throbber
Antibody-basedtherapyofleukaemia
`
`http://www.expertreviews.org/
`
`expert reviews
`
`in molecular medicine
`
`Antibody-based therapy of leukaemia
`
`John C. Morris* and Thomas A. Waldmann
`Over the past decade, monoclonal antibodies have dramatically impacted the
`treatment of haematological malignancies, as evidenced by the effect of
`rituximab on the response rate and survival of patients with follicular and
`diffuse large B cell non-Hodgkin’s lymphoma. Currently, only two monoclonal
`antibodies – the anti-CD33 immunotoxin gemtuzumab ozogamicin and the
`CD52-directed antibody alemtuzumab – are approved for
`treatment of
`relapsed acute myeloid leukaemia in older patients and B cell chronic
`lymphocytic leukaemia,
`respectively. Although not approved for such
`treatment, alemtuzumab is also active against T cell prolymphocytic
`leukaemia, cutaneous T cell lymphoma and Se´ zary syndrome, and adult T cell
`leukaemia and lymphoma. In addition, rituximab has demonstrated activity
`against B cell chronic lymphocytic and hairy cell
`leukaemia. Monoclonal
`antibodies targeting CD4, CD19, CD20, CD22, CD23, CD25, CD45, CD66 and
`CD122 are now being studied in the clinic for the treatment of leukaemia.
`Here, we discuss how these new antibodies have been engineered to reduce
`immunogenicity and improve antibody targeting and binding.
`Improved
`interactions with Fc receptors on immune effector cells can enhance
`destruction of target cells through antibody-dependent cellular cytotoxicity
`and complement-mediated cell
`lysis. The antibodies can also be armed
`with cellular toxins or radionuclides to enhance the destruction of leukaemia cells.
`
`In his Croonian Lecture in 1900, the renowned
`immunologist Paul Ehrlich proposed that
`‘immunisations such as these which are of
`great
`theoretic
`interest may come
`to be
`available
`for
`clinical
`application attacking
`epithelium new formations,
`particularly
`carcinoma by means of specific anti-epithelial
`sera’ (Ref. 1). Unfortunately, Erlich’s dream of
`the ‘magic bullet’ of antibodies as a cancer
`treatment
`remained
`elusive
`until
`the
`groundbreaking work of Ko¨ hler and Milestein
`
`in the mid-1970s in which they developed
`techniques for generating specific monoclonal
`antibodies (Ref. 2). Despite the enthusiasm for
`antibodies as an anticancer
`therapy, early
`clinical results were discouraging. A number
`of
`improvements
`in
`understanding
`and
`biotechnology were
`required
`before
`the
`promise could be kept. Only now at
`the
`beginning of
`the twenty-first century have
`monoclonal antibodies come into their own as
`a treatment for cancer.
`
`Metabolism Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD USA.
`
`*Corresponding author: John C. Morris, Co-Director Clinical Trials, Metabolism Branch, Center for
`Cancer Research National Cancer Institute, Mark O. Hatfield Clinical Research Center, Room 4-5330,
`10 Center Drive, Bethesda, Maryland 20892-1457, USA. Tel: +1 301 402 2912; Fax: +1 301 402 1001;
`E-mail: jmorris@mail.nih.gov
`
`Accession information: doi:10.1017/S1462399409001215; Vol. 11; e29; September 2009
`& Cambridge University Press 2009
`
`1
`
`Downloaded from https://www.cambridge.org/core. IP address: 66.77.102.222, on 31 Jan 2022 at 15:34:09, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
`https://doi.org/10.1017/S1462399409001215
`
`DRL EXHIBIT 1027 PAGE 1
`
`

