FitzGerald D. Characterization of a cellular pro-tease that cleaves Pseudomonas exotoxin. Infect Immun 1992;60:497–502.

Anti-CD22 Recombinant Immunotoxins

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6405

on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 9: Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin … · chain proteins, making them more amenable for construc-tion of recombinant chimeric proteins (14). Recombinant immunotoxins

2011;17:6398-6405. Clin Cancer Res   Robert J. Kreitman and Ira Pastan  Moxetumomab PasudotoxAntibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin

  Updated version

  http://clincancerres.aacrjournals.org/content/17/20/6398

Access the most recent version of this article at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/17/20/6398.full#ref-list-1

This article cites 59 articles, 40 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/17/20/6398.full#related-urls

This article has been cited by 27 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/17/20/6398To request permission to re-use all or part of this article, use this link

on December 3, 2020. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from