`

`Antibody-basedtherapyofleukaemia
`
`expert reviews
`
`in molecular medicine
`
`(1) its expression should be restricted to the
`leukaemic cells. If the antigen is expressed on
`normal cells,
`the loss of
`these cells should
`not result
`in serious complications such as
`life-threatening
`cytopaenias
`or
`prolonged
`immunosuppression; (2) the target should be
`expressed at high density on the leukaemic cells
`to provide an adequate number of antibody
`binding sites. Studies suggest
`that
`tumour
`responses correlate with target density. The
`lower responsiveness of CD20-expressing CLL
`to rituximab compared with follicular B cell
`NHL appears to be due to the lower level of
`CD20 expressed in CLL (Ref. 10). Escape
`mutants
`that
`lose
`antigen expression are
`unaffected because there is no target for the
`antibody to bind; (3) for unmodified or unarmed
`monoclonal antibodies, target antigens should
`not undergo internalisation (downmodulation).
`Internalisation
`of
`the
`antigen – antibody
`complex
`reduces
`the number
`of
`targets
`available
`for
`binding; however,
`antigen –
`antibody internalisation can be an advantage
`with immunotoxins. In addition, enhancement
`of antitumour activity is often seen when
`monoclonal antibodies are used in combination
`with cytotoxic chemotherapy (Refs 11, 12, 13).
`
`Monoclonal antibodies targeting myeloid
`leukaemia
`Acute myeloid leukaemia (AML) is the most
`common acute leukaemia of adults and it
`accounts for about 15% of childhood leukaemias
`(Ref. 14). At diagnosis, 55% of AML patients are
`age 65 or older. Between 50% and 80% of
`previously untreated patients achieve a complete
`remission with combinations of cytarabine and
`an anthracycline. For patients achieving a
`remission, up to 40% will survive for 5 years;
`however, these outcomes are worse for older
`patients. Therapeutic options are limited in
`relapsed patients over the age of 60 because of
`lower
`response rates,
`increased toxicity of
`treatment, and the frequent comorbidities present
`in this population. For older patients who failed
`to respond or relapsed after first-line therapy,
`the prognosis is often limited. The introduction of
`the CD33-targeted immunotoxin, gemtuzumab
`ozogamicin, as well as a number of novel
`monoclonal antibodies directed against other
`myeloid
`antigens
`currently
`in
`clinical
`development, offer new promise to these patients
`(Table 1).
`
`http://www.expertreviews.org/
`
`and Drug
`Food
`the US
`Currently,
`Administration (FDA) has
`approved nine
`monoclonal antibodies for
`the treatment of
`cancer. Of these, only two, alemtuzumab and
`gemtuzumab ozogamicin, have indications in
`leukaemia. Rituximab
`approved
`for
`the
`treatment of B cell non-Hodgkin’s lymphoma
`(NHL), has demonstrated activity in B cell
`chronic lymphocytic leukaemia (CLL) and hairy
`cell leukaemia (HCL) (Refs 3, 4). In addition, a
`number of antibodies directed against novel
`antigens, or
`that have been engineered to
`improve effector function, or armed with toxins
`or radioisotopes to increase killing ability are
`currently being studied in various leukaemias.
`The vast majority of
`early
`therapeutic
`monoclonal antibodies were of rodent origin, and
`therefore exhibited a number of unfavorable
`characteristics. Most notably,
`the nonhuman
`framework was immunogenic and frequently
`induced human antirodent antibody responses
`after very few treatments. This resulted in the
`loss of therapeutic effect, and increased the risk
`of infusional reactions and toxicity. In addition,
`nonhuman
`antibodies
`exhibit
`unfavorable
`pharmacokinetics with
`shorter
`half-lives
`and reduced Fc
`receptor binding that
`is
`required
`for
`antibody-dependent
`cellular
`cytotoxicity (ADCC) (Refs 5, 6). To overcome
`these problems, most approved therapeutic
`monoclonal antibodies have been modified
`using
`recombinant DNA technology
`to
`express human framework sequences to reduce
`immunogenicity and improve pharmacokinetics
`(Ref. 7). ‘Chimeric’ antibodies have undergone
`substitution of up to 70% of the nonhuman
`framework sequences. In ‘humanised’ antibodies,
`up to 90% of nonhuman sequences have been
`replaced,
`leaving only the original nonhuman
`complementary determining regions,
`further
`reducing immunogenicity. Advancements
`in
`transgenic
`technology
`have
`allowed
`the
`generation of monoclonal
`antibodies with
`fully ‘human’ sequences, high antigen affinities
`and little or no immunogenicity (Ref. 8). Further
`efforts have focused on engineering the effector
`function of antibodies by Fc subtype switching
`to improve the antibody’s ability to activate
`complement to lyse target cells, and to enhance
`antibody–Fc-receptor binding on macrophages
`and other effector cells to increase ADCC (Ref. 9).
`The ideal
`leukaemia antigen for antibody
`therapy should exhibit certain characteristics:
`
`Accession information: doi:10.1017/S1462399409001215; Vol. 11; e29; September 2009
`& Cambridge University Press 2009
`
`2
`
`Downloaded from https://www.cambridge.org/core. IP address: 66.77.102.222, on 31 Jan 2022 at 15:34:09, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
`https://doi.org/10.1017/S1462399409001215
`
`DRL EXHIBIT 1027 PAGE 2
`
`

`

`Antibody-basedtherapyofleukaemia
`
`http://www.expertreviews.org/
`
`expert reviews
`
`in molecular medicine
`
`CD33-targeted antibodies
`CD33 is a 67 kDa glycoprotein sialoadhesin
`receptor (Ref. 15). Its intracytoplasmic domain
`expresses two immunoreceptor tyrosine-based
`inhibitory motifs (ITIMs). CD33 is believed to
`have an important role in modulating leuko-
`cyte function, inhibiting cellular activation and
`proliferation,
`inducing
`apoptosis,
`and
`modulating cytokine secretion (Ref. 16). It is
`expressed on normal and maturing myeloid
`progenitor cells, but not on early normal
`haematopoietic stem cells. Over 90% of acute
`myeloid leukaemia blast cells express CD33, but
`it is not found on nonhaematopoietic tissues
`(Ref. 17).
`
`Gemtuzumab ozogamicin
`Gemtuzumab ozogamicin (MylotargTM, Wyeth,
`Madison, NJ)
`is an immunotoxin composed
`of a humanised IgG4k monoclonal antibody
`(hP67.6) that recognises CD33 conjugated to the
`antitumour antibiotic g1-calicheamicin, which is
`active against a broad spectrum of leukaemic
`
`lines. Gemtuzumab is
`and solid tumour cell
`derived from the murine anti-CD33 antibody
`p67.6 (Ref. 18) and it is linked to N-acetyl-g1-
`calicheamicin (Ref. 19). It is approved for the
`treatment of CD33-positive AML in patients 60
`years of age or older in first relapse.
`Upon binding to CD33, the antibody–receptor
`complex is internalised and g1-calicheamicin is
`released through hydrolysis of the N-acetyl linker
`(Ref. 20). Calicheamicin binds to the minor
`groove of DNA and results in site-specific
`double-stranded DNA breaks. Given that CD33
`stimulation is normally inhibitory, gentuzumab
`may also exert effects through receptor binding.
`In vitro, gemtuzumab ozogamicin is almost 105
`times more toxic to CD33-expressing cells than to
`cells lacking CD33 (Ref. 21).
`At least 85% saturation of CD33 on leukaemic
`blasts is achieved with an initial dose of 9 mg/m2.
`Saturating levels of the antibody are believed
`to enhance efficacy. After a 9 mg/m2 dose,
`gentuzumab achieves a mean peak plasma
`concentration (Cmax) of 2.86 1.35 mg/ml and has
`
`Table 1. Monoclonal antibodies for the treatment of myeloid leukaemia
`
`Monoclonal
`antibody
`
`Gemtuzumab
`ozogamicin
`(MyelotargTM,
`hP67.6)
`
`Lintuzumab
`(HuM195, SGN-33)
`
`Anti-CD44 (A3D8)
`
`Anti-CD45 (BC8,
`YTH 24.5,
`YAMEL568)
`
`Anti-CD66
`(BW250/183)
`
`Anti-CD123
`(26292(Fv)-PE38-
`KDEL)
`
`Target
`antigen
`
`CD33
`
`Isotype
`
`Humanised IgG4-
`g1-calicheamicin
`immunotoxin
`
`Approval and
`indication
`
`Refs
`
`FDA approved; AML
`
`18, 24
`
`CD33
`
`Humanised IgG1
`
`Phase III; AML
`
`Adhesion
`glycoprotein
`
`Leukocyte
`protein tyrosine
`phosphatase
`
`CEA family
`granulocyte
`antigen
`
`CD123 (IL-3Ra)
`
`Murine IgG1
`
`Radiolabelled 131I
`murine IgG1; 99mTc
`murine IgG2b; 111In rat
`IgG2a-imaging/
`biodistribution
`
`Radiolabelled 188Re 90Y
`murine IgG1
`
`Murine anti-CD123-Fv-
`Pseudomonas exotoxin A
`immunotoxin
`
`Preclinical
`development; AML
`
`Phase I–II; AML,
`allogeneic stem cell
`transplantation,
`imaging
`
`Phase I–II; AML,
`allogeneic stem cell
`transplantation
`
`Preclinical
`development; AML
`
`35
`
`46
`
`39, 40
`
`42,
`43, 44
`
`49
`
`Abbreviations: AML, acute myeloid leukaemia; CEA, carcinoembryonic antigen; FDA, Food and Drug
`Administration; I, iodine; IL-3Ra, interleukin-3 receptor-a; In, indium; Re, rhenium; Tc, technetium; Y, yttrium.
`
`Accession information: doi:10.1017/S1462399409001215; Vol. 11; e29; September 2009
`& Cambridge University Press 2009
`
`3
`
`Downloaded from https://www.cambridge.org/core. IP address: 66.77.102.222, on 31 Jan 2022 at 15:34:09, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
`https://doi.org/10.1017/S1462399409001215
`
`DRL EXHIBIT 1027 PAGE 3
`
`

`

`Antibody-basedtherapyofleukaemia
`
`expert reviews
`
`in molecular medicine
`
`lintuzumab
`CMC. Similarly to gentuzumab,
`undergoes internalisation. In a Phase III trial in
`191 adults with refractory AML or AML at
`first
`relapse,
`lintuzumab
`in
`combination
`with mitoxantrone,
`etoposide
`and cytosine
`arabinoside was compared with chemotherapy
`alone (Ref. 35). A response rate of 36% was
`observed in the
`combined antibody and
`chemotherapy group compared with 28% in
`those undergoing chemotherapy alone, with no
`difference in overall survival noted between the
`two groups. Both M195
`and lintuzumab
`conjugated to a- or b-particle-emitting isotopes
`have been used to purge bone marrow of
`leukaemic blasts in patients undergoing stem cell
`transplantation (Refs 36, 37).
`
`CD45-targeted antibodies
`CD45, also known as common leukocyte antigen,
`is a 200 kDa protein tyrosine phosphatase
`expressed at high levels on leukocytes and
`leukocyte precursors,
`including AML blasts
`(Ref. 38). CD45 functions as a regulator of T-
`and B-cell-receptor signalling. CD45 does not
`undergo internalisation. Anti-CD45
`labeled
`with 131I has been studied in Phase I and Phase
`II trials in AML patients as part of an allogeneic
`bone marrow transplant conditioning regimen
`in combination with cyclophosphamide and
`total body irradiation, or with busulfan and
`cyclophosphamide (Refs 39, 40). The 3 year
`disease-free survival rate was 61% in 46 treated
`patients. After adjusting for differences in risk,
`the hazard of mortality of the treatment group
`was 0.65 (95% CI, 0.39 – 1.08) compared with 509
`similar patients from the International Bone
`Marrow Transplant Registry, suggesting benefit
`from this approach (Ref. 40).
`
`CD66-targeted antibodies
`CD66 is a heavily glycosylated carcinoembryonic
`antigen (CEA)-related glycoprotein that
`is
`involved in adhesion to E-selectin surface
`glycoprotein. CD66 is expressed at high levels on
`granulocytes and it does not appear to be shed or
`internalised (Ref. 41). Radiolabeled anti-CD66
`with stem cell transplantation has been studied in
`AML (Refs 42, 43, 44). One trial compared the
`biodistribution of 99mTc-anti-CD66 versus 99mTc-
`anti-CD45 monoclonal antibodies in 12 AML
`patients undergoing stem cell transplantation and
`found that the anti-CD66 antibody (BW250/183)
`showed
`a
`favorable
`biodistribution
`and
`
`http://www.expertreviews.org/
`
`a plasma half-life of 72.4 42.0 hours (Ref. 22).
`Higher concentrations are observed with second
`doses. This is thought to be due to reduced
`tumour burden and lower availability of CD33 to
`bind gemtuzumab following the initial dose. The
`area under the curve (AUC) for g1-calicheamicin
`tracks with that of gemtuzumab, indicating that
`g1-calicheamicin remains
`conjugated to the
`antibody in the plasma.
`A Phase I trial defined the maximum tolerated
`dose of gemtuzumab ozogamicin administered as
`a 2 hour infusion as 9 mg/m2 (Ref. 23). The
`recommended treatment course is a total of two
`doses with at
`least 14 days between doses.
`A combined analysis of three open-label Phase II
`studies in 277 adults with recurrent CD33-
`positive AML reported an overall response rate of
`26% (Ref. 24). The response rate for patients 60
`years of age was 24% compared with 28% for
`those less than 60 years old. Gemtuzumab
`ozogamicin has also demonstrated activity as
`first-line single-agent therapy, as well as when
`used as consolidation therapy after remission-
`induction with chemotherapy, or used in
`combination with chemotherapy in CD33-postive
`AML (Refs 25, 26, 27, 28). Improved results were
`achieved when
`it was
`combined with
`chemotherapy at relapse over treatment with
`salvage
`chemotherapy alone
`(Refs 29, 30).
`Gemtuzumab ozogamicin combined with arsenic
`trioxide and all-trans-retinoic acid has also shown
`promise
`in
`recurrent
`acute promyelocytic
`leukaemia (Ref. 31).
`Toxicity includes chills, fever, nausea, vomiting,
`tachycardia, headaches,
`labile blood pressure,
`myelosuppression, stomatitis, elevation of liver
`transaminases and bilirubin, and rarely, hepatic
`veno-occlusive disease. Tumour lysis syndrome
`and acute adult respiratory distress syndrome
`have
`also
`been
`reported. Resistance
`to
`gemtuzumab ozogamicin has been correlated
`with expression of
`the multidrug resistance
`(MDR-1) p-glycoprotein and increased serum
`soluble CD33 antigen levels (Refs 32, 33, 34).
`
`Lintuzumab
`Lintuzumab (SGN-33, HuM195, Seattle Genetics,
`Bothell, WA) is a humanised IgG1 anti-CD33
`monoclonal antibody derived from the murine
`M195 antibody that exhibits a high affinity for
`CD33. Since it is humanised, lintuzumab is less
`immunogenic than M195. Unlike the IgG4-based
`gentuzumab,
`lintuzumab exhibits ADCC and
`
`Accession information: doi:10.1017/S1462399409001215; Vol. 11; e29; September 2009
`& Cambridge University Press 2009
`
`4
`
`Downloaded from https://www.cambridge.org/core. IP address: 66.77.102.222, on 31 Jan 2022 at 15:34:09, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
`https://doi.org/10.1017/S1462399409001215
`
`DRL EXHIBIT 1027 PAGE 4
`
`

`

`Antibody-basedtherapyofleukaemia
`
`expert reviews
`
`in molecular medicine
`
`underlying disease; it is greatest in follicular and
`diffuse large B cell lymphoma and lower in CLL
`and HCL. CD20 is nonmodulating and it is not
`shed, making it an ideal target for antibody
`therapy (Ref. 52).
`
`Rituximab
`Rituximab (RituxanTM, MabTheraTM, Genentech,
`South San Francisco, CA) is a chimeric anti-CD20
`monoclonal antibody. It was generated from the
`murine anti-human CD20 antibody 2B8 by
`cloning the light- and heavy-chain comple-
`mentary determining regions (CDR) on to a
`human IgG1 heavy-chain and kappa light-chain
`constant region (Ref. 53).
`In 1997, Rituximab
`became the first monoclonal antibody to be
`licensed for the treatment of cancer. Since then, it
`has
`revolutionised the treatment of B cell
`malignancies. Rituximab is approved by the FDA
`for: (1) the treatment of relapsed or refractory,
`low-grade or follicular, CD20-positive, B cell
`NHL as a single agent; (2) previously untreated
`follicular, CD20-positive, B cell NHL in
`combination
`with
`chemotherapy;
`(3)
`nonprogressing low-grade, CD20-positive B cell
`NHL,
`as
`a
`single
`agent,
`after first-line
`chemotherapy, and (4) previously untreated
`diffuse large B cell, CD20-positive NHL in
`combination with CHOP (cyclophosphamide,
`doxorubicin, vincristine, prednisone) or other
`anthracycline-based chemotherapy regimens.
`Rituximab exhibits powerful B-cell-depleting
`properties. The mechanism of action is complex
`and may vary by disease
`(Ref.
`54).
`Its
`mechanism includes
`induction of apoptosis,
`inhibition of cell growth, complement-mediated
`cytotoxicity
`(CMC),
`sensitisation
`to
`both
`chemotherapy
`and radiation,
`and ADCC.
`Rituximab enhances sensitivity to chemotherapy
`by downregulating expression of Bcl-2 (Ref. 55).
`Some preclinical work suggested that
`full
`cytotoxic activity could only be achieved in
`mice expressing the stimulatory g-chain of the
`FcgRIII receptor (Ref. 56). Activation of allelic
`variants of the FcgRIIIa gene correlates with an
`improved response to rituximab in patients
`with follicular NHL, but not in patients with
`CLL (Refs 57, 58). Rituximab enhances caspase-
`9, caspase-3 and poly(ADP-ribose) polymerase
`(PARP) cleavage in CLL cells, suggesting that
`induction of
`apoptosis
`is
`the dominant
`mechanism of action in CLL, whereas CMC and
`ADCC appear to be the dominant effects in
`
`http://www.expertreviews.org/
`
`pharmacokinetics compared with the anti-CD45
`antibody (Ref. 45). Zenz and colleagues treated 20
`adults with allogeneic stem cell transplantation
`(188Re)-labeled anti-CD66
`and Rhenium-188
`for
`Philadelphia-chromosome-positive
`acute
`lymphoblastic leukaemia or advanced chronic
`myeloid leukaemia with acceptable toxicity;
`however, no reduction in the incidence of
`leukaemic relapse was observed (Ref. 44).
`
`Antibodies targeted against myeloid
`leukaemia stem cell antigen (CD44 and
`CD123)
`CD44 (phagocytic glycoprotein I) and CD123
`(interleukin-3 receptor a) represent potential
`targets for antibody-based therapy of myeloid
`leukaemia (Refs 46, 47). Cancer stem cells are
`increasingly
`recognised
`as
`a
`specialised
`population
`of
`selfrenewing
`tumour
`cells
`(Ref. 48). Ultimate success might
`lie in our
`ability to effectively target leukaemia stem cells.
`the a-subunit of
`CD123,
`the interleukin-3
`receptor (IL-3Ra) is highly expressed on various
`blasts and leukaemic stem cells. Du and co-
`workers studied the activity of a series of
`single-chain Fv antibody fragments fused to a
`38 kDa fragment of Pseudomonas exotoxin A
`(Ref. 49). One of these, 26292(Fv)-PE38-KDEL
`was cytotoxic to several CD123-expressing cell
`lines in vitro.
`
`Monoclonal antibodies targeting lymphoid
`leukaemia: B cell leukaemia antigens
`A list of antibodies currently in development or
`clinical
`trial
`for
`the
`treatment of B cell
`leukaemia is provided in Table 2. The various
`antibodies are considered in detail within their
`respective groups below.
`
`CD20-targeted antibodies
`CD20 is a 31–37 kDa antigen expressed on almost
`all normal B cells and most B cell malignancies
`(Ref. 50). It is a nonglycosylated phosphoprotein
`member of the membrane-spanning 4A gene
`family.
`Its function is unclear, but CD20 is
`believed to have a role in B cell activation and
`regulation of B cell growth, as well as acting as a
`cell membrane calcium channel (Ref. 51). CD20 is
`expressed at almost all
`stages of B cell
`development,
`except
`in pro-B cells
`and
`terminally differentiated plasma cells. CD20 is
`expressed on the malignant B cells of NHL, CLL
`and HCL. The number of targets varies with the
`
`Accession information: doi:10.1017/S1462399409001215; Vol. 11; e29; September 2009
`& Cambridge University Press 2009
`
`5
`
`Downloaded from https://www.cambridge.org/core. IP address: 66.77.102.222, on 31 Jan 2022 at 15:34:09, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
`https://doi.org/10.1017/S1462399409001215
`
`DRL EXHIBIT 1027 PAGE 5
`
`

`

`http://www.expertreviews.org/
`
`expert reviews
`
`in molecular medicine
`
`Table 2. Monoclonal antibodies for the treatment of B cell leukaemia
`
`Monoclonal antibody
`
`XmAb 5574
`
`Rituximab (RituxanTM,
`MabTheraTM, C2B8)
`
`Ofatumumab
`(HuMax-CD20TM)
`
`Ocrelizumab
`
`Target
`antigen
`
`CD19
`
`Isotype
`
`Approval and
`indication
`
`Humanised and Fc
`enhanced mouse IgG1
`
`Preclinical
`development
`
`CD20
`
`Humanised mouse IgG1
`
`CD20
`
`Fully human IgG1
`
`CD20
`
`Humanised IgG1
`
`Veltuzumab
`(IMMUNO-106, hA20)
`
`CD20
`
`Humanised and
`enhanced IgG2a
`
`Epratuzumab (hLL2)
`
`CD22
`
`Humanised IgG1
`
`FDA approved;
`follicular and diffuse
`large B cell NHL
`
`Phase I–II; follicular
`B cell NHL
`
`Phase II; follicular B
`cell NHL
`
`Phase I; follicular B
`cell NHL and CLL
`
`Phase I–II; follicular
`B cell NHL
`
`Antibody-basedtherapyofleukaemia
`
`Refs
`
`92
`
`3, 63
`
`79, 80
`
`83, 84
`
`85, 86
`
`99, 103
`
`107
`
`CD22
`
`CD22
`
`CD23
`
`Inotuzumab ozogamicin
`(CMC-544, G5/44)
`
`BL22, HA22
`
`Lumiliximab
`
`Alemtuzumab
`(CampathTM,
`MabCampathTM,
`Campath-1H)
`
`Humanised IgG4–N-
`acetyl-g1-calicheamicin
`
`Preclinical
`development
`
`Murine IgG1 Fv(RFB4)-
`Pseudomonas exotoxin
`A immunotoxin
`
`Cynomolgus macaque–
`human IgG1
`
`Phase I–II; HCL and
`B cell CLL
`
`104, 106
`
`Phase I–II–III; CLL
`
`111, 113
`
`CD52
`
`Humanised rat IgG2b
`
`FDA approved; B cell
`CLL
`
`115, 119
`
`Abbreviations: CLL, chronic lymphocytic leukaemia; FDA, Food and Drug Administration; HCL, hairy cell
`leukaemia; NHL, non-Hodgkin’s lymphoma.
`
`NHL (Ref. 59). Mechanisms of resistance to
`rituximab are unclear, but might
`involve
`alterations
`in CD20
`expression,
`reduced
`antibody
`penetration
`of
`tumour masses,
`increased expression of complement inhibitors,
`enhanced prosurvival pathways and inhibition
`of ADCC.
`Phase I studies failed to identify a maximum
`tolerated dose
`and established rituximab
`375 mg/m2/week for 4 weeks as standard (Refs
`60, 61). Pharmacokinetic studies showed a
`plasma half-life of 76.3 hours achieved with the
`first infusion that increased to 205.8 hours after
`the fourth infusion (Ref. 62). The efficacy of
`rituximab was established in a pivotal Phase II
`trial
`in 166 patients with relapsed NHL
`
`In low-grade and follicular B cell
`(Ref. 63).
`NHL patients who relapsed or failed primary
`therapy, extending treatment to eight weekly
`doses appeared to increase the number of
`responses,
`improve complete response rate,
`time
`to progression and the duration of
`response (Ref. 64). The 8 week course is
`often used for
`the treatment of bulky or
`resistant disease, or for patients undergoing
`retreatment.
`Rituximab is not approved for the treatment of
`CLL or other B cell
`leukaemias; however, a
`substantial number of studies demonstrated its
`activity in previously treated and untreated
`CLL (Refs 3, 65, 66, 67). One early trial treated a
`variety of malignant B cell disorders including
`
`Accession information: doi:10.1017/S1462399409001215; Vol. 11; e29; September 2009
`& Cambridge University Press 2009
`
`6
`
`Downloaded from https://www.cambridge.org/core. IP address: 66.77.102.222, on 31 Jan 2022 at 15:34:09, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
`https://doi.org/10.1017/S1462399409001215
`
`DRL EXHIBIT 1027 PAGE 6
`
`

`

`Antibody-basedtherapyofleukaemia
`
`expert reviews
`
`in molecular medicine
`
`74). Rituximab has also demonstrated activity in
`a number of other B cell leukaemias including
`hairy cell leukaemia (HCL), where response rates
`of 25–80% have been reported (Refs 75, 76).
`Rituximab toxicities include acute infusional
`reactions,
`fever,
`chills,
`nausea,
`vomiting,
`bronchospasm and hypotension, most often
`with the initial infusion. Other serious adverse
`events include the potentially fatal cytokine
`release syndrome (Ref. 77). B cell
`counts
`nadir 24 –72 hours after the initial rituximab
`infusion and begin to return to normal values
`at an average of 9 months after
`therapy.
`The risk of serious infection is marginally
`increased in the months following rituximab
`treatment.
`
`Ofatumumab
`(ArzzeraTM, GlaxoSmithKline,
`Ofatumumab
`Philadelphia, PA; HuMax-CD20TM, Genmab,
`Copenhagen, Denmark) is a second-generation
`anti-CD20 monoclonal antibody (Ref. 78). The
`lower levels of CD20 expression in CLL and
`other B cell malignancies
`suggest
`that an
`antibody designed with enhanced Fc effector
`function may be required. Based on this, a
`number of CD20-targeted monoclonal antibodies
`have been generated with enhanced effector
`IgG1k immunoglobulin
`function using the
`framework. These enhanced antibodies strongly
`bind to CD20-positive cells, efficiently recruit
`mononuclear
`cells
`(ADCC) and lyse
`even
`rituximab-resistant targets (Ref. 79). Ofatumumab
`exhibits
`a higher
`affinity for CD20
`than
`rituximab; it attaches to a different epitope closer
`to the cell surface, which makes it easier for
`effector cells to kill the target cell and activate
`complement.
`Ofatumumab has shown activity in low-grade
`follicular B cell NHL and is currently in clinical
`trials for CLL (Refs 80, 81). One hundred and
`thirty-eight patients with fludarabine
`and
`alemtuzumab refractory (double refractory, DR)
`or bulky (.5 cm lymph nodes) fludarabine-
`refractory (BFR) CLL received eight weekly
`infusions of ofatumumab followed by four
`monthly infusions (Ref. 82). The overall response
`rate was 51% for the DR group and 44% for the
`BFR group, with one patient achieving a
`complete response. Serious toxicities included
`infections,
`granulocytopaenia
`and anaemia.
`An FDA Oncologic Drugs Advisory Committee
`has
`recommended accelerated approval of
`
`http://www.expertreviews.org/
`
`15 patients with small lymphocytic lymphoma or
`chronic lymphocytic leukaemia (SLL/CLL) and
`mantle cell
`lymphoma (MCL)
`in leukaemic
`phase with rituximab 375 mg/m2/week for 4
`weeks, and concluded that
`the efficacy of
`rituximab against SLL/CLL and MCL was
`limited (Ref.
`68). O’Brien and colleagues
`examined the efficacy of higher doses rituximab
`in CLL (Ref. 69). Forty patients with CLL and
`ten with other mature B cell leukaemias were
`treated with four weekly infusions of rituximab.
`The initial dose was 375 mg/m2 in all patients,
`with the subsequent doses escalated in groups
`of patients from 500 to 2250 mg/m2. Toxicity
`with the first dose was significantly more
`common in patients with non-CLL B cell
`leukaemias, occurring in 5 out of 10 patients,
`versus 1 out of 40 patients with CLL. The
`overall response rate was 36% in CLL and 60%
`in the other B cell
`leukaemias. The lower
`responsiveness
`of CLL to
`rituximab was
`attributed to the lower density of CD20 targets on
`CLL cells
`compared with
`other B cell
`malignancies (Ref. 70). Similarly to follicular
`and diffuse large B cell NHL, combinations
`of rituximab and chemotherapy demonstrated
`improved
`responses
`and
`progression-free
`survival compared with treatment with either
`chemotherapy or rituximab alone in CLL. The
`CALGB 9712 clinical trial randomised concurrent
`treatment with fludarabine and rituximab versus
`sequential
`treatment
`in 104 symptomatic CLL
`patients (Ref. 71). Patients receiving concurrent
`treatment experienced greater haematological
`toxicity and infusional reactions compared with
`the sequential
`treatment group; however,
`the
`response rate of the concurrent group was 90%
`compared with 77% in the sequential treatment
`group. In a retrospective analysis, these data were
`combined with that of a second multicenter
`clinical trial of fludarabine in CLL (CALGB 9011)
`(Ref. 72). In multivariate analyses controlling for
`pretreatment characteristics, patients receiving
`fludarabine and rituximab had a significantly
`better progression-free survival (P , 0.0001) and
`(P ¼ 0.0006)
`overall
`survival
`than patients
`receiving fludarabine alone. The number of
`infectious events was similar between the two
`treatments. A number of clinical trials in CLL of
`rituximab in combination with fludarabine and
`cyclophosphamide at varying drug doses, or
`sequential or concurrent treatment, demonstrated
`molecular responses in excess of 85% (Refs 73,
`
`Accession information: doi:10.1017/S1462399409001215; Vol. 11; e29; September 2009
`& Cambridge University Press 2009
`
`7
`
`Downloaded from https://www.cambridge.org/core. IP address: 66.77.102.222, on 31 Jan 2022 at 15:34:09, subject to the Cambridge Core terms of use, available at https://www.cambridge.org/core/terms.
`https://doi.org/10.1017/S1462399409001215
`
`DRL EXHIBIT 1027 PAGE 7
`
`

`

`Antibody-basedtherapyofleukaemia
`
`expert reviews
`
`in molecular medicine
`
`XmAb 5574
`is a
`XmAb 5574 (Xencor, Monrovia, CA)
`humanised anti-CD19 monoclonal antibody that
`has an engineered Fc domain to increase
`binding to Fcg receptors and enhance Fc-
`mediated effector function. In vitro, XmAb 5574
`enhanced ADCC 1000-fold relative to an anti-
`CD19 IgG1 analogue against a broad range of
`B cell
`lymphoma and leukaemia cell
`lines
`including primary ALL and mantle
`cell
`lymphoma (Ref. 92). XmAb 5574 increased
`effector
`cell phagocytosis
`and target
`cell
`apoptosis.
`In vivo, XmAb 5574
`inhibited
`tumour growth in established lymphoma
`mouse xenograft models, and demonstrated
`more potent antitumour activity than its IgG1
`analogue. Comparison with a variant antibody
`incapable of Fcg-receptor binding showed that
`engagement of these receptors was critical for
`efficacy. In a nonhuman primate model, XmAb
`5574 depleted B cells from the peripheral blood,
`lymph nodes and spleen (Ref. 93). XmAb 5574
`has not yet entered clinical trials.
`Anti-CD19 antibodies and immunomagnetic
`separation have been used to deplete B cells
`from the bone marrow in CLL patients
`undergoing autologous transplants (Refs 94, 95,
`96). In one study, 20 patients with poor-risk
`CLL underwent stem-cell mobilisation with
`chemotherapy
`and
`granulocyte-colony
`stimulating factor (Ref. 96). B cell depletion was

`cells and
`performed using selection for CD34
`selection against B cells with anti-CD19, -CD20,
`-CD23 and -CD37 immuno- magnetic beads.
`With a median follow-up of 20 months, 17
`patients were in complete remission.
`
`Anti-CD19 immunotoxins
`The internalisation of CD19 after antibody
`binding suggests that an immunotoxin might be
`an effective strategy for treatment of CD19-
`expressing leukaemias. One study examined the
`use of an anti-CD19-based immunotoxin in
`acute
`lymphocytic
`leukaemia
`(ALL)
`for
`eradication of residual disease after treatment
`(Ref. 97). After initial chemotherapy, 46 adults
`with previously untreated CD19-positive ALL
`received an immunotoxin composed of
`the
`murine B4
`anti-CD19
`antibody linked to
`modified ricin. The most
`frequent
`toxicities
`were transient elevation of liver transaminases
`and lymphopaenia. Molecular studies, however,
`failed to show a consistent decrease in the
`
`http://www.expertreviews.org/
`
`ofatumumab for the treatment of fludarabine and
`alemtuzumab-refractory CLL.
`
`Ocrelizumab
`Ocrelizumab (rhumAb 2H7v.16, Biogen-IDEC,
`Cambridge, MA) is a recombinant humanised
`anti-human CD20 antibody (Ref. 83)

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket