AU2013257387A1 - Monoclonal antibodies against glypican-3 - Google Patents

Monoclonal antibodies against glypican-3 Download PDF

Info

Publication number
AU2013257387A1
AU2013257387A1 AU2013257387A AU2013257387A AU2013257387A1 AU 2013257387 A1 AU2013257387 A1 AU 2013257387A1 AU 2013257387 A AU2013257387 A AU 2013257387A AU 2013257387 A AU2013257387 A AU 2013257387A AU 2013257387 A1 AU2013257387 A1 AU 2013257387A1
Authority
AU
Australia
Prior art keywords
antibody
seq
glypican
human
variable region
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2013257387A
Inventor
Haichun Huang
Heidi Leblanc
Li-Sheng Lu
Chin Pan
Timothy Sproul
Jonathan Alexander Terrett
Mark Yamanaka
Dapeng Yao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ER Squibb and Sons LLC
Original Assignee
ER Squibb and Sons LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2008275985A external-priority patent/AU2008275985B2/en
Application filed by ER Squibb and Sons LLC filed Critical ER Squibb and Sons LLC
Priority to AU2013257387A priority Critical patent/AU2013257387A1/en
Publication of AU2013257387A1 publication Critical patent/AU2013257387A1/en
Assigned to E. R. SQUIBB & SONS, L.L.C. reassignment E. R. SQUIBB & SONS, L.L.C. Alteration of Name(s) of Applicant(s) under S113 Assignors: MEDAREX, INC
Abandoned legal-status Critical Current

Links

Abstract

Provided are isolated monoclonal antibodies, particularly human monoclonal antibodies that specifically bind to Glypican-3 with high affinity. Nucleic acid molecules encoding Glypican-3 antibodies, expression vectors, host cells and methods for expressing the Glypican-3 antibodies are also provided. Immunoconjugates, bispecific molecules and pharmaceutical compositions comprising Glypican-3 antibodies are also disclosed. Method for detecting Glypican-3, as well as methods for treating various Glypican-3 related conditions, including heptacellular cancer, are disclosed.

Description

MXI-347PC Regulation 3.2 5 AUSTRALIA PATENTS ACT, 1990 COMPLETE SPECIFICATION 10 FOR A STANDARD PATENT DIVISIONAL 15 20 25 30 Name of Applicant: MEDAREX, INC Actual Inventors: TERRETT, Jonathan Alexander; LU, Li-sheng; HUANG, Haichun; YAO, Dapeng; PAN, Chin; LEBLANC, Heidi; SPROUL, Timothy; YAMANAKA, Mark Address for service AJ PARK, Level 11, 60 Marcus Clarke Street, Canberra ACT in Australia: 2601, Australia Invention Title: Monoclonal Antibodies Against Glypican-3 Original Application: Australian patent application 2008275985 dated 17 July 2008 The following statement is a full description of this invention, including the best method of performing it known to us. 35 1 MXI-347PC Monoclonal Antibodies Against Glypican-3 Background of the Invention Glypican-3 is an oncofetal antigen that belongs to the glypican family of 5 glycosyl-phosphatidylinositol-anchored heparin sulfate proteoglycans. Glypicans are characterized by a covalent linkage to complex polysaccharide chains called heparinsulphate glycosaminoglycans. Glypicans are involved in cell signaling at the cellular-extracellular matrix interface. (Sasisekharan et al., Nature Reviews | Cancer, Volume 2 (2002).) To date, six distinct members of the human glypican family have 10 been identified. Cell membrane-bound Glypican-3 is composed of two subunits, linked by one or more disulfide bonds. Glypican-3 is expressed in fetal liver and placenta during development and is down-regulated or silenced in normal adult tissues. Mutations and depletions in the Glypican-3 gene are responsible for the Simpson-Golabi-Behmel or Simpson 15 dysmorphia syndrome in humans. Glypican-3 is expressed in various cancers and, in particular, hepatocellular carcinoma ("HCC"), melanoma, Wilm's tumor, and hepatoblastoma. (Jakubovic and Jothy; Ex. Mol. Path. 82:184-189 (2007); Nakatsura and Nishimura, Biodrugs 19(2):71-77 (2005).) HCC is the third leading cause of cancer-related deaths worldwide. Each year, 20 HCC accounts for about 1 million deaths. (Nakatsura and Nishimura, Biodrugs 19(2):71-77 (2005).) Hepatitis B virus, hepatitis C virus, and chronic heavy alcohol use leading to cirrhosis of the liver remain the most common causes of HCC. Its incidence has increased dramatically in the United States because of the spread of hepatitis C virus infection and is expected to increase for the next 2 decades. HCC is treated primarily by 25 liver transplantation or tumor resection. Patient prognosis is dependent on both the underlying liver function and the stage at which the tumor is diagnosed. (Parikh and Hyman, Am JMed. 120(3):194-202 (2007).) Effective HCC treatment strategies are needed. Summary of the Invention 30 The present invention provides isolated monoclonal antibodies, in particular human monoclonal antibodies, that bind to Glypican-3 and that exhibit numerous desirable properties. These properties include high affinity binding to human Glypican 3. 2 MXI-347PC In one aspect, the invention pertains to an isolated monoclonal antibody, or an antigen-binding portion thereof, wherein the antibody: (a) binds to human Glypican-3 with a KD Of 1X10 7 M or less; (b) binds to human CHO cells transfected with Glypican-3. 5 Preferably the antibody is a human antibody, although in alternative embodiments the antibody can be a murine antibody, a chimeric antibody or humanized antibody. In one embodiment, the antibody binds to human Glypican-3 with a KD of 5 x 10-8 M or less, binds to human Glypican-3 with a KD of 2 x 10-8 M or less, binds to 10 human Glypican-3 with a KD Of 1 x 10-8 M or less, binds to human Glypican-3 with a KD of 5x10- 9 M or less, binds to human Glypican-3 with a KD of 4x10- 9 M or less, binds to human Glypican-3 with a KD of 3x10- 9 M or less, or binds to human Glypican-3 with a KD of 2 x 10~9 M or less. In another embodiment, the invention provides an isolated monoclonal antibody, 15 or antigen binding portion thereof, wherein the antibody cross-competes for binding to Glypican-3 with a reference antibody comprising: (a) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:19, 20, and 21; and 20 (b) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:22, 23, and 24. In various embodiments, the reference antibody comprises: (a) a heavy chain variable region comprising the amino acid sequence of SEQ 25 ID NO:19; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO:22; or the reference antibody comprises: 3 MXI-347PC (a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO:20; and (b) a light chain variable region comprising the amino acid sequence of SEQ 5 ID NO:23. or the reference antibody comprises: (a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO:21; and 10 (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO:24. In another aspect, the invention provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a heavy chain variable region that is the 15 product of or derived from a human VH 5-51 gene, wherein the antibody specifically binds Glypican-3. The invention also provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a light chain variable region that is the product of or derived from a human VK A27 gene, wherein the antibody specifically binds Glypican-3. 20 In a preferred embodiment, the invention provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising: (a) a heavy chain variable region of a human VH5-51 gene; and (b) a light chain variable region of a human VK A27 gene; wherein the antibody 25 specifically binds to Glypican-3. In another aspect, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising: a heavy chain variable region that comprises CDR1, CDR2, and CDR3 sequences; and a light chain variable region that comprises CDR1, CDR2, and CDR3 30 sequences, wherein: 4 MXI-347PC (a) the heavy chain variable region CDR3 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs:7, 8, and 9, and conservative modifications thereof; (b) the light chain variable region CDR3 sequence comprises an amino acid 5 sequence selected from the group consisting of amino acid sequence of SEQ ID NOs:16, 17, and 18, and conservative modifications thereof; (c) the antibody binds to human Glypican-3 with a KD Of 1X10 7 M or less; or (d) binds to human CHO cells transfected with Glypican-3. Preferably, the heavy chain variable region CDR2 sequence comprises an amino 10 acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs:4, 5, and 6, and conservative modifications thereof; and the light chain variable region CDR2 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs:13, 14, and 15, and conservative modifications thereof. Preferably, the heavy chain variable region CDR1 sequence 15 comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs:1, 2, and 3, and conservative modifications thereof; and the light chain variable region CDR1 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs:10, 11, and 12, and conservative modifications thereof. 20 A preferred combination comprises: (a) a heavy chain variable region CDR1 comprising SEQ ID NO: 1; (b) a heavy chain variable region CDR2 comprising SEQ ID NO:4; (c) a heavy chain variable region CDR3 comprising SEQ ID NO:7; (d) a light chain variable region CDR1 comprising SEQ ID NO:10; 25 (e) a light chain variable region CDR2 comprising SEQ ID NO:13; and (f) a light chain variable region CDR3 comprising SEQ ID NO:16. Another preferred combination comprises: (a) a heavy chain variable region CDR1 comprising SEQ ID NO:2; (b) a heavy chain variable region CDR2 comprising SEQ ID NO:5; 30 (c) a heavy chain variable region CDR3 comprising SEQ ID NO:8; (d) a light chain variable region CDR1 comprising SEQ ID NO: 11; (e) a light chain variable region CDR2 comprising SEQ ID NO:14; and (f) a light chain variable region CDR3 comprising SEQ ID NO:17. 5 MXI-347PC Another preferred combination comprises: (a) a heavy chain variable region CDR1 comprising SEQ ID NO:3; (b) a heavy chain variable region CDR2 comprising SEQ ID NO:6; (c) a heavy chain variable region CDR3 comprising SEQ ID NO:9; 5 (d) a light chain variable region CDR1 comprising SEQ ID NO:12; (e) a light chain variable region CDR2 comprising SEQ ID NO:15; and (f) a light chain variable region CDR3 comprising SEQ ID NO: 18. Other preferred antibodies of the invention, or antigen binding portions thereof comprise: 10 (a) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 19, 20 and 21; and (b) a light chain variable region comprising an amino acid sequence selected from 15 the group consisting of SEQ ID NOs:22, 23, and 24; wherein the antibody specifically binds Glypican-3. A preferred combination comprises: (a) a heavy chain variable region comprising the amino acid sequence of SEQ ID 20 NO:19; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO:22. Another preferred combination comprises: 25 (a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO:20; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID 30 (c) NO:23. Another preferred combination comprises: 6 MXI-347PC (a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO:21; and (b) a light chain variable region comprising the amino acid sequence of SEQ 5 ID NO:24. In another aspect of the invention, antibodies, or antigen-binding portions thereof, are provided that compete for binding to Glypican-3 with any of the aforementioned antibodies. 10 The antibodies of the invention can be, for example, full-length antibodies, for example of an IgGI or IgG4 isotype. Alternatively, the antibodies can be antibody fragments, such as Fab, Fab' or Fab'2 fragments, or single chain antibodies. The invention also provides an immunoconjugate comprising an antibody of the invention, or antigen-binding portion thereof, linked to a therapeutic agent, such as a 15 cytotoxin or a radioactive isotope. The invention also provides a bispecific molecule comprising an antibody, or antigen-binding portion thereof, of the invention, linked to a second functional moiety having a different binding specificity than said antibody, or antigen binding portion thereof. Compositions comprising an antibody, or antigen-binding portion thereof, or 20 immunoconjugate or bispecific molecule of the invention and a pharmaceutically acceptable carrier are also provided. Nucleic acid molecules encoding the antibodies, or antigen-binding portions thereof, of the invention are also encompassed by the invention, as well as expression vectors comprising such nucleic acids and host cells comprising such expression vectors. 25 The invention provides methods of inhibiting the growth of tumor cells expressing Glypican-3 The methods comprise contacting the cells with an anti Glypican-3 antibody, or antigen-binding portion thereof, in an amount effective to inhibit growth of the tumor cells. In a preferred embodiment, the tumor cells are derived from liver tissue. Additionally, the antibody, or antigen-binding portion thereof, may be 30 immunoconjugates. The immunoconjugates of the invention may be therapeutic agents, for example, cytotoxins or radioactive isotopes. 7 MXI-347PC Other features and advantages of the instant invention will be apparent from the following detailed description and examples which should not be construed as limiting. The contents of all references, Genbank entries, patents and published patent applications cited throughout this application are expressly incorporated herein by 5 reference. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1A shows the nucleotide sequence (SEQ ID NO:25) and amino acid sequence (SEQ ID NO:19) of the heavy chain variable region of the 4A6 human monoclonal antibody. The CDR1 (SEQ ID NO:1), CDR2 (SEQ ID NO:4) and CDR3 10 (SEQ ID NO:7) regions are delineated and the V, D and J germline derivations are indicated. Figure lB shows the nucleotide sequence (SEQ ID NO:28) and amino acid sequence (SEQ ID NO:22) of the light chain variable region of the 4A6 human monoclonal antibody. The CDR1 (SEQ ID NO:10), CDR2 (SEQ ID NO:13) and CDR3 15 (SEQ ID NO: 16) regions are delineated and the V and J germline derivations are indicated. Figure 2A shows the nucleotide sequence (SEQ ID NO:26) and amino acid sequence (SEQ ID NO:20) of the heavy chain variable region of the 1 1E7 human monoclonal antibody. The CDR1 (SEQ ID NO:2), CDR2 (SEQ ID NO:5) and CDR3 20 (SEQ ID NO:8) regions are delineated and the V and J germline derivations are indicated. Figure 2B shows the nucleotide sequence (SEQ ID NO:29) and amino acid sequence (SEQ ID NO:23) of the light chain variable region of the 1 1E7 human monoclonal antibody. The CDR1 (SEQ ID NO: 11), CDR2 (SEQ ID NO:14) and CDR3 25 (SEQ ID NO: 17) regions are delineated and the V and J germline derivations are indicated. Figure 3A shows the nucleotide sequence (SEQ ID NO:27) and amino acid sequence (SEQ ID NO:21) of the heavy chain variable region of the 16D10 human monoclonal antibody. The CDR1 (SEQ ID NO:3), CDR2 (SEQ ID NO:6) and CDR3 30 (SEQ ID NO:9) regions are delineated and the V and J germline derivations are indicated. 8 MXI-347PC Figure 3B shows the nucleotide sequence (SEQ ID NO:30) and amino acid sequence (SEQ ID NO:24) of the light chain variable region of the 16D10 human monoclonal antibody. The CDR1 (SEQ ID NO:12), CDR2 (SEQ ID NO:15) and CDR3 (SEQ ID NO: 18) regions are delineated and the V and J germline derivations are 5 indicated. Figure 4 shows the alignment of the amino acid sequence of the heavy chain variable region of 4A6 (SEQ ID NO:19) with the human germline VH 5-51 amino acid sequence (SEQ ID NO:3 1) and the human germline JH JH4b amino acid sequence (SEQ ID NO:32). 10 Figure 5 shows the alignment of the amino acid sequence of the heavy chain variable region of 1 1E7 (SEQ ID NO:20) with the human germline VH 5-51 amino acid sequences (SEQ ID NO:3 1) and the human germline JH JH4b amino acid sequence (SEQ ID NO:32). Figure 6 shows the alignment of the amino acid sequence of the heavy chain 15 variable region of 16D10 (SEQ ID NO:21) with the human germline VH 5-51 amino acid sequences (SEQ ID NO:3 1) and the human germline JH JH4b amino acid sequence (SEQ ID NO:32). Figure 7 shows the alignment of the amino acid sequence of the light chain variable region of 4A6 (SEQ ID NO:22) with the human germline Vk A27 amino acid 20 sequence (SEQ ID NO:33) and the human germline JK JK4 amino acid sequence (SEQ ID NO:34). Figure 8 shows the alignment of the amino acid sequence of the light chain variable region of 1 1E7 (SEQ ID NO:23) with the human germline Vk A27 amino acid sequence (SEQ ID NO:33) and the human germline JK JK4 amino acid sequence (SEQ 25 ID NO:34). Figure 9 shows the alignment of the amino acid sequence of the light chain variable region of 16D10 (SEQ ID NO:24) with the human germline Vk A27 amino acid sequence (SEQ ID NO:33) and the human germline JK JK1 amino acid sequence (SEQ ID NO:35). 30 Figure 10 shows the binding affinity of the 4A6, 1 1E7, and 16D10 monoclonal antibodies for the HCC cell line Hep-3B. Binding was measured using a Fluorescence Activated Cell Sorter ("FACS") assay. 9 MXI-347PC Figure 11 shows that monoclonal antibodies 4A6, 1 1E7, and 16D10 specifically bind to the HCC cell lines Hep-3B and Hep-G2. Binding was measured using a FACS assay. Figure 12 shows that monoclonal antibodies 4A6, 1 1E7, and 16D10 specifically 5 bind CHO cells stably transfected with a Glypican-3 expression vector but not to the parental CHO cell line. Binding was measured using a FACS assay. Figure 13 shows that monoclonal antibody 4A6 binds to liver cancer cells taken from patients. Antibody binding was revealed in an immunohistochemistry assay. Figure 14 shows that monoclonal antibodies 4A6, 1 1E7, and 16D10 can mediate 10 natural killer ("NK") T-cell-mediated killing of Hep-3B cells. Killing was measured with an antibody-dependent cellular cytotoxicity ("ADCC") assay. Figure 15 shows that monoclonal antibodies 4A6, 1 1E7, and 16D10 are internalized by Hep-3B cells following binding to the cell-surface Glypican 3. Internalization was measured by a Hum-ZAP assay (Advanced Targeting Systems, San 15 Diego, CA). Figure 16 shows that monoclonal antibodies 4A6, 1 1E7, and 16D10 are internalized by Hep-3B cells following binding to the cell-surface Glypican 3. Internalization was revealed in an immunofluorescence assay. DETAILED DESCRIPTION OF THE INVENTION 20 The present invention relates to isolated monoclonal antibodies, particularly human monoclonal antibodies that bind specifically to Glypican-3 with high affinity. In certain embodiments, the antibodies of the invention are derived from particular heavy and light chain germline sequences and/or comprise particular structural features such as CDR regions comprising particular amino acid sequences. The invention provides 25 isolated antibodies, methods of making such antibodies, immunoconjugates and bispecific molecules comprising such antibodies and pharmaceutical compositions containing the antibodies, immunoconjugates or bispecific molecules of the invention. The invention also relates to methods of using the antibodies, such as to detect Glypican 3, as well as to treat diseases associated with expression of Glypican-3, such as liver cell 30 malignancies that express Glypican-3. Accordingly, the invention also provides methods of using the anti-Glypican-3 antibodies of the invention to treat liver cell malignancies, for example, HCC. 10 MXI-347PC In order that the present invention may be more readily understood, certain terms are first defined. Additional definitions are set forth throughout the detailed description. The terms "Glypican-3, "glypican proteoglycan 3," "GPC3," "OTTHUMPO000062492," "GTR2-2," "SGB," "DGSX," "SDYS," "SGBS," "OCI-5," 5 and "SGBS 1" are used interchangeably, and include variants, isoforms and species homologs of human Glypican-3. Accordingly, human antibodies of this disclosure may, in certain cases, cross-react with Glypican-3 from species other than human. In certain embodiments, the antibodies may be completely specific for one or more human Glypican-3 proteins and may not exhibit species or other types of non-human cross 10 reactivity. The complete amino acid sequence of an exemplary human Glypican-3 has Genbank/NCBI accession number NM_004484 (SEQ ID NO:36). The term "immune response" refers to the action of, for example, lymphocytes, antigen presenting cells, phagocytic cells, granulocytes, and soluble macromolecules produced by the above cells or the liver (including antibodies, cytokines, and 15 complement) that results in selective damage to, destruction of, or elimination from the human body of invading pathogens, cells or tissues infected with pathogens, cancerous cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues. A "signal transduction pathway" refers to the biochemical relationship between 20 several of signal transduction molecules that play a role in the transmission of a signal from one portion of a cell to another portion of a cell. As used herein, the phrase "cell surface receptor" includes, for example, molecules and complexes of molecules capable of receiving a signal and the transmission of such a signal across the plasma membrane of a cell. An example of a "cell surface receptor" of the present invention is the 25 Glypican-3 receptor. The term "antibody" as referred to herein includes whole antibodies and any antigen binding fragment (i.e., "antigen-binding portion") or single chains thereof. An "antibody" refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion 30 thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CHi, CH 2 and CH 3 . Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The 11 MXI-347PC light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, 5 arranged from amino-terminus to carboxy-terminus in the following order: FRI, CDR1, FR2, CDR2, FR3, CDR3, and FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first 10 component (Clq) of the classical complement system. The term "antigen-binding portion" of an antibody (or simply "antibody portion"), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., Glypican-3). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length 15 antibody. Examples of binding fragments encompassed within the term "antigen binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fab' fragment, which is essentially an Fab with part of the hinge region (see, 20 FUNDAMENTAL IMMUNOLOGY (Paul ed., 3.sup.rd ed. 1993); (iv) a Fd fragment consisting of the VH and CHI domains; (v) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (vi) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; (vii) an isolated complementarity determining region (CDR); and (viii) a nanobody, a heavy chain variable region 25 containing a single variable domain and two constant domains. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 30 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen binding portion" of an antibody. These antibody fragments are obtained using 12 MXI-347PC conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. An "isolated antibody," as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an 5 isolated antibody that specifically binds Glypican-3 is substantially free of antibodies that specifically bind antigens other than Glypican-3). An isolated antibody that specifically binds Glypican-3 may, however, have cross-reactivity to other antigens, such as Glypican-3 molecules from other species. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals. 10 The terms "monoclonal antibody" or "monoclonal antibody composition" as used herein refer to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope. The term "human antibody," as used herein, is intended to include antibodies 15 having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. Furthermore, if the antibody contains a constant region, the constant region also is derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., 20 mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). However, the term "human antibody," as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. 25 The term "human monoclonal antibody" refers to antibodies displaying a single binding specificity which have variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. In one embodiment, the human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, 30 having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell. 13 MXI-347PC The term "recombinant human antibody," as used herein, includes all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom 5 (described further below), (b) antibodies isolated from a host cell transformed to express the human antibody, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial human antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human 10 antibodies have variable regions in which the framework and CDR regions are derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies 15 are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo. As used herein, "isotype" refers to the antibody class (e.g., IgM or IgGl) that is encoded by the heavy chain constant region genes. 20 The phrases "an antibody recognizing an antigen" and "an antibody specific for an antigen" are used interchangeably herein with the term "an antibody which binds specifically to an antigen." The term "human antibody derivatives" refers to any modified form of the human antibody, e.g., a conjugate of the antibody and another agent or antibody. 25 The term "humanized antibody" is intended to refer to antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications may be made within the human framework sequences. The term "chimeric antibody" is intended to refer to antibodies in which the 30 variable region sequences are derived from one species and the constant region sequences are derived from another species, such as an antibody in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody. 14 MXI-347PC As used herein, an antibody that "specifically binds to human Glypican-3" is intended to refer to an antibody that binds to human Glypican-3 with a KD Of 1 X 10 7 M or less, more preferably 5 x 10-8 M or less, more preferably 3 x 10-8 M or less, more preferably 5 x 10-8 M or less, even more preferably 5 x 10-9 M or less. 5 The term "does not substantially bind" to a protein or cells, as used herein, means does not bind or does not bind with a high affinity to the protein or cells, i.e. binds to the protein or cells with a KD Of 1 X 10-6 M or more, more preferably 1 X 10-5 M or more, more preferably 1 x 10-4 M or more, more preferably 1 x 10-3 M or more, even more preferably 1 x 10-2 M or more. 10 The term "Kassoc" or "Ka," as used herein, is intended to refer to the association rate of a particular antibody-antigen interaction, whereas the term "Kdis" or "Kd," as used herein, is intended to refer to the dissociation rate of a particular antibody-antigen interaction. The term "KD," as used herein, is intended to refer to the dissociation constant, which is obtained from the ratio of Kd to Ka (i.e,. Kd/Ka) and is expressed as a 15 molar concentration (M). KD values for antibodies can be determined using methods well established in the art. A preferred method for determining the KD of an antibody is by using surface plasmon resonance, preferably using a biosensor system such as a Biacore* system. As used herein, the term "high affinity" for an IgG antibody refers to an antibody 20 having a KD of 1 x 10~ M or less, more preferably 5 x 10-8 M or less, even more preferably 1x10- 8 M or less, even more preferably 5 x 10-9 M or less and even more preferably 1 x 10-9 M or less for a target antigen. However, "high affinity" binding can vary for other antibody isotypes. For example, "high affinity" binding for an IgM isotype refers to an antibody having a KD of 10-6 M or less, more preferably 10~ M or 25 less, even more preferably 10-8 M or less. As used herein, the term "subject" includes any human or nonhuman animal. The term "nonhuman animal" includes all vertebrates, e.g., mammals and non mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc. 30 Various aspects of the invention are described in further detail in the following subsections. 15 MXI-347PC Anti-Glypican-3 Antibodies The antibodies of the invention are characterized by particular functional features or properties of the antibodies. For example, the antibodies bind specifically to human Glypican-3. Preferably, an antibody of the invention binds to Glypican-3 with high 5 affinity, for example with a KD of 1 x 10- 7 M or less. The anti-Glypican-3 antibodies of the invention preferably exhibit one or more of the following characteristics: (a) binds to human Glypican-3 with a KD Of 1X10 7 M or less; (b) binds to human CHO cells transfected with Glypican-3. Preferably, the antibody binds to human Glypican-3 with a KD of 5 x 10-8 M or 10 less, binds to human Glypican-3 with a KD of 2 x 10-8 M or less, binds to human Glypican-3 with a KD of 5x10- 9 M or less, binds to human Glypican-3 with a KD Of 4x10- 9 M or less, binds to human Glypican-3 with a KD of 3x10~ 9 M or less, or binds to human Glypican-3 with a KD of 2.1 x 10 M or less. The antibody preferably binds to an antigenic epitope present in Glypican-3, 15 which epitope is not present in other proteins. The antibody typically binds to Glypican 3 but does not bind to other proteins, or binds to other proteins with a low affinity, such as with a KD Of I X 10-6 M or more, more preferably 1 X 10-5 M or more, more preferably 1 x 10 4M or more, more preferably 1 x 10-3 M or more, even more preferably 1 x 10-2 M or more. Preferably, the antibody does not substantially bind to related proteins, for 20 example, the antibody does not substantially bind to Glypican- 1, Glypican-2, Glypican-4 or Glypican-6. In one embodiment, the antibody may be internalized into a cell expressing Glypican-3. Standard assays to evaluate antibody internalization are known in the art, including, for example, a HumZap internalization assay. 25 Standard assays to evaluate the binding ability of the antibodies toward Glypican-3 are known in the art, including for example, ELISAs, Western blots, RIAs, and flow cytometry analysis. Suitable assays are described in detail in the Examples. The binding kinetics (e.g., binding affinity) of the antibodies also can be assessed by standard assays known in the art, such as by Biacore system analysis. To assess 30 binding to tumor cells, e.g. Hep-3b or Hep-G2 (ATCC Deposit No. HB-8064 and HB 8065, respectively), cells can be obtained from publicly available sources, such as the American Type Culture Collection, and used in standard assays, such as flow cytometric analysis. 16 MXI-347PC Monoclonal Antibodies 4A6, 1 E7, and 16D10 Preferred antibodies of the invention are the human monoclonal antibodies 4A6, 1 1E7, and 16D 10, isolated and structurally characterized as described in Examples 1 and 2. The VH amino acid sequences of 4A6, 1 1E7, and 16D10 are shown in SEQ ID 5 NOs:19, 20, and 21, respectively. The VL amino acid sequences of 4A6, 1 1E7, and 16D10 are shown in SEQ ID NOs:22, 23, and 24, respectively. Given that each of these antibodies can bind to Glypican-3, the VH and VL sequences can be "mixed and matched" to create other anti-Glypican-3 binding molecules of the invention. Glypican-3 binding of such "mixed and matched" 10 antibodies can be tested using the binding assays described above and in the Examples (e.g., ELISAs). Preferably, when VH and VL chains are mixed and matched, a VH sequence from a particular VH/VL pairing is replaced with a structurally similar VH sequence. Likewise, preferably a VL sequence from a particular VH/VL pairing is replaced with a structurally similar VL sequence. 15 Accordingly, in one aspect, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof comprising: (a) a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 19, 20, and 21; and 20 (b) a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 22, 23, and 24; wherein the antibody specifically binds Glypican-3, preferably human Glypican 3. 25 Preferred heavy and light chain combinations include: (a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 19; and (b) a light chain variable region comprising the amino acid 30 sequence of SEQ ID NO:22; or (b) a heavy chain variable region comprising the amino acid sequence of 17 MXI-347PC SEQ ID NO:20; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO:23. (c) a heavy chain variable region comprising the amino acid sequence 5 of SEQ ID NO:21; and (b) a light chain variable region comprising the amino acid sequence of SEQ ID NO:24. In another aspect, the invention provides antibodies that comprise the heavy 10 chain and light chain CDRls, CDR2s and CDR3s of 4A6, 1 1E7, and 16D10, or combinations thereof. The amino acid sequences of the VH CDRls of 4A6, 1 1E7, and 16D10 are shown in SEQ ID NOs:1, 2 and 3, respectively. The amino acid sequences of the VH CDR2s of 4A6, 1 1E7, and 16D10 are shown in SEQ ID NOs:4, 5, and 6, respectively. The amino acid sequences of the VH CDR3s of 4A6, 1 1E7, and 16D10 are 15 shown in SEQ ID NOs:7, 8, and 9, respectively. The amino acid sequences of the Vk CDRls of 4A6, 1 1E7, and 16D10 are shown in SEQ ID NOs:10, 11, and 12, respectively. The amino acid sequences of the Vk CDR2s of 4A6, 1 1E7, and 16D10 are shown in SEQ ID NOs:13, 14, and 15, respectively. The amino acid sequences of the Vk CDR3s of 4A6, 1 1E7, and 16D10 are shown in SEQ ID NOs:16, 17, and 18, 20 respectively. The CDR regions are delineated using the Kabat system (Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). Given that each of these antibodies can bind to Glypican-3 and that antigen binding specificity is provided primarily by the CDR1, CDR2, and CDR3 regions, the 25 VH CDR1, CDR2, and CDR3 sequences and Vk CDR1, CDR2, and CDR3 sequences can be "mixed and matched" (i.e., CDRs from different antibodies can be mixed and match, although each antibody must contain a VH CDR1, CDR2, and CDR3 and a Vk CDR1, CDR2, and CDR3) to create other anti-Glypican-3 binding molecules of the invention. Glypican-3 binding of such "mixed and matched" antibodies can be tested 30 using the binding assays described above and in the Examples (e.g., ELISAs, Biacore" analysis). Preferably, when VH CDR sequences are mixed and matched, the CDR1, CDR2 and/or CDR3 sequence from a particular VH sequence is replaced with a structurally similar CDR sequence(s). Likewise, when Vk CDR sequences are mixed 18 MXI-347PC and matched, the CDR1, CDR2 and/or CDR3 sequence from a particular Vk sequence preferably is replaced with a structurally similar CDR sequence(s). It will be readily apparent to the ordinarily skilled artisan that novel VH and VL sequences can be created by substituting one or more VH and/or VL CDR region sequences with structurally 5 similar sequences from the CDR sequences disclosed herein for monoclonal antibodies 4A6, 11E7, and 16D10. Accordingly, in another aspect, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof comprising: (a) a heavy chain variable region CDR1 comprising an amino acid sequence 10 selected from the group consisting of SEQ ID NOs:1, 2 and 3; (b) a heavy chain variable region CDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:4, 5, and 6; (c) a heavy chain variable region CDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:7, 8, and 9; 15 (d) a light chain variable region CDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:10, 11, and 12; (e) a light chain variable region CDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:13, 14, and 15; and (f) a light chain variable region CDR3 comprising an amino acid sequence 20 (g) selected from the group consisting of SEQ ID NOs: 16, 17, and 18; wherein the antibody specifically binds Glypican-3, preferably human Glypican 3. In a preferred embodiment, the antibody comprises: (a) a heavy chain variable region CDR1 comprising SEQ ID NO: 1; 25 (b) a heavy chain variable region CDR2 comprising SEQ ID NO:4; (c) a heavy chain variable region CDR3 comprising SEQ ID NO:7; (d) a light chain variable region CDR1 comprising SEQ ID NO:10; (e) a light chain variable region CDR2 comprising SEQ ID NO:13; and (f) a light chain variable region CDR3 comprising SEQ ID NO:16. 30 In another preferred embodiment, the antibody comprises: (a) a heavy chain variable region CDR1 comprising SEQ ID NO:2; (b) a heavy chain variable region CDR2 comprising SEQ ID NO:5; (c) a heavy chain variable region CDR3 comprising SEQ ID NO:8; 19 MXI-347PC (d) a light chain variable region CDR1 comprising SEQ ID NO: 11; (e) a light chain variable region CDR2 comprising SEQ ID NO:14; and (f) a light chain variable region CDR3 comprising SEQ ID NO:17. In another preferred embodiment, the antibody comprises: 5 (a) a heavy chain variable region CDR1 comprising SEQ ID NO:3; (b) a heavy chain variable region CDR2 comprising SEQ ID NO:6; (c) a heavy chain variable region CDR3 comprising SEQ ID NO:9; (d) a light chain variable region CDR1 comprising SEQ ID NO:12; (e) a light chain variable region CDR2 comprising SEQ ID NO:15; and 10 (f) a light chain variable region CDR3 comprising SEQ ID NO:18. It is well known in the art that the CDR3 domain, independently from the CDR1 and/or CDR2 domain(s), alone can determine the binding specificity of an antibody for a cognate antigen and that multiple antibodies can predictably be generated having the same binding specificity based on a common CDR3 sequence. See, for example, 15 Klimka et al., British J. of Cancer 83(2):252-260 (2000) (describing the production of a humanized anti-CD30 antibody using only the heavy chain variable domain CDR3 of murine anti-CD30 antibody Ki-4); Beiboer et al., J. Mol. Biol. 296:833-849 (2000) (describing recombinant epithelial glycoprotein-2 (EGP-2) antibodies using only the heavy chain CDR3 sequence of the parental murine MOC-31 anti-EGP-2 antibody); 20 Rader et al., Proc. Natl. Acad. Sci. US.A. 95:8910-8915 (1998) (describing a panel of humanized anti-integrin avp 3 antibodies using a heavy and light chain variable CDR3 domain of a murine anti-integrin ay3 antibody LM609 wherein each member antibody comprises a distinct sequence outside the CDR3 domain and capable of binding the same epitope as the parent murine antibody with affinities as high or higher than the 25 parent murine antibody); Barbas et al., J. Am. Chem. Soc. 116:2161-2162 (1994) (disclosing that the CDR3 domain provides the most significant contribution to antigen binding); Barbas et al., Proc. Natl. Acad. Sci. US.A. 92:2529-2533 (1995) (describing the grafting of heavy chain CDR3 sequences of three Fabs (SI-1, SI-40, and SI-32) against human placental DNA onto the heavy chain of an anti-tetanus toxoid Fab thereby 30 replacing the existing heavy chain CDR3 and demonstrating that the CDR3 domain alone conferred binding specificity); and Ditzel et al., J Immunol. 157:739-749 (1996) (describing grafting studies wherein transfer of only the heavy chain CDR3 of a parent polyspecific Fab LNA3 to a heavy chain of a monospecific IgG tetanus toxoid-binding 20 MXI-347PC Fab p313 antibody was sufficient to retain binding specificity of the parent Fab). Each of these references is hereby incorporated by reference in its entirety. Accordingly, the present invention provides monoclonal antibodies comprising one or more heavy and/or light chain CDR3 domains from an antibody derived from a 5 human or non-human animal, wherein the monoclonal antibody is capable of specifically binding to Glypican-3. Within certain aspects, the present invention provides monoclonal antibodies comprising one or more heavy and/or light chain CDR3 domain from a non-human antibody, such as a mouse or rat antibody, wherein the monoclonal antibody is capable of specifically binding to Glypican-3. Within some 10 embodiments, such inventive antibodies comprising one or more heavy and/or light chain CDR3 domain from a non-human antibody (a) are capable of competing for binding with; (b) retain the functional characteristics; (c) bind to the same epitope; and/or (d) have a similar binding affinity as the corresponding parental non-human antibody. 15 Within other aspects, the present invention provides monoclonal antibodies comprising one or more heavy and/or light chain CDR3 domain from a human antibody, such as, for example, a human antibody obtained from a non-human animal, wherein the human antibody is capable of specifically binding to Glypican-3. Within other aspects, the present invention provides monoclonal antibodies comprising one or more heavy 20 and/or light chain CDR3 domain from a first human antibody, such as, for example, a human antibody obtained from a non-human animal, wherein the first human antibody is capable of specifically binding to Glypican-3 and wherein the CDR3 domain from the first human antibody replaces a CDR3 domain in a human antibody that is lacking binding specificity for Glypican-3 to generate a second human antibody that is capable 25 of specifically binding to Glypican-3. Within some embodiments, such inventive antibodies comprising one or more heavy and/or light chain CDR3 domains from the first human antibody (a) are capable of competing for binding with; (b) retain the functional characteristics; (c) bind to the same epitope; and/or (d) have a similar binding affinity as the corresponding parental first human antibody. In preferred embodiments, 30 the first human antibody is 4A6, 1 1E7, and 16D 10. 21 MXI-347PC Antibodies Having Particular Germline Sequences In certain embodiments, an antibody of the invention comprises a heavy chain variable region from a particular germline heavy chain immunoglobulin gene and/or a light chain variable region from a particular germline light chain immunoglobulin gene. 5 For example, in a preferred embodiment, the invention provides an isolated monoclonal antibody, or an antigen-binding portion thereof, comprising a heavy chain variable region that is the product of or derived from a human VH 5-51 gene, wherein the antibody specifically binds Glypican-3. In another preferred embodiment, the invention provides an isolated monoclonal antibody, or an antigen-binding portion thereof, 10 comprising a light chain variable region that is the product of or derived from a human VK A27 gene, wherein the antibody specifically binds Glypican-3. In yet another preferred embodiment, the invention provides an isolated monoclonal antibody, or antigen-binding portion thereof, wherein the antibody: (a) comprises a heavy chain variable region that is the product of or derived 15 from a human VH 5-51 gene (which gene encodes the amino acid sequences set forth in SEQ ID NO:3 1); (b) comprises a light chain variable region that is the product of or derived 20 from a human VK A27 gene (which gene encodes the amino acid sequences set forth in SEQ ID NO:33); and (c) specifically binds to Glypican-3, preferably human Glypican-3. 25 Examples of antibodies having VH 5-51 and VK A27 are 4A6, 1 E7, and 16D10 As used herein, a human antibody comprises heavy or light chain variable regions that is "the product of' or "derived from" a particular germline sequence if the variable regions of the antibody are obtained from a system that uses human germline immunoglobulin genes. Such systems include immunizing a transgenic mouse carrying 30 human immunoglobulin genes with the antigen of interest or screening a human immunoglobulin gene library displayed on phage with the antigen of interest. A human antibody that is "the product of' or "derived from" a human germline immunoglobulin sequence can be identified as such by comparing the amino acid sequence of the human 22 MXI-347PC antibody to the amino acid sequences of human germline immunoglobulins and selecting the human germline immunoglobulin sequence that is closest in sequence (i.e., greatest % identity) to the sequence of the human antibody. A human antibody that is "the product of" or "derived from" a particular human germline immunoglobulin sequence 5 may contain amino acid differences as compared to the germline sequence, due to, for example, naturally-occurring somatic mutations or intentional introduction of site directed mutation. However, a selected human antibody typically is at least 90% identical in amino acids sequence to an amino acid sequence encoded by a human germline immunoglobulin gene and contains amino acid residues that identify the human 10 antibody as being human when compared to the germline immunoglobulin amino acid sequences of other species (e.g., murine germline sequences). In certain cases, a human antibody may be at least 95%, or even at least 96%, 97%, 98%, or 99% identical in amino acid sequence to the amino acid sequence encoded by the germline immunoglobulin gene. Typically, a human antibody derived from a particular human 15 germline sequence will display no more than 10 amino acid differences from the amino acid sequence encoded by the human germline immunoglobulin gene. In certain cases, the human antibody may display no more than 5, or even no more than 4, 3, 2, or 1 amino acid difference from the amino acid sequence encoded by the germline immunoglobulin gene. 20 Homologous Antibodies In yet another embodiment, an antibody of the invention comprises heavy and light chain variable regions comprising amino acid sequences that are homologous to the amino acid sequences of the preferred antibodies described herein, and wherein the antibodies retain the desired functional properties of the anti-Glypican-3 antibodies of 25 the invention. For example, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising a heavy chain variable region and a light chain variable region, wherein: (a) the heavy chain variable region comprises an amino acid sequence that is 30 at least 80% homologous to an amino acid sequence selected from the group consisting of SEQ ID NOs:19, 20 and 21; 23 MXI-347PC (b) the light chain variable region comprises an amino acid sequence that is at least 80% homologous to an amino acid sequence selected from the group consisting 5 of SEQ ID NOs:22, 23, and 24; and (c) the antibody binds to human Glypican-3 with a KD Of 1X10 7 M or less. The antibody may also bind to CHO cells transfected with human Glypican-3. In various embodiments, the antibody can be, for example, a human antibody, a humanized antibody or a chimeric antibody. 10 In other embodiments, the VH and/or VL amino acid sequences may be 85%, 90%, 95%, 96%, 97%, 98% or 99% homologous to the sequences set forth above. An antibody having VH and VL regions having high (i.e., 80% or greater) homology to the VH and VL regions of the sequences set forth above, can be obtained by mutagenesis (e.g., site-directed or PCR-mediated mutagenesis) of nucleic acid molecules encoding 15 SEQ ID NOs:25, 26, 27, 28, 29, and 30, followed by testing of the encoded altered antibody for the ability to bind glypican-3 using the functional assays described herein. As used herein, the percent homology between two amino acid sequences is equivalent to the percent identity between the two sequences. The percent identity between the two sequences is a function of the number of identical positions shared by 20 the sequences (i.e., % homology = # of identical positions/total # of positions x 100), taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below. 25 The percent identity between two amino acid sequences can be determined using the algorithm of E. Meyers and W. Miller (Comput. Apple. Biosci., 4:11-17 (1988)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. In addition, the percent identity between two amino acid sequences can be determined using the Needleman and 30 Wunsch (J Mol. Biol. 48:444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.geg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6. 24 MXI-347PC Additionally or alternatively, the protein sequences of the present invention can further be used as a "query sequence" to perform a search against public databases to, for example, identify related sequences. Such searches can be performed using the XBLAST program (version 2.0) of Altschul, et al. (1990) J Mol. Biol. 215:403-10. 5 BLAST protein searches can be performed with the XBLAST program, score = 50, wordlength = 3 to obtain amino acid sequences homologous to the antibody molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25(17):3389-3402. When utilizing BLAST and Gapped BLAST programs, the default 10 parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See http://www.ncbi.nlm.nih.gov. Antibodies with Conservative Modifications In certain embodiments, an antibody of the invention comprises a heavy chain variable region comprising CDR1, CDR2 and CDR3 sequences and a light chain 15 variable region comprising CDR1, CDR2 and CDR3 sequences, wherein one or more of these CDR sequences comprise specified amino acid sequences based on the preferred antibodies described herein (e.g., 4A6, 1 1E7, and 16D 10), or conservative modifications thereof, and wherein the antibodies retain the desired functional properties of the anti Glypican-3 antibodies of the invention. Accordingly, the invention provides an isolated 20 monoclonal antibody, or antigen binding portion thereof, comprising a heavy chain variable region comprising CDR1, CDR2, and CDR3 sequences and a light chain variable region comprising CDR1, CDR2, and CDR3 sequences, wherein: (a) the heavy chain variable region CDR3 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID 25 NOs:7, 8, and 9, and conservative modifications thereof; (b) the light chain variable region CDR3 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequence of SEQ ID NOs:16, 17, and 18, and conservative modifications thereof; and (c) the antibody binds to human Glypican-3 with a KD Of 1X10 7 M or less. 30 The antibody may also bind to CHO cells transfected with human Glypican-3. In a preferred embodiment, the heavy chain variable region CDR2 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs:4, 5, and 6, and conservative modifications thereof; and the 25 MXI-347PC light chain variable region CDR2 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs:13, 14, and 15, and conservative modifications thereof. In another preferred embodiment, the heavy chain variable region CDR1 sequence comprises an amino acid sequence selected from the 5 group consisting of amino acid sequences of SEQ ID NOs:1, 2, and 3 and conservative modifications thereof; and the light chain variable region CDR1 sequence comprises an amino acid sequence selected from the group consisting of amino acid sequences of SEQ ID NOs:10, 11, and 12, and conservative modifications thereof. In various embodiments, the antibody can be, for example, human antibodies, 10 humanized antibodies or chimeric antibodies. As used herein, the term "conservative sequence modifications" is intended to refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications 15 can be introduced into an antibody of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids 20 with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, 25 phenylalanine, tryptophan, histidine). Thus, one or more amino acid residues within the CDR regions of an antibody of the invention can be replaced with other amino acid residues from the same side chain family and the altered antibody can be tested for the ability to bind glypican-3 using the functional assays described herein. The heavy chain CDR1 sequence of SEQ ID NO:1, 2, or 3 may comprise one or 30 more conservative sequence modification, such as one, two, three, four, five or more amino acid substitutions, additions or deletions; the light chain CDR1 sequence of SEQ ID NO:10, 11, or 12 may comprise one or more conservative sequence modification, such as one, two, three, four, five or more amino acid substitutions, additions or 26 MXI-347PC deletions; the heavy chain CDR2 sequence shown in SEQ ID NO:4, 5, or 6 may comprise one or more conservative sequence modification, such as one, two, three, four, five or more amino acid substitutions, additions or deletions; the light chain CDR2 sequence shown in SEQ ID NO:13, 14, or 15 may comprise one or more conservative 5 sequence modification, such as one, two, three, four, five or more amino acid substitutions, additions or deletions; the heavy chain CDR3 sequence shown in SEQ ID NO:7, 8, or 9 may comprise one or more conservative sequence modification, such as one, two, three, four, five or more amino acid substitutions, additions or deletions; and/or the light chain CDR3 sequence shown in SEQ ID NO:16, 17, or 18 may comprise 10 one or more conservative sequence modification, such as one, two, three, four, five or more amino acid substitutions, additions or deletions. Antibodies that Bind to the Same Epitope as Anti-Glypican-3 Antibodies of the Invention In another embodiment, the invention provides antibodies that bind to the same epitope on human Glypican-3 as any of the Glypican-3 monoclonal antibodies of the 15 invention (i. e., antibodies that have the ability to cross-compete for binding to Glypican 3 with any of the monoclonal antibodies of the invention). In preferred embodiments, the reference antibody for cross-competition studies can be the monoclonal antibody 4A6 (having VH and VL sequences as shown in SEQ ID NOs: 19 and 22, respectively), the monoclonal antibody 1 1E7 (having VH and VL sequences as shown in SEQ ID 20 NOs:20 and 23, respectively), or the monoclonal antibody 16D10 (having VH and VL sequences as shown in SEQ ID NOs:21 and 24, respectively). Such cross-competing antibodies can be identified based on their ability to cross-compete with 4A6, 1 1E7, or 16D10 in standard Glypican-3 binding assays. For example, Biacore analysis, ELISA assays or flow cytometry may be used to demonstrate cross-competition with the 25 antibodies of the current invention. The ability of a test antibody to inhibit the binding of, for example, 4A6, 1 1E7, or 16D 10, to human Glypican-3 demonstrates that the test antibody can compete with 4A6, 1 1E7, or 16D10 for binding to human Glypican-3 and thus binds to the same epitope on human Glypican-3 as 4A6, 1 1E7, or 16D 10. In a preferred embodiment, the antibody that binds to the same epitope on human Glypican-3 30 as 4A6, 1 1E7, or 16D10 is a human monoclonal antibody. Such human monoclonal antibodies can be prepared and isolated as described in the Examples. 27 MXI-347PC Engineered and Modified Antibodies An antibody of the invention further can be prepared using an antibody having one or more of the VH and/or VL sequences disclosed herein can be used as starting material to engineer a modified antibody, which modified antibody may have altered 5 properties as compared to the starting antibody. An antibody can be engineered by modifying one or more amino acids within one or both variable regions (i.e., VH and/or VL), for example within one or more CDR regions and/or within one or more framework regions. Additionally or alternatively, an antibody can be engineered by modifying residues within the constant region(s), for example to alter the effector function(s) of the 10 antibody. In certain embodiments, CDR grafting can be used to engineer variable regions of antibodies. Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs 15 are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of specific naturally occurring antibodies by constructing expression vectors that include CDR sequences from the specific naturally occurring antibody grafted onto framework 20 sequences from a different antibody with different properties (see, e.g., Riechmann, L. et al. (1998) Nature 332:323-327; Jones, P. et al. (1986) Nature 321:522-525; Queen, C. et al. (1989) Proc. Natl. Acad. See. US.A. 86:10029-10033; U.S. Patent No. 5,225,539 to Winter, and U.S. Patent Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen et al.) 25 Accordingly, another embodiment of the invention pertains to an isolated monoclonal antibody, or antigen binding portion thereof, comprising a heavy chain variable region comprising CDR1, CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:1, 2, and 3, SEQ ID NOs:4, 5, and 6, and SEQ ID NOs:7, 8, and 9, respectively, and a light chain variable 30 region comprising CDR1, CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 10, 11, and 12, SEQ ID NOs:13, 14, and 15, and SEQ ID NOs:16, 17, and 18, respectively. Thus, such 28 MXI-347PC antibodies contain the VH and VL CDR sequences of monoclonal antibodies 4A6, 1 1E7, or 16D 10 yet may contain different framework sequences from these antibodies. Such framework sequences can be obtained from public DNA databases or published references that include germline antibody gene sequences. For example, 5 germline DNA sequences for human heavy and light chain variable region genes can be found in the "VBase" human germline sequence database (available on the Internet at www.mrc-cpe.cam.ac.uk/vbase), as well as in Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; Tomlinson, I. M., et al. (1992) "The 10 Repertoire of Human Germline VH Sequences Reveals about Fifty Groups of VH Segments with Different Hypervariable Loops" J Mol. Biol. 227:776-798; and Cox, J. P. L. et al. (1994) "A Directory of Human Germ-line VH Segments Reveals a Strong Bias in their Usage" Eur. J. Immunol. 24:827-836; the contents of each of which are expressly incorporated herein by reference. As another example, the germline DNA 15 sequences for human heavy and light chain variable region genes can be found in the Genbank database. For example, the following heavy chain germline sequences found in the HCo7 HuMAb mouse are available in the accompanying Genbank accession numbers: 1-69 (NG_0010109, NT_024637 and BC070333), 3-33 (NG_0010109 and NT_024637) and 3-7 (NG_0010109 and NT_024637). As another example, the 20 following heavy chain germline sequences found in the HCo12 HuMAb mouse are available in the accompanying Genbank accession numbers: 1-69 (NG_0010109, NT_024637 and BC070333), 5-51 (NG_0010109 and NT_024637), 4-34 (NG_0010109 and NT_024637), 3-30.3 (CAJ556644) and 3-23 (AJ406678). Antibody protein sequences are compared against a compiled protein sequence 25 database using one of the sequence similarity searching methods called the Gapped BLAST (Altschul et al. (1997) Nucleic Acids Research 25:3389-3402), which is well known to those skilled in the art. BLAST is a heuristic algorithm in that a statistically significant alignment between the antibody sequence and the database sequence is likely to contain high-scoring segment pairs (HSP) of aligned words. Segment pairs whose 30 scores cannot be improved by extension or trimming is called a hit. Briefly, the nucleotide sequences of VBASE origin (http://vbase.mrc cpe. cam. ac.uk/vbasel1list2.php) are translated and the region between and including FRI through FR3 framework region is retained. The database sequences have an average 29 MXI-347PC length of 98 residues. Duplicate sequences which are exact matches over the entire length of the protein are removed. A BLAST search for proteins using the program blastp with default, standard parameters except the low complexity filter, which is turned off, and the substitution matrix of BLOSUM62, filters for top 5 hits yielding 5 sequence matches. The nucleotide sequences are translated in all six frames and the frame with no stop codons in the matching segment of the database sequence is considered the potential hit. This is in turn confirmed using the BLAST program tblastx, which translates the antibody sequence in all six frames and compares those translations to the VBASE nucleotide sequences dynamically translated in all six frames. 10 The identities are exact amino acid matches between the antibody sequence and the protein database over the entire length of the sequence. The positives (identities + substitution match) are not identical but amino acid substitutions guided by the BLOSUM62 substitution matrix. If the antibody sequence matches two of the database sequences with same identity, the hit with most positives would be decided to be the 15 matching sequence hit. Preferred framework sequences for use in the antibodies of the invention are those that are structurally similar to the framework sequences used by selected antibodies of the invention, e.g., similar to the VH 5-51 framework sequences (SEQ ID NO:3 1) and/or the VK A27 framework sequences (SEQ ID NO:33) used by preferred 20 monoclonal antibodies of the invention. The VH CDR1, CDR2, and CDR3 sequences, and the VK CDR1, CDR2, and CDR3 sequences, can be grafted onto framework regions that have the identical sequence as that found in the germline immunoglobulin gene from which the framework sequence derive, or the CDR sequences can be grafted onto framework regions that contain one or more mutations as compared to the germline 25 sequences. For example, it has been found that in certain instances it is beneficial to mutate residues within the framework regions to maintain or enhance the antigen binding ability of the antibody (see e.g., U.S. Patent Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen et al.). Another type of variable region modification is to mutate amino acid residues 30 within the VH and/or VK CDR1, CDR2 and/or CDR3 regions to thereby improve one or more binding properties (e.g., affinity) of the antibody of interest. Site-directed mutagenesis or PCR-mediated mutagenesis can be performed to introduce the mutation(s) and the effect on antibody binding, or other functional property of interest, 30 MXI-347PC can be evaluated in in vitro or in vivo assays as described herein and provided in the Examples. Preferably conservative modifications (as discussed above) are introduced. The mutations may be amino acid substitutions, additions or deletions, but are preferably substitutions. Moreover, typically no more than one, two, three, four or five residues 5 within a CDR region are altered. Accordingly, in another embodiment, the instant disclosure provides isolated anti-Glypican-3 monoclonal antibodies, or antigen binding portions thereof, comprising a heavy chain variable region comprising: (a) a VH CDR1 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:1, 2, and 3, or an 10 amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs:1, 2 and 3; (b) a VH CDR2 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:4, 5, and 6, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs:4, 5, and 6; (c) a 15 VH CDR3 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:7, 8, and 9, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs:7, 8, and 9; (d) a VK CDR1 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 10, 11, and 12, or an amino acid sequence having one, 20 two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 10, 11, and 12; (e) a VK CDR2 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 13, 14, and 15, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 13, 14, and 15; and (f) a VK CDR3 region 25 comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 16, 17, and 18, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 16, 17, and 18. Engineered antibodies of the invention include those in which modifications have been made to framework residues within VH and/or VK, e.g. to improve the 30 properties of the antibody. Typically such framework modifications are made to decrease the immunogenicity of the antibody. For example, one approach is to "backmutate" one or more framework residues to the corresponding germline sequence. More specifically, an antibody that has undergone somatic mutation may contain 31 MXI-347PC framework residues that differ from the germline sequence from which the antibody is derived. Such residues can be identified by comparing the antibody framework sequences to the germline sequences from which the antibody is derived. For example, for 4A6, using the Kabat numbering system, amino acid residue #73 (within FR3) of VH 5 is an arginine (SEQ ID NO: 19) whereas this residue in the corresponding VH 5-51 germline sequence is lysine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, the somatic mutations can be "backmutated" to the germline sequence by, for example, site-directed mutagenesis or PCR-mediated mutagenesis (e.g., residue #73 (residue #8 of FR3) of the VH of 4A6 can be 10 "backmutated" from arginine to lysine). As another example, for 4A6, amino acid residue #76 (within FR3) of VH is arginine (SEQ ID NO: 19) whereas this residue in the corresponding VH 5-51 germline sequence is a serine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #76 (residue #11 of FR3) of the VH Of 15 4A6 can be "backmutated" from arginine to serine. Such "backmutated" antibodies are also intended to be encompassed by the invention. As another example, for 4A6, amino acid residue #89 (within FR3) of VH is leucine (SEQ ID NO:19) whereas this residue in the corresponding VH 5-51 germline sequence is a methionine (SEQ ID NO:3 1). To return the framework region sequences 20 to their germline configuration, for example, residue #89 (residue #27 of FR3) of the VH of 4A6 can be "backmutated" from leucine to methionine. Such "backmutated" antibodies are also intended to be encompassed by the invention. As another example, for 1 1E7, amino acid residue #76 (within FR3) of VH is arginine (SEQ ID NO:20) whereas this residue in the corresponding VH 5-51 germline 25 sequence is a serine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #76 (residue #11 of FR3) of the VH Of 1 1E7 can be "backmutated" from arginine to serine. Such "backmutated" antibodies are also intended to be encompassed by the invention. As another example, for 16D 10, amino acid residue #10 (within FRI) of VH is 30 aspartate (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline sequence is a glutamate (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #10 (residue #10 of FRI) of the VH Of 32 MXI-347PC 16D10 can be "backmutated" from aspartate to glutamate. Such "backmutated" antibodies are also intended to be encompassed by the invention. As another example, for 16D10, amino acid residue #12 (within FRI) of VH is threonine (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline 5 sequence is a lysine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #12 (residue #12 of FRI) of the VH Of 16D10 can be "backmutated" from threonine to lysine. Such "backmutated" antibodies are also intended to be encompassed by the invention. As another example, for 16D 10, amino acid residue #24 (within FRI) of VH is 10 valine (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline sequence is a glycine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #24 (residue #24 of FRI) of the VH Of 16D10 can be "backmutated" from valine to glycine. Such "backmutated" antibodies are also intended to be encompassed by the invention. 15 As another example, for 16D10, amino acid residue #28 (within FRI) of VH is arginine (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline sequence is a serine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #28 (residue #28 of FRI) of the VH Of 16D 10 can be "backmutated" from arginine to serine. Such "backmutated" antibodies 20 are also intended to be encompassed by the invention. As another example, for 16D 10, amino acid residue #37 (within FR2) of VH is methionine (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline sequence is a valine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #37 (residue #2 of FR2) 25 of the VH of 16D10 can be "backmutated" from methionine to valine. Such "backmutated" antibodies are also intended to be encompassed by the invention. As another example, for 16D 10, amino acid residue #41 (within FR2) of VH is serine (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline sequence is a proline (SEQ ID NO:3 1). To return the framework region sequences to 30 their germline configuration, for example, residue #41 (residue #6 of FR2) of the VH Of 16D10 can be "backmutated" from serine to proline. Such "backmutated" antibodies are also intended to be encompassed by the invention. 33 MXI-347PC As another example, for 16D 10, amino acid residue #66 (within FR3) of VH is histidine (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline sequence is a glutamine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #66 (residue #1 of FR3) of the VH Of 5 16D10 can be "backmutated" from histidine to glutamine. Such "backmutated" antibodies are also intended to be encompassed by the invention. As another example, for 16D 10, amino acid residue #76 (within FR3) of VH is asparagine (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline sequence is a serine (SEQ ID NO:3 1). To return the framework region sequences to 10 their germline configuration, for example, residue #76 (residue #11 of FR3) of the VH Of 16D 10 can be "backmutated" from asparagine to serine. Such "backmutated" antibodies are also intended to be encompassed by the invention. As another example, for 16D 10, amino acid residue #81 (within FR3) of VH is arginine (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline 15 sequence is a glutamine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #81 (residue #16 of FR3) of the VH Of 16D10 can be "backmutated" from arginine to glutamine. Such "backmutated" antibodies are also intended to be encompassed by the invention. As another example, for 16D 10, amino acid residue #89 (within FR3) of VH is 20 isoleucine (SEQ ID NO:21) whereas this residue in the corresponding VH 5-51 germline sequence is a methionine (SEQ ID NO:3 1). To return the framework region sequences to their germline configuration, for example, residue #89 (residue #27 of FR3) of the VH of 16D10 can be "backmutated" from isoleucine to methionine. Such "backmutated" antibodies are also intended to be encompassed by the invention. 25 Similarly, the invention provides for the backmutation of framework residues in the light chain. For example, for 16D 10, amino acid residue #3 (within FRI) of VK is leucine (SEQ ID NO:24) whereas this residue in the corresponding VK A27 germline sequence is a valine (SEQ ID NO:33). To return the framework region sequences to their germline configuration, for example, residue #3 (residue #3 of FRI) of the VK of 30 16D10 can be "backmutated" from leucine to valine. Another type of framework modification involves mutating one or more residues within the framework region, or even within one or more CDR regions, to remove T cell epitopes to thereby reduce the potential immunogenicity of the antibody. This approach 34 MXI-347PC is also referred to as "deimmunization" and is described in further detail in U.S. Patent Publication No. 20030153043 by Carr et al. In addition or alternative to modifications made within the framework or CDR regions, antibodies of the invention may be engineered to include modifications within 5 the Fe region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fe receptor binding, and/or antigen-dependent cellular cytotoxicity. Furthermore, an antibody of the invention may be chemically modified (e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation, again to alter one or more functional properties of the 10 antibody. Each of these embodiments is described in further detail below. The numbering of residues in the Fe region is that of the EU index of Kabat. In one embodiment, the hinge region of CHI is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further in U.S. Patent No. 5,677,425 by Bodmer et al. The 15 number of cysteine residues in the hinge region of CH1 is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody. In another embodiment, the Fe hinge region of an antibody is mutated to decrease the biological half life of the antibody. More specifically, one or more amino 20 acid mutations are introduced into the CH2-CH3 domain interface region of the Fe hinge fragment such that the antibody has impaired Staphylococcyl protein A (SpA) binding relative to native Fc-hinge domain SpA binding. This approach is described in further detail in U.S. Patent No. 6,165,745 by Ward et al. In another embodiment, the antibody is modified to increase its biological half 25 life. Various approaches are possible. For example, one or more of the following mutations can be introduced: T252L, T254S, and T256F, as described in U.S. Patent No. 6,277,375 to Ward. Alternatively, to increase the biological half life, the antibody can be altered within the CHI or CL region to contain a salvage receptor binding epitope taken from two loops of a CH2 domain of an Fe region of an IgG, as described in U.S. 30 Patent Nos. 5,869,046 and 6,121,022 by Presta et al. In yet other embodiments, the Fe region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector function(s) of the antibody. For example, one or more amino acids selected from amino acid residues 234, 35 MXI-347PC 235, 236, 237, 297, 318, 320 and 322 can be replaced with a different amino acid residue such that the antibody has an altered affinity for an effector ligand but retains the antigen-binding ability of the parent antibody. The effector ligand to which affinity is altered can be, for example, an Fe receptor or the C1 component of complement. This 5 approach is described in further detail in U.S. Patent Nos. 5,624,821 and 5,648,260, both by Winter et al. In another example, one or more amino acids selected from amino acid residues 329, 331 and 322 can be replaced with a different amino acid residue such that the antibody has altered Cl q binding and/or reduced or abolished complement dependent 10 cytotoxicity (CDC). This approach is described in further detail in U.S. Patent Nos. 6,194,551 by Idusogie et al. In another example, one or more amino acid residues within amino acid positions 231 and 239 are altered to thereby alter the ability of the antibody to fix complement. This approach is described further in PCT Publication WO 94/29351 by Bodmer et al. 15 In yet another example, the Fe region is modified to increase the ability of the antibody to mediate antibody dependent cellular cytotoxicity (ADCC) and/or to increase the affinity of the antibody for an Fey receptor by modifying one or more amino acids at the following positions: 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269,270,272,276,278,280,283,285,286,289,290,292,293,294,295,296,298, 20 301,303,305,307,309,312,315,320,322,324,326,327,329,330,331,333,334, 335,337,338,340,360,373,376,378,382,388,389,398,414,416,419,430,434, 435, 437, 438 or 439. This approach is described further in PCT Publication WO 00/42072 by Presta. Moreover, the binding sites on human IgGI for FcyR1, FcyRII, FcyRIII and FcRn have been mapped and variants with improved binding have been 25 described (see Shields, R.L. et al. (2001) J. Biol. Chem. 276:6591-6604). Specific mutations at positions 256, 290, 298, 333, 334 and 339 were shown to improve binding to FcyRIII. Additionally, the following combination mutants were shown to improve FcyRIII binding: T256A/S298A, S298A/E333A, S298A/K224A and S298A/E333A/K334A. 30 In still another embodiment, the glycosylation of an antibody is modified. For example, an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation). Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen. Such carbohydrate modifications can be accomplished by, for example, altering 36 MXI-347PC one or more sites of glycosylation within the antibody sequence. For example, one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site. Such aglycosylation may increase the affinity of the antibody for antigen. Such an 5 approach is described in further detail in U.S. Patent Nos. 5,714,350 and 6,350,861 by Co et al. Additionally or alternatively, an antibody can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNac structures. Such altered 10 glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies. Such carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an 15 antibody with altered glycosylation. For example, the cell lines Ms704, Ms705, and Ms709 lack the fucosyltransferase gene, FUT8 (alpha (1,6) fucosyltransferase), such that antibodies expressed in the Ms704, Ms705, and Ms709 cell lines lack fucose on their carbohydrates. The Ms704, Ms705, and Ms709 FUT8' cell lines were created by the targeted disruption of the FUT8 gene in CHO/DG44 cells using two replacement vectors 20 (see U.S. Patent Publication No. 20040110704 by Yamane et al. and Yamane-Ohnuki et al. (2004) Biotechnol Bioeng 87:614-22). As another example, EP 1,176,195 by Hanai et al. describes a cell line with a functionally disrupted FUT8 gene, which encodes a fucosyl transferase, such that antibodies expressed in such a cell line exhibit hypofucosylation by reducing or eliminating the alpha 1,6 bond-related enzyme. Hanai 25 et al. also describe cell lines which have a low enzyme activity for adding fucose to the N-acetylglucosamine that binds to the Fe region of the antibody or does not have the enzyme activity, for example the rat myeloma cell line YB2/0 (ATCC CRL 1662). PCT Publication WO 03/035835 by Presta describes a variant CHO cell line, Lec13 cells, with reduced ability to attach fucose to Asn(297)-linked carbohydrates, also resulting in 30 hypofucosylation of antibodies expressed in that host cell (see also Shields, R.L. et al. (2002) J. Biol. Chem. 277:26733-26740). PCT Publication WO 99/54342 by Umana et al. describes cell lines engineered to express glycoprotein-modifying glycosyl transferases (e.g., beta(1,4)-N-acetylglucosaminyltransferase III (GnTIII)) such that 37 MXI-347PC antibodies expressed in the engineered cell lines exhibit increased bisecting GlcNac structures which results in increased ADCC activity of the antibodies (see also Umana et al. (1999) Nat. Biotech. 17:176-180). Alternatively, the fucose residues of the antibody may be cleaved off using a fucosidase enzyme. For example, the fucosidase alpha-L 5 fucosidase removes fucosyl residues from antibodies (Tarentino, A.L. et al. (1975) Biochem. 14:5516-23). Another modification of the antibodies herein that is contemplated by the invention is pegylation. An antibody can be pegylated to, for example, increase the biological (e.g., serum) half life of the antibody. To pegylate an antibody, the antibody, 10 or fragment thereof, typically is reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment. Preferably, the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer). As used 15 herein, the term "polyethylene glycol" is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (Cl-C10) alkoxy- or aryloxy-polyethylene glycol or polyethylene glycol-maleimide. In certain embodiments, the antibody to be pegylated is an aglycosylated antibody. Methods for pegylating proteins are known in the art and can be applied to the antibodies of the invention. See 20 for example, EP 0 154 316 by Nishimura et al. and EP 0 401 384 by Ishikawa et al. Antibody Physical Properties The antibodies of the present invention may be further characterized by the various physical properties of the anti-Glypican-3 antibodies. Various assays may be used to detect and/or differentiate different classes of antibodies based on these physical 25 properties. In some embodiments, antibodies of the present invention may contain one or more glycosylation sites in either the light or heavy chain variable region. The presence of one or more glycosylation sites in the variable region may result in increased immunogenicity of the antibody or an alteration of the pK of the antibody due to altered 30 antigen binding (Marshall et al (1972) Annu Rev Biochem 41:673-702; Gala FA and Morrison SL (2004) JImmunol 172:5489-94; Wallick et al (1988) JExp Med 168:1099 109; Spiro RG (2002) Glycobiology 12:43R-56R; Parekh et al (1985) Nature 316:452-7; Mimura et al. (2000) Mol Immunol 37:697-706). Glycosylation has been known to 38 MXI-347PC occur at motifs containing an N-X-S/T sequence. Variable region glycosylation may be tested using a Glycoblot assay, which cleaves the antibody to produce a Fab, and then tests for glycosylation using an assay that measures periodate oxidation and Schiff base formation. Alternatively, variable region glycosylation may be tested using Dionex light 5 chromatography (Dionex-LC), which cleaves saccharides from a Fab into monosaccharides and analyzes the individual saccharide content. In some instances, it is preferred to have an anti-Glypican-3 antibody that does not contain variable region glycosylation. This can be achieved either by selecting antibodies that do not contain the glycosylation motif in the variable region or by mutating residues within the 10 glycosylation motif using standard techniques well known in the art. In a preferred embodiment, the antibodies of the present invention do not contain asparagine isomerism sites. A deamidation or isoaspartic acid effect may occur on N-G or D-G sequences, respectively. The deamidation or isoaspartic acid effect results in the creation of isoaspartic acid which decreases the stability of an antibody by creating a 15 kinked structure off a side chain carboxy terminus rather than the main chain. The creation of isoaspartic acid can be measured using an iso-quant assay, which uses a reverse-phase HPLC to test for isoaspartic acid. Each antibody will have a unique isoelectric point (pI), but generally antibodies will fall in the pH range of between 6 and 9.5. The pI for an IgGI antibody typically 20 falls within the pH range of 7-9.5 and the pI for an IgG4 antibody typically falls within the pH range of 6-8. Antibodies may have a pI that is outside this range. Although the effects are generally unknown, there is speculation that antibodies with a pI outside the normal range may have some unfolding and instability under in vivo conditions. The isoelectric point may be tested using a capillary isoelectric focusing assay, which creates 25 a pH gradient and may utilize laser focusing for increased accuracy (Janini et al (2002) Electrophoresis 23:1605-11; Ma et al. (2001) Chromatographia 53:S75-89; Hunt et al (1998) JChromatogr A 800:355-67). In some instances, it is preferred to have an anti Glypican-3 antibody that contains a pI value that falls in the normal range. This can be achieved either by selecting antibodies with a pI in the normal range, or by mutating 30 charged surface residues using standard techniques well known in the art. Each antibody will have a melting temperature that is indicative of thermal stability (Krishnamurthy R and Manning MC (2002) Curr Pharm Biotechnol 3:361-71). A higher thermal stability indicates greater overall antibody stability in vivo. The 39 MXI-347PC melting point of an antibody may be measure using techniques such as differential scanning calorimetry (Chen et al (2003) Pharm Res 20:1952-60; Ghirlando et al (1999) Immunol Lett 68:47-52). Tmi indicates the temperature of the initial unfolding of the antibody. TM2 indicates the temperature of complete unfolding of the antibody. 5 Generally, it is preferred that the Tmi of an antibody of the present invention is greater than 60 0 C, preferably greater than 65 0 C, even more preferably greater than 70 0 C. Alternatively, the thermal stability of an antibody may be measure using circular dichroism (Murray et al. (2002) J. Chromatogr Sci 40:343-9). In a preferred embodiment, antibodies are selected that do not rapidly degrade. 10 Fragmentation of an anti-Glypican-3 antibody may be measured using capillary electrophoresis (CE) and MALDI-MS, as is well understood in the art (Alexander AJ and Hughes DE (1995) Anal Chem 67:3626-32). In another preferred embodiment, antibodies are selected that have minimal aggregation effects. Aggregation may lead to triggering of an unwanted immune 15 response and/or altered or unfavorable pharmacokinetic properties. Generally, antibodies are acceptable with aggregation of 25% or less, preferably 20% or less, even more preferably 15% or less, even more preferably 10% or less and even more preferably 5% or less. Aggregation may be measured by several techniques well known in the art, including size-exclusion column (SEC) high performance liquid 20 chromatography (HPLC), and light scattering to identify monomers, dimers, trimers or multimers. Methods of Engineering Antibodies As discussed above, the anti-Glypican-3 antibodies having VH and VK sequences disclosed herein can be used to create new anti-Glypican-3 antibodies by modifying the 25 VH and/or VK sequences, or the constant region(s) attached thereto. Thus, in another aspect of the invention, the structural features of an anti-Glypican-3 antibody of the invention, e.g. 4A6, 1 1E7, and 16D 10, are used to create structurally related anti Glypican-3 antibodies that retain at least one functional property of the antibodies of the invention, such as binding to human Glypican-3. For example, one or more CDR 30 regions of 4A6, 1 1E7, and 16D 10, or mutations thereof, can be combined recombinantly with known framework regions and/or other CDRs to create additional, recombinantly engineered, anti-Glypican-3 antibodies of the invention, as discussed above. Other types of modifications include those described in the previous section. The starting material 40 MXI-347PC for the engineering method is one or more of the VH and/or VK sequences provided herein, or one or more CDR regions thereof. To create the engineered antibody, it is not necessary to actually prepare (i. e., express as a protein) an antibody having one or more of the VH and/or VK sequences provided herein, or one or more CDR regions thereof. 5 Rather, the information contained in the sequence(s) is used as the starting material to create a "second generation" sequence(s) derived from the original sequence(s) and then the "second generation" sequence(s) is prepared and expressed as a protein. Accordingly, in another embodiment, the invention provides a method for preparing an anti-Glypican-3 antibody comprising: 10 (a) providing: (i) a heavy chain variable region antibody sequence comprising a CDR1 sequence selected from the group consisting of SEQ ID NOs:1, 2, and 3, a CDR2 sequence selected from the group consisting of SEQ ID NOs:4, 5, and 6, 15 and/or a CDR3 sequence selected from the group consisting of SEQ ID NOs:7, 8, and 9; and/or (ii) a light chain variable region antibody sequence comprising a CDR1 sequence selected from the group consisting of SEQ ID NOs: 10, 11, and 12, a CDR2 20 sequence selected from the group consisting of SEQ ID NOs: 13, 14, and 15, and/or a CDR3 sequence selected from the group consisting of SEQ ID NOs: 16, 17, and 18; (b) altering at least one amino acid residue within the heavy chain variable 25 regionantibody sequence and/or the light chain variable region antibody sequence to create atleast one altered antibody sequence; and (c) expressing the altered antibody sequence as a protein. Standard molecular biology techniques can be used to prepare and express the 30 altered antibody sequence. 41 MXI-347PC Preferably, the antibody encoded by the altered antibody sequence(s) is one that retains one, some or all of the functional properties of the anti-Glypican-3 antibodies described herein, which functional properties include, but are not limited to: (i) binds to human Glypican-3 with a KD Of 1X10 7 M or less; 5 (ii) binds to human CHO cells transfected with Glypican-3. The functional properties of the altered antibodies can be assessed using standard assays available in the art and/or described herein, such as those set forth in the Examples (e.g., flow cytometry, binding assays). In certain embodiments of the methods of engineering antibodies of the 10 invention, mutations can be introduced randomly or selectively along all or part of an anti-Glypican-3 antibody coding sequence and the resulting modified anti-Glypican-3 antibodies can be screened for binding activity and/or other functional properties as described herein. Mutational methods have been described in the art. For example, PCT Publication WO 02/092780 by Short describes methods for creating and screening 15 antibody mutations using saturation mutagenesis, synthetic ligation assembly, or a combination thereof. Alternatively, PCT Publication WO 03/074679 by Lazar et al. describes methods of using computational screening methods to optimize physiochemical properties of antibodies. Nucleic Acid Molecules Encoding Antibodies of the Invention 20 Another aspect of the invention pertains to nucleic acid molecules that encode the antibodies of the invention. The nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form. A nucleic acid is "isolated" or "rendered substantially pure" when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by 25 standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and others well known in the art. See, F. Ausubel, et al., ed. (1987) Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York. A nucleic acid of the invention can be, for example, DNA or RNA and may or may not contain intronic sequences. In a preferred 30 embodiment, the nucleic acid is a cDNA molecule. Nucleic acids of the invention can be obtained using standard molecular biology techniques. For antibodies expressed by hybridomas (e.g., hybridomas prepared from transgenic mice carrying human immunoglobulin genes as described further below), 42 MXI-347PC cDNAs encoding the light and heavy chains of the antibody made by the hybridoma can be obtained by standard PCR amplification or cDNA cloning techniques. For antibodies obtained from an immunoglobulin gene library (e.g., using phage display techniques), nucleic acid encoding the antibody can be recovered from the library. 5 Preferred nucleic acids molecules of the invention are those encoding the VH and VL sequences of the 4A6, 1 1E7, or 16D10 monoclonal antibodies. DNA sequences encoding the VH sequences of 4A6, 1 1E7, or 16D10 are shown in SEQ ID NOs:25, 26, and 27, respectively. DNA sequences encoding the VL sequences of 4A6, 1 1E7, or 16D10 are shown in SEQ ID NOs:28, 29, and 30, respectively. 10 Other preferred nucleic acids of the invention are nucleic acids having at least 80% sequence identity, such as at least 85%, at least 90%, at least 95%, at least 98% or at least 99% sequence identity, with one of the sequences shown in SEQ ID NO:25, 26, 27, 28, 29, and 30, which nucleic acids encode an antibody of the invention, or an antigen-binding portion thereof. 15 The percent identity between two nucleic acid sequences is the number of positions in the sequence in which the nucleotide is identical, taking into account the number of gaps and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical 20 algorithm, such as the algorithm of Meyers and Miller or the XBLAST program of Altschul described above. Still further, preferred nucleic acids of the invention comprise one or more CDR encoding portions of the nucleic acid sequences shown in SEQ ID NOs: 25, 26, 27, 28, 29, and 30. In this embodiment, the nucleic acid may encode the heavy chain CDR1, 25 CDR2 and/or CDR3 sequence of 4A6, 1 1E7, or 16D10 or the light chain CDR1, CDR2 and/or CDR3 sequence of 4A6, 1 1E7, or 16D 10. Nucleic acids which have at least 80%, such as at least 85%, at least 90%, at least 95%, at least 98% or at least 99% sequence identity, with such a CDR-encoding portion of SEQ ID NO: 25, 26, 27, 28, 29, or 30 are also preferred nucleic acids of the invention. 30 Such nucleic acids may differ from the corresponding portion of SEQ ID NO: 25, 26, 27, 28, 29, or 30 in a non-CDR coding region and/or in a CDR-coding region. Where the difference is in a CDR-coding region, the nucleic acid CDR region encoded by the 43 MXI-347PC nucleic acid typically comprises one or more conservative sequence modification as defined herein compared to the corresponding CDR sequence of 4A6, 1 1E7, and 16D 10. Once DNA fragments encoding VH and VL segments are obtained, these DNA fragments can be further manipulated by standard recombinant DNA techniques, for 5 example to convert the variable region genes to full-length antibody chain genes, to Fab fragment genes or to a scFv gene. In these manipulations, a VL- or VH-encoding DNA fragment is operatively linked to another DNA fragment encoding another protein, such as an antibody constant region or a flexible linker. The term "operatively linked," as used in this context, is intended to mean that the two DNA fragments are joined such 10 that the amino acid sequences encoded by the two DNA fragments remain in-frame. The isolated DNA encoding the VH region can be converted to a full-length heavy chain gene by operatively linking the VH-encoding DNA to another DNA molecule encoding heavy chain constant regions (CHI, CH2 and CH3). The sequences of human heavy chain constant region genes are known in the art (see e.g., Kabat, E. A., 15 el al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification. The heavy chain constant region can be an IgGI, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region, but most preferably is an IgGI or IgG4 constant region. For a Fab 20 fragment heavy chain gene, the VH-encoding DNA can be operatively linked to another DNA molecule encoding only the heavy chain CHI constant region. The isolated DNA encoding the VL region can be converted to a full-length light chain gene (as well as a Fab light chain gene) by operatively linking the VL-encoding DNA to another DNA molecule encoding the light chain constant region, CL. The 25 sequences of human light chain constant region genes are known in the art (see e.g., Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification. In preferred embodiments, the light chain constant region can be a kappa 30 or lambda constant region. To create a scFv gene, the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4 -Ser)3, such that the VH and VL sequences can be expressed as a 44 MXI-347PC contiguous single-chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al. (1988) Science 242:423-426; Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883; McCafferty et al., (1990) Nature 348:552-554). Production of Monoclonal Antibodies 5 Monoclonal antibodies (mAbs) of the present invention can be produced by a variety of techniques, including conventional monoclonal antibody methodology e.g., the standard somatic cell hybridization technique of Kohler and Milstein (1975) Nature 256: 495. Although somatic cell hybridization procedures are preferred, in principle, other techniques for producing monoclonal antibody can be employed e.g., viral or 10 oncogenic transformation of B lymphocytes. The preferred animal system for preparing hybridomas is the murine system. Hybridoma production in the mouse is a very well-established procedure. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also 15 known. Chimeric or humanized antibodies of the present invention can be prepared based on the sequence of a non-human monoclonal antibody prepared as described above. DNA encoding the heavy and light chain immunoglobulins can be obtained from the non-human hybridoma of interest and engineered to contain non-murine (e.g., human) 20 immunoglobulin sequences using standard molecular biology techniques. For example, to create a chimeric antibody, murine variable regions can be linked to human constant regions using methods known in the art (see e.g., U.S. Patent No. 4,816,567 to Cabilly et al.). To create a humanized antibody, murine CDR regions can be inserted into a human framework using methods known in the art (see e.g., U.S. Patent No. 5,225,539 to 25 Winter, and U.S. Patent Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen et al.). In a preferred embodiment, the antibodies of the invention are human monoclonal antibodies. Such human monoclonal antibodies directed against Glypican-3 can be generated using transgenic or transchromosomic mice carrying parts of the 30 human immune system rather than the mouse system. These transgenic and transchromosomic mice include mice referred to herein as the HuMAb Mouse® and KM Mouse, respectively, and are collectively referred to herein as "human Ig mice." 45 MXI-347PC The HuMAb Mouse® (Medarex®, Inc.) contains human immunoglobulin gene miniloci that encode unrearranged human heavy (pi and y) and K light chain immunoglobulin sequences, together with targeted mutations that inactivate the endogenous pt and K chain loci (see e.g., Lonberg, et al. (1994) Nature 368(6474): 856 5 859). Accordingly, the mice exhibit reduced expression of mouse IgM or K, and in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgGK monoclonal antibodies (Lonberg, N. et al. (1994), supra; reviewed in Lonberg, N. (1994) Handbook of Experimental Pharmacology 113:49-101; Lonberg, N. and Huszar, D. 10 (1995) Intern. Rev. Immunol. 13: 65-93, and Harding, F. and Lonberg, N. (1995) Ann. N.Y. Acad. Sci. 764:536-546). Preparation and use of the HuMAb Mouse®, and the genomic modifications carried by such mice, is further described in Taylor, L. et al. (1992) Nucleic Acids Research 20:6287-6295; Chen, J. et al. (1993) International Immunology 5: 647-656; Tuaillon et al. (1993) Proc. Natl. Acad. Sci. USA 90:3720 15 3724; Choi et al. (1993) Nature Genetics 4:117-123; Chen, J. et al. (1993) EMBO J. 12: 821-830; Tuaillon et al. (1994) J. Immunol. 152:2912-2920; Taylor, L. et al. (1994) International Immunology 6: 579-591; and Fishwild, D. et al. (1996) Nature Biotechnology 14: 845-85 1, the contents of all of which are hereby specifically incorporated by reference in their entirety. See further, U.S. Patent Nos. 5,545,806; 20 5,569,825; 5,625,126; 5,633,425; 5,789,650; 5,877,397; 5,661,016; 5,814,318; 5,874,299; and 5,770,429; all to Lonberg and Kay; U.S. Patent No. 5,545,807 to Surani et al.; PCT Publication Nos. WO 92/03918, WO 93/12227, WO 94/25585, WO 97/13852, WO 98/24884 and WO 99/45962, all to Lonberg and Kay; and PCT Publication No. WO 01/14424 to Korman et al. 25 In another embodiment, human antibodies of the invention can be raised using a mouse that carries human immunoglobulin sequences on transgenes and transchomosomes, such as a mouse that carries a human heavy chain transgene and a human light chain transchromosome. This mouse is referred to herein as a "KM mouse," and is described in detail in PCT Publication WO 02/43478 to Ishida et al. 30 Still further, alternative transgenic animal systems expressing human immunoglobulin genes are available in the art and can be used to raise anti-Glypican-3 antibodies of the invention. For example, an alternative transgenic system referred to as the Xenomouse (Abgenix, Inc.) can be used; such mice are described in, for example, 46 MXI-347PC U.S. Patent Nos. 5,939,598; 6,075,181; 6,114,598; 6,150,584 and 6,162,963 to Kucherlapati et al. Moreover, alternative transchromosomic animal systems expressing human immunoglobulin genes are available in the art and can be used to raise anti-Glypican-3 5 antibodies of the invention. For example, mice carrying both a human heavy chain transchromosome and a human light chain tranchromosome, referred to as "TC mice" can be used; such mice are described in Tomizuka et al. (2000) Proc. Natl. Acad. Sci. USA 97:722-727. Furthermore, cows carrying human heavy and light chain transchromosomes have been described in the art (Kuroiwa et al. (2002) Nature 10 Biotechnology 20:889-894) and PCT application No. WO/2002/092812 and can be used to raise anti-Glypican-3 antibodies of the invention. Human monoclonal antibodies of the invention can also be prepared using phage display methods for screening libraries of human immunoglobulin genes. Such phage display methods for isolating human antibodies are established in the art. See for 15 example: U.S. Patent Nos. 5,223,409; 5,403,484; and 5,571,698 to Ladner et al.; U.S. Patent Nos. 5,427,908 and 5,580,717 to Dower et al.; U.S. Patent Nos. 5,969,108 and 6,172,197 to McCafferty et al.; and U.S. Patent Nos. 5,885,793; 6,521,404; 6,544,731; 6,555,313; 6,582,915 and 6,593,081 to Griffiths et al. Human monoclonal antibodies of the invention can also be prepared using SCID 20 mice into which human immune cells have been reconstituted such that a human antibody response can be generated upon immunization. Such mice are described in, for example, U.S. Patent Nos. 5,476,996 and 5,698,767 to Wilson et al. Immunization of Human Ig Mice When human Ig mice are used to raise human antibodies of the invention, such 25 mice can be immunized with a purified or enriched preparation of Glypican-3 antigen and/or recombinant Glypican-3, or cells expressing Glypican-3, or a Glypican-3 fusion protein, as described by Lonberg, N. et al. (1994) Nature 368(6474): 856-859; Fishwild, D. et al. (1996) Nature Biotechnology 14: 845-85 1; and PCT Publication WO 98/24884 and WO 01/14424. Preferably, the mice will be 6-16 weeks of age upon the first 30 infusion. For example, a purified or recombinant preparation (5-50 tg) of Glypican-3 antigen can be used to immunize the human Ig mice intraperitoneally. 47 MXI-347PC Detailed procedures to generate fully human monoclonal antibodies to Glypican 3 are described in Example 1 below. Cumulative experience with various antigens has shown that the transgenic mice respond when initially immunized intraperitoneally (IP) with antigen in complete Freund's adjuvant, followed by every other week IP 5 immunizations (up to a total of 6) with antigen in incomplete Freund's adjuvant. However, adjuvants other than Freund's are also found to be effective. In addition, whole cells in the absence of adjuvant are found to be highly immunogenic. The immune response can be monitored over the course of the immunization protocol with plasma samples being obtained by retroorbital bleeds. The plasma can be screened by ELISA 10 (as described below), and mice with sufficient titers of anti-Glypican-3 human immunoglobulin can be used for fusions. Mice can be boosted intravenously with antigen 3 days before sacrifice and removal of the spleen. It is expected that 2-3 fusions for each immunization may need to be performed. Between 6 and 24 mice are typically immunized for each antigen. In one embodiment, mouse strains bearing an HCo7, 15 HCo 12 or HCo 17 human heavy chain transgene strains may used. Alternatively or additionally, the KM Mouse® strain can be used. In addition, two or more of these strains can be bred together into a single mouse having a plurality of different human heavy chain transgenes. Generation of Hybridomas Producing Human Monoclonal Antibodies 20 To generate hybridomas producing human monoclonal antibodies of the invention, splenocytes and/or lymph node cells from immunized mice can be isolated and fused to an appropriate immortalized cell line, such as a mouse myeloma cell line. The resulting hybridomas can be screened for the production of antigen-specific antibodies. For example, single cell suspensions of splenic lymphocytes from 25 immunized mice can be fused to one-sixth the number of P3X63-Ag8.653 nonsecreting mouse myeloma cells (ATCC, CRL 1580) with 50% PEG. Alternatively, the single cell suspension of splenic lymphocytes from immunized mice can be fused using an electric field based electrofusion method, using a CytoPulse large chamber cell fusion electroporator (CytoPulse Sciences, Inc., Glen Burnie Maryland). Cells are plated at 30 approximately 2 x 10 5 in flat bottom microtiter plate, followed by a two week incubation in selective medium containing 20% fetal Clone Serum, 18% "653" conditioned media, 5% origen (IGEN), 4 mM L-glutamine, 1 mM sodium pyruvate, 5mM HEPES, 0.055 mM 2-mercaptoethanol, 50 units/ml penicillin, 50 mg/ml streptomycin, 50 mg/ml 48 MXI-347PC gentamycin and IX HAT (Sigma; the HAT is added 24 hours after the fusion). After approximately two weeks, cells can be cultured in medium in which the HAT is replaced with HT. Individual wells can then be screened by ELISA for human monoclonal IgM and IgG antibodies. Once extensive hybridoma growth occurs, medium can be observed 5 usually after 10-14 days. The antibody secreting hybridomas can be replated, screened again, and if still positive for human IgG, the monoclonal antibodies can be subeloned at least twice by limiting dilution. The stable subclones can then be cultured in vitro to generate small amounts of antibody in tissue culture medium for characterization. To purify human monoclonal antibodies, selected hybridomas can be grown in 10 two-liter spinner-flasks for monoclonal antibody purification. Supernatants can be filtered and concentrated before affinity chromatography with protein A-sepharose (Pharmacia, Piscataway, N.J.). Eluted IgG can be checked by gel electrophoresis and high performance liquid chromatography to ensure purity. The buffer solution can be exchanged into PBS, and the concentration can be determined by OD280 using 1.43 15 extinction coefficient. The monoclonal antibodies can be aliquoted and stored at -80' C. Generation of Transfectomas Producing Monoclonal Antibodies Antibodies of the invention also can be produced in a host cell transfectoma using, for example, a combination of recombinant DNA techniques and gene transfection methods as is well known in the art (e.g., Morrison, S. (1985) Science 20 229:1202). For example, to express the antibodies, or antibody fragments thereof, DNAs encoding partial or full-length light and heavy chains, can be obtained by standard molecular biology techniques (e.g., PCR amplification or cDNA cloning using a hybridoma that expresses the antibody of interest) and the DNAs can be inserted into 25 expression vectors such that the genes are operatively linked to transcriptional and translational control sequences. In this context, the term "operatively linked" is intended to mean that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene. The expression vector 30 and expression control sequences are chosen to be compatible with the expression host cell used. The antibody light chain gene and the antibody heavy chain gene can be inserted into separate vector or, more typically, both genes are inserted into the same expression vector. The antibody genes are inserted into the expression vector by 49 MXI-347PC standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present). The light and heavy chain variable regions of the antibodies described herein can be used to create full-length antibody genes of any antibody isotype by inserting them into expression 5 vectors already encoding heavy chain constant and light chain constant regions of the desired isotype such that the VH segment is operatively linked to the CH segment(s) within the vector and the VK segment is operatively linked to the CL segment within the vector. Additionally or alternatively, the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell. The 10 antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene. The signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein). In addition to the antibody chain genes, the recombinant expression vectors of 15 the invention carry regulatory sequences that control the expression of the antibody chain genes in a host cell. The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes. Such regulatory sequences are described, for example, in Goeddel (Gene Expression 20 Technology. Methods in Enzymology 185, Academic Press, San Diego, CA (1990)). It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences, may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. Preferred regulatory sequences for mammalian host cell expression include viral 25 elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV), Simian Virus 40 (SV40), adenovirus, (e.g., the adenovirus major late promoter (AdMLP) and polyoma. Alternatively, nonviral regulatory sequences may be used, such as the ubiquitin promoter or p-globin promoter. Still further, regulatory elements composed of 30 sequences from different sources, such as the SRa promoter system, which contains sequences from the SV40 early promoter and the long terminal repeat of human T cell leukemia virus type 1 (Takebe, Y. et al. (1988) Mol. Cell. Biol. 8:466-472). 50 MXI-347PC In addition to the antibody chain genes and regulatory sequences, the recombinant expression vectors of the invention may carry additional sequences, such as sequences that regulate replication of the vector in host cells (e.g., origins of replication) and selectable marker genes. The selectable marker gene facilitates selection of host 5 cells into which the vector has been introduced (see, e.g., U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al.). For example, typically the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced. Preferred selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr- host cells with 10 methotrexate selection/amplification) and the neo gene (for G418 selection). For expression of the light and heavy chains, the expression vector(s) encoding the heavy and light chains is transfected into a host cell by standard techniques. The various forms of the term "transfection" are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or 15 eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE dextran transfection and the like. Although it is theoretically possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells, and most preferably mammalian host cells, is the most preferred because such eukaryotic cells, and in particular mammalian cells, are more 20 likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody. Prokaryotic expression of antibody genes has been reported to be ineffective for production of high yields of active antibody (Boss, M. A. and Wood, C. R. (1985) Immunology Today 6:12-13). Preferred mammalian host cells for expressing the recombinant antibodies of the 25 invention include Chinese Hamster Ovary (CHO cells) (including dhfr- CHO cells, described in Urlaub and Chasin, (1980) Proc. NatI. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R. J. Kaufman and P. A. Sharp (1982) J. Mol. Biol. 159:601-621), NSO myeloma cells, COS cells and SP2 cells. In particular, for use with NSO myeloma cells, another preferred expression system is the 30 GS gene expression system disclosed in WO 87/04462 (to Wilson), WO 89/01036 (to Bebbington) and EP 338,841 (to Bebbington). When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for 51 MXI-347PC expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods. Characterization of Antibody Binding to Antigen 5 Antibodies of the invention can be tested for binding to Glypican-3 by, for example, standard ELISA. Briefly, microtiter plates are coated with purified Glypican-3 at 0.25 L ptg/ml in PBS, and then blocked with 50% bovine serum albumin in PBS. Dilutions of antibody (e.g., dilutions of plasma from Glypican-3-immunized mice) are added to each well and incubated for 1-2 hours at 37'C. The plates are washed with 10 PBS/Tween and then incubated with secondary reagent (e.g., for human antibodies, a goat-anti-human IgG Fe-specific polyclonal reagent) conjugated to alkaline phosphatase for 1 hour at 37'C. After washing, the plates are developed with pNPP substrate (1 mg/ml), and analyzed at OD of 405-650. Preferably, mice which develop the highest titers will be used for fusions. 15 An ELISA assay as described above can also be used to screen for hybridomas that show positive reactivity with a Glypican-3 immunogen. Hybridomas that bind with high avidity to Glypican-3 are subeloned and further characterized. One clone from each hybridoma, which retains the reactivity of the parent cells (by ELISA), can be chosen for making a 5-10 vial cell bank stored at -140'C, and for antibody purification. 20 To purify anti-Glypican-3 antibodies, selected hybridomas can be grown in two liter spinner-flasks for monoclonal antibody purification. Supernatants can be filtered and concentrated before affinity chromatography with protein A-sepharose (Pharmacia, Piscataway, NJ). Eluted IgG can be checked by gel electrophoresis and high performance liquid chromatography to ensure purity. The buffer solution can be 25 exchanged into PBS, and the concentration can be determined by OD280 using 1.43 extinction coefficient. The monoclonal antibodies can be aliquoted and stored at -80'C. To determine if the selected anti-Glypican-3 monoclonal antibodies bind to unique epitopes, each antibody can be biotinylated using commercially available reagents (Pierce, Rockford, IL). Competition studies using unlabeled monoclonal 30 antibodies and biotinylated monoclonal antibodies can be performed using Glypican-3 coated-ELISA plates as described above. Biotinylated mAb binding can be detected with a strep-avidin-alkaline phosphatase probe. 52 MXI-347PC To determine the isotype of purified antibodies, isotype ELISAs can be performed using reagents specific for antibodies of a particular isotype. For example, to determine the isotype of a human monoclonal antibody, wells of microtiter plates can be coated with 1 ptg/ml of anti-human immunoglobulin overnight at 4'C. After blocking 5 with 1% BSA, the plates are reacted with 1 tg /ml or less of test monoclonal antibodies or purified isotype controls, at ambient temperature for one to two hours. The wells can then be reacted with either human IgGI or human IgM-specific alkaline phosphatase conjugated probes. Plates are developed and analyzed as described above. Anti-Glypican-3 human IgGs can be further tested for reactivity with Glypican-3 10 antigen by Western blotting. Briefly, Glypican-3 can be prepared and subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis. After electrophoresis, the separated antigens are transferred to nitrocellulose membranes, blocked with 10% fetal calf serum, and probed with the monoclonal antibodies to be tested. Human IgG binding can be detected using anti-human IgG alkaline phosphatase and developed with 15 BCIP/NBT substrate tablets (Sigma Chem. Co., St. Louis, Mo.). The binding specificity of an antibody of the invention may also be determined by monitoring binding of the antibody to cells expressing Glypican-3, for example by flow cytometry. Typically, a cell line, such as a CHO cell line, may be transfected with an expression vector encoding a transmembrane form of Glypican-3. The transfected 20 protein may comprise a tag, such as a mye-tag, preferably at the N-terminus, for detection using an antibody to the tag. Binding of an antibody of the invention to Glypican-3 may be determined by incubating the transfected cells with the antibody, and detecting bound antibody. Binding of an antibody to the tag on the transfected protein may be used as a positive control. 25 The specificity of an antibody of the invention for Glypican-3 may be further studied by determining whether or not the antibody binds to other proteins (e.g. Glypican- 1, Glypican-2, Glypican-4 or Glypican-6) or to Glypican-3 itself using the same methods by which binding to Glypican-3 is determined. Immunoconjugates 30 In another aspect, the present invention features an anti-Glypican-3 antibody, or a fragment thereof, conjugated to a therapeutic moiety, such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radiotoxin. Such conjugates are referred to herein as "immunoconjugates". Immunoconjugates that include one or more cytotoxins are 53 MXI-347PC referred to as "immunotoxins." A cytotoxin or cytotoxic agent includes any agent that is detrimental to (e.g., kills) cells. Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, 5 mithramycin, actinomycin D, 1 -dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents also include, for example, antimetabolites (e.g., methotrexate, 6 mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) 10 and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine). 15 Other preferred examples of therapeutic cytotoxins that can be conjugated to an antibody of the invention include duocarmycins, calicheamicins, maytansines and auristatins, and derivatives thereof. An example of a calicheamicin antibody conjugate is commercially available (Mylotarg@; American Home Products). Cytotoxins can be conjugated to antibodies of the invention using linker 20 technology available in the art. Examples of linker types that have been used to conjugate a cytotoxin to an antibody include, but are not limited to, hydrazones, thioethers, esters, disulfides and peptide-containing linkers. A linker can be chosen that is, for example, susceptible to cleavage by low pH within the lysosomal compartment or susceptible to cleavage by proteases, such as proteases preferentially expressed in tumor 25 tissue such as cathepsins (e.g., cathepsins B, C, D). Examples of cytotoxins are described, for example, in U.S. Patent Nos. 6,989,452, 7,087,600, and 7,129,261, and in PCT Application Nos. PCT/US02/17210, PCT/US2005/017804, PCT/US06/37793, PCT/US06/060050, PCT/US2006/060711, WO/2006/110476, and in U.S. Patent Application No. 60/891,028, all of which are 30 incorporated herein by reference in their entirety. For further discussion of types of cytotoxins, linkers and methods for conjugating therapeutic agents to antibodies, see also Saito, G. et al. (2003) Adv. Drug Deliv. Rev. 55:199-215; Trail, P.A. et al. (2003) Cancer Immunol. Immunother. 52:328-337; Payne, G. (2003) Cancer Cell 3:207-212; 54 MXI-347PC Allen, T.M. (2002) Nat. Rev. Cancer 2:750-763; Pastan, I. and Kreitman, R. J. (2002) Curr. Opin. Investig. Drugs 3:1089-1091; Senter, P.D. and Springer, C.J. (2001) Adv. Drug Deliv. Rev. 53:247-264. Antibodies of the present invention also can be conjugated to a radioactive 5 isotope to generate cytotoxic radiopharmaceuticals, also referred to as radioimmunoconjugates. Examples of radioactive isotopes that can be conjugated to antibodies for use diagnostically or therapeutically include, but are not limited to, iodine m, indium", yttrium 0 and lutetiumm. Method for preparing radioimmunconjugates are established in the art. Examples of radioimmunoconjugates 10 are commercially available, including Zevalin@ (IDEC Pharmaceuticals) and Bexxar@ (Corixa Pharmaceuticals), and similar methods can be used to prepare radioimmunoconjugates using the antibodies of the invention. The antibody conjugates of the invention can be used to modify a given biological response, and the drug moiety is not to be construed as limited to classical 15 chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, an enzymatically active toxin, or active fragment thereof, such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor or interferon-y; or, biological response modifiers such as, for example, lymphokines, 20 interleukin-1 ("IL-1"), interleukin-2 ("IL-2"), interleukin-6 ("IL-6"), granulocyte macrophage colony stimulating factor ("GM-CSF"), granulocyte colony stimulating factor ("G-CSF"), or other growth factors. Techniques for conjugating such therapeutic moiety to antibodies are well known, see, e.g., Arnon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs 25 In Cancer Therapy," in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Hellstrom et al., "Antibodies For Drug Delivery," in Controlled Drug Delivery ( 2 nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review," in Monoclonal Antibodies '84: Biological And Clinical 30 Applications, Pinchera et al. (eds.), pp. 475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy," in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., Immunol. Rev., 62:119-58 (1982). 55 MXI-347PC Bispecific Molecules In another aspect, the present invention features bispecific molecules comprising an anti-Glypican-3 antibody, or a fragment thereof, of the invention. An antibody of the invention, or antigen-binding portions thereof, can be derivatized or linked to another 5 functional molecule, e.g., another peptide or protein (e.g., another antibody or ligand for a receptor) to generate a bispecific molecule that binds to at least two different binding sites or target molecules. The antibody of the invention may in fact be derivatized or linked to more than one other functional molecule to generate multispecific molecules that bind to more than two different binding sites and/or target molecules; such 10 multispecific molecules are also intended to be encompassed by the term "bispecific molecule" as used herein. To create a bispecific molecule of the invention, an antibody of the invention can be functionally linked (e.g., by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other binding molecules, such as another antibody, antibody fragment, peptide or binding mimetic, such that a bispecific 15 molecule results. Accordingly, the present invention includes bispecific molecules comprising at least one first binding specificity for Glypican-3 and a second binding specificity for a second target epitope. In a particular embodiment of the invention, the second target epitope is an Fe receptor, e.g., human FcyRI (CD64) or a human Fe receptor (CD89). 20 Therefore, the invention includes bispecific molecules capable of binding both to FcyR or FcaR expressing effector cells (e.g., monocytes, macrophages or polymorphonuclear cells (PMNs)), and to target cells expressing Glypican-3. These bispecific molecules target Glypican-3 expressing cells to effector cell and trigger Fe receptor-mediated effector cell activities, such as phagocytosis of a Glypican-3 expressing cells, antibody 25 dependent cell-mediated cytotoxicity (ADCC), cytokine release, or generation of superoxide anion. In an embodiment of the invention in which the bispecific molecule is multispecific, the molecule can further include a third binding specificity, in addition to an anti-Fe binding specificity and an anti-Glypican-3 binding specificity. In one 30 embodiment, the third binding specificity is an anti-enhancement factor (EF) portion, e.g., a molecule which binds to a surface protein involved in cytotoxic activity and thereby increases the immune response against the target cell. The "anti-enhancement factor portion" can be an antibody, functional antibody fragment or a ligand that binds to 56 MXI-347PC a given molecule, e.g., an antigen or a receptor, and thereby results in an enhancement of the effect of the binding determinants for the Fe receptor or target cell antigen. The "anti-enhancement factor portion" can bind an Fe receptor or a target cell antigen. Alternatively, the anti-enhancement factor portion can bind to an entity that is different 5 from the entity to which the first and second binding specificities bind. For example, the anti-enhancement factor portion can bind a cytotoxic T-cell (e.g. via CD2, CD3, CD8, CD28, CD4, CD40, ICAM- 1 or other immune cell that results in an increased immune response against the target cell). In one embodiment, the bispecific molecules of the invention comprise as a 10 binding specificity at least one antibody, or an antibody fragment thereof, including, e.g., an Fab, Fab', F(ab') 2 , Fv, Fd, dAb or a single chain Fv. The antibody may also be a light chain or heavy chain dimer, or any minimal fragment thereof such as a Fv or a single chain construct as described in U.S. Patent No. 4,946,778 to Ladner et al., the contents of which is expressly incorporated by reference. 15 In one embodiment, the binding specificity for an Fey receptor is provided by a monoclonal antibody, the binding of which is not blocked by human immunoglobulin G (IgG). As used herein, the term "IgG receptor" refers to any of the eight y-chain genes located on chromosome 1. These genes encode a total of twelve transmembrane or soluble receptor isoforms which are grouped into three Fcy receptor classes: FcyRI 20 (CD64), FcyRII(CD32), and FcyRIII (CD16). In one preferred embodiment, the Fey receptor a human high affinity FcyRI. The human FcyRI is a 72 kDa molecule, which shows high affinity for monomeric IgG (108 - 109 M 1 ). The production and characterization of certain preferred anti-Fcy D Dmonoclonal antibodies are described in PCT Publication WO 88/00052 and in U.S. Patent No. 25 4,954,617 to Fanger et al., the teachings of which are fully incorporated by reference herein. These antibodies bind to an epitope of FcyRI, FcyRII or FcyRIII at a site which is distinct from the Fcy binding site of the receptor and, thus, their binding is not blocked substantially by physiological levels of IgG. Specific anti-FcyRI antibodies useful in this invention are mAb 22, mAb 32, mAb 44, mAb 62 and mAb 197. The hybridoma 30 producing mAb 32 is available from the American Type Culture Collection, ATCC Accession No. HB9469. In other embodiments, the anti-Fcy receptor antibody is a humanized form of monoclonal antibody 22 (H22). The production and characterization of the H22 antibody is described in Graziano, R.F. et al. (1995) J. Immunol 155 (10): 57 MXI-347PC 4996-5002 and PCT Publication WO 94/10332 to Tempest et al.. The H22 antibody producing cell line was deposited at the American Type Culture Collection under the designation HA022CL1 and has the accession no. CRL 11177. In still other preferred embodiments, the binding specificity for an Fe receptor is 5 provided by an antibody that binds to a human IgA receptor, e.g., an Fc-alpha receptor (FeaRI (CD89)), the binding of which is preferably not blocked by human immunoglobulin A (IgA). The term "IgA receptor" is intended to include the gene product of one a-gene (FeaRI) located on chromosome 19. This gene is known to encode several alternatively spliced transmembrane isoforms of 55 to 110 kDa. FcaRI 10 (CD89) is constitutively expressed on monocytes/macrophages, eosinophilic and neutrophilic granulocytes, but not on non-effector cell populations. FcURI has medium affinity (,z 5 x 107 M 1 ) for both IgAl and IgA2, which is increased upon exposure to cytokines such as G-CSF or GM-CSF (Morton, H.C. et al. (1996) Critical Reviews in Immunology 16:423-440). Four FeaRI-specific monoclonal antibodies, identified as A3, 15 A59, A62 and A77, which bind FcaRI outside the IgA ligand binding domain, have been described (Monteiro, R.C. et al. (1992) J. Immunol. 148:1764). FcaRl and FcyRI are preferred trigger receptors for use in the bispecific molecules of the invention because they are (1) expressed primarily on immune effector cells, e.g., monocytes, PMNs, macrophages and dendritic cells; (2) expressed at high 20 levels (e.g., 5,000-100,000 per cell); (3) mediators of cytotoxic activities (e.g., ADCC, phagocytosis); and (4) mediate enhanced antigen presentation of antigens, including self-antigens, targeted to them. While human monoclonal antibodies are preferred, other antibodies which can be employed in the bispecific molecules of the invention are murine, chimeric and 25 humanized monoclonal antibodies. The bispecific molecules of the present invention can be prepared by conjugating the constituent binding specificities, e.g., the anti-FcR and anti-Glypican-3 binding specificities, using methods known in the art. For example, each binding specificity of the bispecific molecule can be generated separately and then conjugated to one another. 30 When the binding specificities are proteins or peptides, a variety of coupling or cross linking agents can be used for covalent conjugation. Examples of cross-linking agents include protein A, carbodiimide, N-succinimidyl-S-acetyl-thioacetate (SATA), 5,5' dithiobis(2-nitrobenzoic acid) (DTNB), o-phenylenedimaleimide (oPDM), N 58 MXI-347PC succinimidyl-3-(2-pyridyldithio)propionate (SPDP), and sulfosuccinimidyl 4-(N maleimidomethyl) cyclohaxane-1-carboxylate (sulfo-SMCC) (see e.g., Karpovsky et al. (1984) J. Exp. Med. 160:1686; Liu, MA et al. (1985) Proc. Natl. Acad. Sci. USA 82:8648). Other methods include those described in Paulus (1985) Behring Ins. Mitt. 5 No. 78, 118-132; Brennan et al. (1985) Science 229:81-83, and Glennie et al. (1987) J. Immunol. 139: 2367-2375). Preferred conjugating agents are SATA and sulfo-SMCC, both available from Pierce Chemical Co. (Rockford, IL). When the binding specificities are antibodies, they can be conjugated via sulfhydryl bonding of the C-terminus hinge regions of the two heavy chains. In a 10 particularly preferred embodiment, the hinge region is modified to contain an odd number of sulfhydryl residues, preferably one, prior to conjugation. Alternatively, both binding specificities can be encoded in the same vector and expressed and assembled in the same host cell. This method is particularly useful where the bispecific molecule is a mAb x mAb, mAb x Fab, Fab x F(ab') 2 or ligand x Fab 15 fusion protein. A bispecific molecule of the invention can be a single chain molecule comprising one single chain antibody and a binding determinant, or a single chain bispecific molecule comprising two binding determinants. Bispecific molecules may comprise at least two single chain molecules. Methods for preparing bispecific molecules are described for example in U.S. Patent Numbers 5,260,203; 5,455,030; 20 4,881,175; 5,132,405; 5,091,513; 5,476,786; 5,013,653; 5,258,498; and 5,482,858, all of which are expressly incorporated herein by reference. Binding of the bispecific molecules to their specific targets can be confirmed by, for example, enzyme-linked immunosorbant assay (ELISA), radioimmunoassay (RIA), FACS analysis, bioassay (e.g., growth inhibition), or Western Blot assay. Each of these 25 assays generally detects the presence of protein-antibody complexes of particular interest by employing a labeled reagent (e.g., an antibody) specific for the complex of interest. For example, the FcR-antibody complexes can be detected using e.g., an enzyme-linked antibody or antibody fragment which recognizes and specifically binds to the antibody-FcR complexes. Alternatively, the complexes can be detected using any of 30 a variety of other immunoassays. For example, the antibody can be radioactively labeled and used in a radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March, 1986, which is incorporated by reference 59 MXI-347PC herein). The radioactive isotope can be detected by such means as the use of a ily Counter or a scintillation counter or by autoradiography. Pharmaceutical Compositions In another aspect, the present invention provides a composition, e.g., a 5 pharmaceutical composition, containing one or a combination of monoclonal antibodies, or antigen-binding portion(s) thereof, of the present invention, formulated together with a pharmaceutically acceptable carrier. Such compositions may include one or a combination of (e.g., two or more different) antibodies, or immunoconjugates or bispecific molecules of the invention. For example, a pharmaceutical composition of the 10 invention can comprise a combination of antibodies (or immunoconjugates or bispecifics) that bind to different epitopes on the target antigen or that have complementary activities. Pharmaceutical compositions of the invention also can be administered in combination therapy, i. e., combined with other agents. For example, the combination 15 therapy can include an anti-Glypican-3 antibody of the present invention combined with at least one other anti-inflammatory or immunosuppressant agent. Examples of therapeutic agents that can be used in combination therapy are described in greater detail below in the section on uses of the antibodies of the invention. As used herein, "pharmaceutically acceptable carrier" includes any and all 20 solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion). Depending on the route of administration, the active compound, i.e., antibody, immunoconjugate, or bispecific 25 molecule, may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound. The pharmaceutical compounds of the invention may include one or more pharmaceutically acceptable salts. A "pharmaceutically acceptable salt" refers to a salt that retains the desired biological activity of the parent compound and does not impart 30 any undesired toxicological effects (see e.g., Berge, S.M., et al. (1977) J Pharm. Sci. 66:1-19). Examples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the 60 MXI-347PC like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like. Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and 5 the like, as well as from nontoxic organic amines, such as N,N' dibenzylethylenediamine, N-methylgluc amine, chloroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like. A pharmaceutical composition of the invention also may include a pharmaceutically acceptable anti-oxidant. Examples of pharmaceutically acceptable 10 antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid 15 (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. Examples of suitable aqueous and nonaqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl 20 oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of 25 microorganisms may be ensured both by sterilization procedures, supra, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought 30 about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin. 61 MXI-347PC Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is 5 incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions. Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a 10 solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance 15 of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and 20 gelatin. Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration. Generally, dispersions are prepared by incorporating the active compound into a sterile 25 vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. 30 The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, and the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that 62 MXI-347PC amount of the composition which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 0.01 per cent to about ninety-nine percent of active ingredient, preferably from about 0.1 per cent to about 70 per cent, most preferably from about 1 per cent to about 30 per cent of active ingredient in 5 combination with a pharmaceutically acceptable carrier. Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially 10 advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The 15 specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals. For administration of the antibody, the dosage ranges from about 0.0001 to 100 20 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight. For example dosages can be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg. An exemplary treatment regime entails administration once per week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 25 months or once every three to 6 months. Preferred dosage regimens for an anti Glypican-3 antibody of the invention include 1 mg/kg body weight or 3 mg/kg body weight via intravenous administration, with the antibody being given using one of the following dosing schedules: (i) every four weeks for six dosages, then every three months; (ii) every three weeks; (iii) 3 mg/kg body weight once followed by 1 mg/kg 30 body weight every three weeks. In some methods, two or more monoclonal antibodies with different binding specificities are administered simultaneously, in which case the dosage of each antibody administered falls within the ranges indicated. Antibody is usually administered on 63 MXI-347PC multiple occasions. Intervals between single dosages can be, for example, weekly, monthly, every three monthgs or yearly. Intervals can also be irregular as indicated by measuring blood levels of antibody to the target antigen in the patient. In some methods, dosage is adjusted to achieve a plasma antibody concentration of about 1-1000 pig /ml 5 and in some methods about 25-300 pig /ml. Alternatively, antibody can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the antibody in the patient. In general, human antibodies show the longest half life, followed by humanized antibodies, chimeric antibodies, and 10 nonhuman antibodies. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is 15 sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime. Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient 20 which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the 25 rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. 30 A "therapeutically effective dosage" of an anti-Glypican-3 antibody of the invention preferably results in a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction. For example, for the treatment of Glypican-3 64 MXI-347PC tumors, a "therapeutically effective dosage" preferably inhibits cell growth or tumor growth by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects. The ability of a compound to inhibit tumor growth can be 5 evaluated in an animal model system predictive of efficacy in human tumors. Alternatively, this property of a composition can be evaluated by examining the ability of the compound to inhibit cell growth, such inhibition can be measured in vitro by assays known to the skilled practitioner. A therapeutically effective amount of a therapeutic compound can decrease tumor size, or otherwise ameliorate symptoms in a 10 subject. One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected. A composition of the present invention can be administered via one or more routes of administration using one or more of a variety of methods known in the art. As 15 will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. Preferred routes of administration for antibodies of the invention include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion. The phrase "parenteral administration" as used herein 20 means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. 25 Alternatively, an antibody of the invention can be administered via a non parenteral route, such as a topical, epidermal or mucosal route of administration, for example, intranasally, orally, vaginally, rectally, sublingually or topically. The active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including 30 implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the 65 MXI-347PC art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. Therapeutic compositions can be administered with medical devices known in the art. For example, in a preferred embodiment, a therapeutic composition of the 5 invention can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. Patent Nos. 5,399,163; 5,383,851; 5,312,335; 5,064,413; 4,941,880; 4,790,824; or 4,596,556. Examples of well-known implants and modules useful in the present invention include: U.S. Patent No. 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate; 10 U.S. Patent No. 4,486,194, which discloses a therapeutic device for administering medicants through the skin; U.S. Patent No. 4,447,233, which discloses a medication infusion pump for delivering medication at a precise infusion rate; U.S. Patent No. 4,447,224, which discloses a variable flow implantable infusion apparatus for continuous drug delivery; U.S. Patent No. 4,439,196, which discloses an osmotic drug 15 delivery system having multi-chamber compartments; and U.S. Patent No. 4,475,196, which discloses an osmotic drug delivery system. These patents are incorporated herein by reference. Many other such implants, delivery systems, and modules are known to those skilled in the art. In certain embodiments, the human monoclonal antibodies of the invention can 20 be formulated to ensure proper distribution in vivo. For example, the blood-brain barrier (BBB) excludes many highly hydrophilic compounds. To ensure that the therapeutic compounds of the invention cross the BBB (if desired), they can be formulated, for example, in liposomes. For methods of manufacturing liposomes, see, e.g., U.S. Patents 4,522,811; 5,374,548; and 5,399,331. The liposomes may comprise one or more 25 moieties which are selectively transported into specific cells or organs, thus enhance targeted drug delivery (see, e.g., V.V. Ranade (1989) J. Clin. Pharmacol. 29:685). Exemplary targeting moieties include folate or biotin (see, e.g., U.S. Patent 5,416,016 to Low et al.); mannosides (Umezawa et al., (1988) Biochem. Biophys. Res. Commun. 153:1038); antibodies (P.G. Bloeman et al. (1995) FEBS Lett. 357:140; M. Owais et al. 30 (1995) Antimicrob. Agents Chemother. 39:180); surfactant protein A receptor (Briscoe et al. (1995) Am. J. Physiol. 1233:134); p 12 0 (Schreier et al. (1994) J Biol. Chem. 269:9090); see also K. Keinanen; M.L. Laukkanen (1994) FEBS Lett. 346:123; J.J. Killion; I.J. Fidler (1994) Immunomethods 4:273. 66 MXI-347PC Uses and Methods The antibodies, particularly the human antibodies, antibody compositions and methods of the present invention have numerous in vitro and in vivo diagnostic and therapeutic utilities involving the diagnosis and treatment of Glypican-3 mediated 5 disorders. For example, these molecules can be administered to cells in culture, in vitro or ex vivo, or to human subjects, e.g., in vivo, to treat, prevent and to diagnose a variety of disorders. As used herein, the term "subject" is intended to include human and non human animals. Non-human animals include all vertebrates, e.g., mammals and non mammals, such as non-human primates, sheep, dogs, cats, cows, horses, chickens, 10 amphibians, and reptiles. Preferred subjects include human patients having disorders mediated by Glypican-3 activity. The methods are particularly suitable for treating human patients having a disorder associated with aberrant Glypican-3 expression. When antibodies to Glypican-3 are administered together with another agent, the two can be administered in either order or simultaneously. 15 Given the specific binding of the antibodies of the invention for Glypican-3, the antibodies of the invention can be used to specifically detect Glypican-3 expression on the surface of cells and, moreover, can be used to purify Glypican-3 via immunoaffinity purification. Furthermore, given the expression of Glypican-3 on various liver tumor cells, the 20 human antibodies, antibody compositions and methods of the present invention can be used to treat a subject with a liver tumorigenic disorder, e.g., a disorder characterized by the presence of tumor cells expressing Glypican-3 including, for example, HCC cells. In one embodiment, the antibodies (e.g., human monoclonal antibodies, multispecific and bispecific molecules and compositions) of the invention can be used to 25 detect levels of Glypican-3, or levels of cells which contain Glypican-3 on their membrane surface, which levels can then be linked to certain disease symptoms. Alternatively, the antibodies can be used to inhibit or block Glypican-3 function which, in turn, can be linked to the prevention or amelioration of certain disease symptoms, thereby implicating Glypican-3 as a mediator of the disease. This can be achieved by 30 contacting a sample and a control sample with the anti- Glypican-3 antibody under conditions that allow for the formation of a complex between the antibody and Glypican-3. Any complexes formed between the antibody and Glypican-3 are detected and compared in the sample and the control. 67 MXI-347PC In another embodiment, the antibodies (e.g., human antibodies, multispecific and bispecific molecules and compositions) of the invention can be initially tested for binding activity associated with therapeutic or diagnostic use in vitro. For example, compositions of the invention can be tested using the flow cytometric assays described 5 in the Examples below. The antibodies (e.g., human antibodies, multispecific and bispecific molecules, immunoconjugates and compositions) of the invention have additional utility in therapy and diagnosis of Glypican-3-related diseases. For example, the human monoclonal antibodies, the multispecific or bispecific molecules and the immunoconjugates can be 10 used to elicit in vivo or in vitro one or more of the following biological activities: to inhibit the growth of and/or kill a cell expressing Glypican-3; to mediate phagocytosis or ADCC of a cell expressing Glypican-3 in the presence of human effector cells, or to block a Glypican-3 ligand from binding to Glypican-3. In a particular embodiment, the antibodies (e.g., human antibodies, multispecific 15 and bispecific molecules and compositions) are used in vivo to treat, prevent or diagnose a variety of Glypican-3-related diseases. Examples of Glypican-3-related diseases include, among others, HCC and other liver cancers. Suitable routes of administering the antibody compositions (e.g., human monoclonal antibodies, multispecific and bispecific molecules and immunoconjugates) 20 of the invention in vivo and in vitro are well known in the art and can be selected by those of ordinary skill. For example, the antibody compositions can be administered by injection (e.g., intravenous or subcutaneous). Suitable dosages of the molecules used will depend on the age and weight of the subject and the concentration and/or formulation of the antibody composition. 25 As previously described, human anti-Glypican-3 antibodies of the invention can be co-administered with one or other more therapeutic agents, e.g., a cytotoxic agent, a radiotoxic agent or an immunosuppressive agent. The antibody can be linked to the agent (as an immunocomplex) or can be administered separate from the agent. In the latter case (separate administration), the antibody can be administered before, after or 30 concurrently with the agent or can be co-administered with other known therapies, e.g., an anti-cancer therapy, e.g., radiation. Such therapeutic agents include, among others, anti-neoplastic agents such as doxorubicin (adriamycin), cisplatin bleomycin sulfate, carmustine, chlorambucil, and cyclophosphamide hydroxyurea which, by themselves, 68 MXI-347PC are only effective at levels which are toxic or subtoxic to a patient. Cisplatin is intravenously administered as a 100 mg/kg dose once every four weeks and adriamycin is intravenously administered as a 60-75 mg/ml dose once every 21 days. Co administration of the human anti-Glypican-3 antibodies, or antigen binding fragments 5 thereof, of the present invention with chemotherapeutic agents provides two anti-cancer agents which operate via different mechanisms which yield a cytotoxic effect to human tumor cells. Such co-administration can solve problems due to development of resistance to drugs or a change in the antigenicity of the tumor cells which would render them unreactive with the antibody. 10 Target-specific effector cells, e.g., effector cells linked to compositions (e.g., human antibodies, multispecific and bispecific molecules) of the invention can also be used as therapeutic agents. Effector cells for targeting can be human leukocytes such as macrophages, neutrophils or monocytes. Other cells include eosinophils, natural killer cells and other IgG- or IgA-receptor bearing cells. If desired, effector cells can be 15 obtained from the subject to be treated. The target-specific effector cells can be administered as a suspension of cells in a physiologically acceptable solution. The number of cells administered can be in the order of 108-109 but will vary depending on the therapeutic purpose. In general, the amount will be sufficient to obtain localization at the target cell, e.g., a tumor cell expressing Glypican-3, and to affect cell killing by, 20 e.g., phagocytosis. Routes of administration can also vary. Therapy with target-specific effector cells can be performed in conjunction with other techniques for removal of targeted cells. For example, anti-tumor therapy using the compositions (e.g., human antibodies, multispecific and bispecific molecules) of the invention and/or effector cells armed with these compositions can be used in conjunction 25 with chemotherapy. Additionally, combination immunotherapy may be used to direct two distinct cytotoxic effector populations toward tumor cell rejection. For example, anti-Glypican-3 antibodies linked to anti-Fe-gamma RI or anti-CD3 may be used in conjunction with IgG- or IgA-receptor specific binding agents. Bispecific and multispecific molecules of the invention can also be used to 30 modulate FcyR or FcyR levels on effector cells, such as by capping and elimination of receptors on the cell surface. Mixtures of anti-Fe receptors can also be used for this purpose. 69 MXI-347PC The compositions (e.g., human, humanized, or chimeric antibodies, multispecific and bispecific molecules and immunoconjugates) of the invention which have complement binding sites, such as portions from IgG1, -2, or -3 or IgM which bind complement, can also be used in the presence of complement. In one embodiment, ex 5 vivo treatment of a population of cells comprising target cells with a binding agent of the invention and appropriate effector cells can be supplemented by the addition of complement or serum containing complement. Phagocytosis of target cells coated with a binding agent of the invention can be improved by binding of complement proteins. In another embodiment target cells coated with the compositions (e.g., human antibodies, 10 multispecific and bispecific molecules) of the invention can also be lysed by complement. In yet another embodiment, the compositions of the invention do not activate complement. The compositions (e.g., human, humanized, or chimeric antibodies, multispecific and bispecific molecules and immunoconjugates) of the invention can also be 15 administered together with complement. In certain embodiments, the instant disclosure provides compositions comprising human antibodies, multispecific or bispecific molecules and serum or complement. These compositions can be advantageous when the complement is located in close proximity to the human antibodies, multispecific or bispecific molecules. Alternatively, the human antibodies, multispecific or bispecific 20 molecules of the invention and the complement or serum can be administered separately. Also within the scope of the present invention are kits comprising the antibody compositions of the invention (e.g., human antibodies, bispecific or multispecific molecules, or immunoconjugates) and instructions for use. The kit can further contain one ore more additional reagents, such as an immunosuppressive reagent, a cytotoxic 25 agent or a radiotoxic agent, or one or more additional human antibodies of the invention (e.g., a human antibody having a complementary activity which binds to an epitope in the Glypican-3 antigen distinct from the first human antibody). Accordingly, patients treated with antibody compositions of the invention can be additionally administered (prior to, simultaneously with, or following administration of a 30 human antibody of the invention) with another therapeutic agent, such as a cytotoxic or radiotoxic agent, which enhances or augments the therapeutic effect of the human antibodies. 70 MXI-347PC In other embodiments, the subject can be additionally treated with an agent that modulates, e.g., enhances or inhibits, the expression or activity of Fey or Fey receptors by, for example, treating the subject with a cytokine. Preferred cytokines for administration during treatment with the multispecific molecule include of granulocyte 5 colony-stimulating factor (G-CSF), granulocyte- macrophage colony-stimulating factor (GM-CSF), interferon- y (IFN-y), and tumor necrosis factor (TNF). The compositions (e.g., human antibodies, multispecific and bispecific molecules) of the invention can also be used to target cells expressing FcyR or Glypican 3, for example for labeling such cells. For such use, the binding agent can be linked to a 10 molecule that can be detected. Thus, the invention provides methods for localizing ex vivo or in vitro cells expressing Fe receptors, such as FcyR, or Glypican-3. The detectable label can be, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. In a particular embodiment, the invention provides methods for detecting the 15 presence of Glypican-3 antigen in a sample, or measuring the amount of Glypican-3 antigen, comprising contacting the sample, and a control sample, with a human monoclonal antibody, or an antigen binding portion thereof, which specifically binds to Glypican-3, under conditions that allow for formation of a complex between the antibody or portion thereof and Glypican-3. The formation of a complex is then 20 detected, wherein a difference complex formation between the sample compared to the control sample is indicative the presence of Glypican-3 antigen in the sample. In other embodiments, the invention provides methods for treating a Glypican-3 mediated disorder in a subject, e.g., HCC and other liver cancers. In yet another embodiment, immunoconjugates of the invention can be used to 25 target compounds (e.g., therapeutic agents, labels, cytotoxins, radiotoxoins immunosuppressants, etc.) to cells which have Glypican-3 cell surface receptors by linking such compounds to the antibody. For example, an anti-Glypican-3 antibody can be conjugated to any of the toxin compounds described in US Patent Nos. 6, 281, 354 and 6,548,530, US patent publication Nos. 20030050331, 20030064984, 20030073852, 30 and 20040087497, or published in WO 03/022806. Thus, the invention also provides methods for localizing ex vivo or in vivo cells expressing Glypican-3 (e.g., with a detectable label, such as a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor). Alternatively, the immunoconjugates can be used to kill cells which 71 MXI-347PC have Glypican-3 cell surface receptors by targeting cytotoxins or radiotoxins to Glypican-3. The present invention is further illustrated by the following examples which should not be construed as further limiting. The contents of all figures and all 5 references, patents and published patent applications cited throughout this application are expressly incorporated herein by reference. EXAMPLES 10 Example 1: Generation of Human Monoclonal Antibodies Against Glypican-3 Antigen A recombinant fusion protein, Glypican-3 -his, was created that comprises the extracellular domain of the Glypican-3 protein linked to a non-Glypican-3 polypeptide, a histidine tag. Glypican-3-his was generated by standard recombinant methods and used 15 as an antigen for immunization. In addition, CHO cells transfected with a Glypican-3 Mye expression construct and a cell line, Hep-G2, were used as antigens for immunization. Transgenic HuMAb Mouse® and KM Mouse® Fully human monoclonal antibodies to Glypican-3 were prepared using an 20 HCo 17 strain of the transgenic HuMAb Mouse* and the KM strain of transgenic transchromosomic mice, each of which express human antibody genes. The HCo17 strain was constructed as described in WO/2005/058815, which is incorporated herein by reference in its entirety. The KM strain was constructed as described in WO 02/43478, which is incorporated herein by reference in its entirety. 25 HuMab Mouse and KM Mouse Immunizations To generate fully human monoclonal antibodies to Glypican-3, mice of the HuMAb Mouse* and KM Mouse* were immunized with Glypican-3-his protein expressed in CHO-S cells, CHO cells expressing the Glypicn-3-Myc protein, and Hep G2 cells. General immunization schemes for the HuMAb Mouse® are described in 30 Lonberg, N. et al (1994) Nature 368(6474): 856-859; Fishwild, D. et al. (1996) Nature Biotechnology 14: 845-851 and PCT Publication WO 98/24884. The mice were 6-16 weeks of age upon the first infusion of antigen. A purified recombinant preparation (5 72 MXI-347PC 25 tg) of Glypican-3 fusion protein or Glypican-3 expression cells (1x10 7 cells) were used to immunize each HuMab mouse and KM mouse Transgenic mice were immunized with antigen in complete Freund's adjuvant or Ribi adjuvant either intraperitonealy (IP), subcutaneously (Sc) or via footpad (FP), 5 followed by 3-21 days IP, Sc or FP immunization (up to a total of 12 immunizations) with the antigen in incomplete Freund's or Ribi adjuvant. The immune response was monitored by retroorbital bleeds. The plasma was screened by ELISA (as described below) and FACS, and mice with sufficient titers of anti-Glypican-3 human immunoglobulin were used for fusions. Mice were boosted intravenously with antigen 3 10 and 2 days before sacrifice and removal of the spleen. As is typical, between ten and 35 fusions for each antigen were performed. Several dozen mice were immunized for each antigen. Selection of a HuMab Mouse® or KM Mouse® Producing Anti-Glypican-3 Antibodies 15 To select a HuMab Mouse® or a KM Mouse* producing antibodies that bound Glypican-3, sera from immunized mice was tested by ELISA as described by Fishwild, D. et al. (1996)(supra). Briefly, microtiter plates were coated with purified recombinant Glypican-3 at 1-2 tg /ml in PBS, 50 pl/wells incubated 4'C overnight then blocked with 200 pl/well of 5% chicken serum in PBS/Tween (0.05%). Dilutions of plasma from 20 Glypican-3-immunized mice were added to each well and incubated for 1-2 hours at ambient temperature. The plates were washed with PBS/Tween and then incubated with a goat-anti-human IgG Fc polyclonal antibody conjugated with horseradish peroxidase (HRP) for 1 hour at room temperature. After washing, the plates were developed with ABTS substrate (Sigma, A- 1888, 0.22 mg/ml) and analyzed by spectrophotometer at OD 25 415-495. Sera from immunized mice were then further screened by flow cytometry for binding to a cell line expressing recombinant human Glypican-3, but not to a control cell line that does not express Glypican-3. Briefly, the binding of anti-Glypican-3 antibodies was assessed by incubating Glypican-3-expressing CHO cells with the anti-Glypican-3 30 antibody at a 1:20 dilution. The cells were washed and binding was detected with a FITC-labeled anti-human IgG Ab. Flow cytometric analyses were performed using a FACScan flow cytometry (Becton Dickinson, San Jose, CA). Mice that developed the highest titers of anti-Glypican-3 antibodies were used for fusions. Fusions were 73 MXI-347PC performed as described below and hybridoma supernatants were tested for anti Glypican-3 activity by ELISA and FACS. Generation of Hybridomas Producing Human Monoclonal Antibodies to Glypican-3: 5 The mouse splenocytes, isolated from a HuMab mouse or a KM mouse", were fused using electric field based electrofusion using a Cyto Pulse large chamber cull fusion electroporator (Cyto Pulse Sciences, Inc., Glen Burnie, MD). The resulting hybridomas were then screened for the production of antigen-specific antibodies. Single cell suspensions of splenic lymphocytes from immunized mice were fused with SP2/0 10 nonsecreting mouse myeloma cells (ATCC, CRL 1581) using electric field based electrofusion using a Cyto Pulse large chamber cull fusion electroporator (Cyto Pulse Sciences, Inc., Glen Burnie, MD). Cells were plated at approximately 1x10 4 cells/well in flat bottom microtiter plates, followed by about two week incubation in selective medium containing 10% fetal bovine serum, 10% P388D1 (ATCC, CRL TIB-63) 15 conditioned medium, 3-5% origen (IGEN) in DMEM (Mediatech, CRL 10013, with high glucose, L-glutamine and sodium pyruvate) plus 5 mM HEPES, 0.055 mM 2 mercaptoethanol, 50 mg/ml gentamycin and Ix HAT (Sigma, CRL P-7185). After 1-2 weeks, cells were cultured in medium in which the HAT was replaced with HT. Individual wells were then screened by ELISA and FACS (described above) for human 20 anti-Glypican-3 monoclonal IgG antibodies. Once extensive hybridoma growth occurred, medium was monitored usually after 10-14 days. The antibody secreting hybridomas were replated, screened again and, if still positive for human IgG, anti Glypican-3 monoclonal antibodies were subcloned at least twice by limiting dilution. The stable subclones were then cultured in vitro to generate small amounts of antibody 25 in tissue culture medium for further characterization. Hybridoma clones 4A6, 1 1E7, AND 16D10, generated from a HuMAb Mouse were selected for further analysis. 30 74 MXI-347PC Example 2: Structural Characterization of Human Monoclonal Antibodies 4A6, 1 1E7, and 16D10 The cDNA sequences encoding the heavy and light chain variable regions of the 4A6, 1 1E7, and 16D10 monoclonal antibodies were obtained from the 4A6, 1 1E7, and 5 16D10 hybridomas, respectively, using standard PCR techniques and were sequenced using standard DNA sequencing techniques. The nucleotide and amino acid sequences of the heavy chain variable region of 4A6 are shown in Figure 1A and in SEQ ID NO:25 and 19, respectively. The nucleotide and amino acid sequences of the light chain variable region of 10 4A6 are shown in Figure lB and in SEQ ID NO:28 and 22, respectively. Comparison of the 4A6 heavy chain immunoglobulin sequence to the known human germline immunoglobulin heavy chain sequences demonstrated that the 4A6 heavy chain utilizes a VH segment from human germline VH 5-51 and a JH segment from human germline JH4b. The alignment of the 4A6 VH sequence to the germline VH 5-51 15 sequence is shown in Figure 4. Further analysis of the 4A6 VH sequence using the Kabat system of CDR region determination led to the delineation of the heavy chain CDR1, CDR2 and CD3 regions as shown in Figures 1A and 4, and in SEQ ID NOs:1, 4 and 7, respectively. Comparison of the 4A6 light chain immunoglobulin sequence to the known 20 human germline immunoglobulin light chain sequences demonstrated that the 4A6 light chain utilizes a VL segment from human germline VK A27 and a JK segment from human germline JK4. The alignment of the 4A6 VL sequence to the germline VK A27 sequence is shown in Figure 7. Further analysis of the 4A6 VL sequence using the Kabat system of CDR region determination led to the delineation of the light chain 25 CDR1, CDR2 and CD3 regions as shown in Figures lB and 7, and in SEQ ID NOs:10, 13, and 16, respectively. The nucleotide and amino acid sequences of the heavy chain variable region of 1 1E7 are shown in Figure 2A and in SEQ ID NO:26 and 20, respectively. The nucleotide and amino acid sequences of the light chain variable region of 30 11E7 are shown in Figure 2B and in SEQ ID NO:29 and 23, respectively. Comparison of the 1 1E7 heavy chain immunoglobulin sequence to the known human germline immunoglobulin heavy chain sequences demonstrated that the 1 1E7 heavy chain utilizes a VH segment from human germline VH 5-51 and a JH segment from 75 MXI-347PC human germline JH4b. The alignment of the 11 E7 VH sequence to the germline VH 5-51 sequence is shown in Figure 5. Further analysis of the 1 1E7 VH sequence using the Kabat system of CDR region determination led to the delineation of the heavy chain CDR1, CDR2 and CD3 regions as shown in Figures 2A and 5, and in SEQ ID NOs:2, 5, 5 and 8, respectively. Comparison of the 1 1E7 light chain immunoglobulin sequence to the known human germline immunoglobulin light chain sequences demonstrated that the 1 1E7 light chain utilizes a VL segment from human germline VK A27 and a JK segment from human germline JK4. The alignment of the 11E7 VL sequence to the germline VK A27 10 sequence is shown in Figure 8. Further analysis of the 1 1E7 VL sequence using the Kabat system of CDR region determination led to the delineation of the light chain CDR1, CDR2 and CD3 regions as shown in Figures 2B and 8, and in SEQ ID NOs: 11, 14, and 17, respectively. The nucleotide and amino acid sequences of the heavy chain variable region of 15 16D10 are shown in Figure 3A and in SEQ ID NO:27 and 21, respectively. The nucleotide and amino acid sequences of the light chain variable region of 16D10 are shown in Figure 3B and in SEQ ID NO:30 and 24, respectively. Comparison of the 16D 10 heavy chain immunoglobulin sequence to the known human germline immunoglobulin heavy chain sequences demonstrated that the 16D 10 20 heavy chain utilizes a VH segment from human germline VH 5-51 and a JH segment from human germline JH4b. The alignment of the 16D10 VH sequence to the germline VH 5 51 sequence is shown in Figure 6. Further analysis of the 16D 10 VH sequence using the Kabat system of CDR region determination led to the delineation of the heavy chain CDR1, CDR2 and CD3 regions as shown in Figures 3A and 6, and in SEQ ID NOs:3, 6, 25 and 9, respectively. Comparison of the 16D 10 light chain immunoglobulin sequence to the known human germline immunoglobulin light chain sequences demonstrated that the 16D 10 light chain utilizes a VL segment from human germline VK A27 and a JK segment from human germline JKl. The alignment of the 16D10 VL sequence to the germline VK 30 A27 sequence is shown in Figure 9. Further analysis of the 16D10 VL sequence using the Kabat system of CDR region determination led to the delineation of the light chain CDR1, CDR2 and CD3 regions as shown in Figures 3B and 9, and in SEQ ID NOs:12, 15, and 18, respectively. 76 MXI-347PC Example 3: Characterization of Binding Specificity and Binding Kinetics of Anti Glypican-3 Human Monoclonal Antibodies In this example, binding affinity, kinetics and specificity of anti-Glypican-3 5 antibodies were examined by Biacore analysis and flow cytometry. Binding affinity and kinetics Anti-Glypican-3 antibodies were characterized for affinities and binding kinetics by Biacore analysis (Biacore AB, Uppsala, Sweden). Glypican-3 was covalently linked to a CM5 chip (carboxymethyl dextran coated chip) via primary amines, using standard 10 amine coupling chemistry and kit provided by Biacore. Binding was measured by flowing a concentration series of the anti-Glypican-3 monoclonal antibodies in HBS-EP buffer (provided by Biacore AB) at 20, 10, 5, 2.5 and 1.25 pig/ml at a flow rate of 40 ptL/min. Background binding was accounted for by flowing a concentration series of IgGI across the same chip. The antigen-antibody association kinetics were followed for 15 3 minutes and the dissociation kinetics were followed for 10 minutes. The association and dissociation curves were fit to a 1:1 Langmuir binding model using BlAevaluation software (Biacore AB). The KD, kon and koff values that were determined are shown in Table 1. 20 Table 1: Biacore binding data for anti-Glypican-3 monoclonal antibodies Anti-Glypican-3 Affinity KD On rate kon Off rate koff antibody x 10- 9 (M) x 106 (1/Ms) x 10- 4 1/s 4A6 0.20 1.6 3.2 16D10 0.31 1.8 5.4 11E7 0.40 1.8 7.3 Binding affinity Measured by FACS Anti-Glypican-3 antibodies were shown to bind to cell-surface Glypican-3 proteins with a high affinity using a fluorescence activated cell sorter ("FACS") assay. 25 The HCC cell line Hep-3B was added at a density of 2 x 105 cells into each well of 96 well plate. The 4A6, 1 1E7, and 16D10 monoclonal antibodies were added at a starting concentration of 20ug/ml and serially diluting the antibody at a 1:3 dilution. The cells were washed and binding was detected with a FITC-labeled anti-human IgG Ab. Flow cytometric analyses were performed using a FACSCalibur flow cytometer (Becton 77 MXI-347PC Dickinson, San Jose, CA). The resulting binding affinities, plotted as the mean fluorescence intensity ("MFI"), is shown in Figure 10. The binding affinities of the antibodies bound the HCC cells in a concentration-dependent manner. The effective concentration ("EC50") for each antibody was determined as the concentration that 5 resulted in a 50% mean fluorescence intensity ("MFI"). In this example, the strongest binding affinity was observed with the 4A6 antibody, which showed a MFI of 0.83. The next best observed affinity was for 16D10, which had an EC50 of 1.39. The lowest observed affinity of the three antibodies was for 11E7, which had an EC50 of 1.98. Binding specificity Measured by FACS 10 Anti-Glypican-3 antibodies were shown to bind to cell-surface Glypican-3 proteins with a high specificity using a FACS assay. The HCC cell lines Hep-3B and Hep-G2 were grown in DMEM + 10% FBS at a density of 1 x 10s cells. The 4A6, 1 1E7, and 16D10 monoclonal antibodies were added at a concentration of 10 pig/ml. Additionally, three negative controls were analyzed for comparison. These negative 15 controls were (1) no stain at all, (2) the secondary antibody added alone, and (3) a hIgG1 isotype control antibody. The cells were washed and binding was detected with a FITC labeled anti-human IgG Ab. Flow cytometric analyses were performed using a FACSCalibur flow cytometer. Figure 11 shows that the anti-Glypican-3 antibodies bound the HCC cells with a high affinity. In contrast, the negative control samples did 20 not show any binding to the HCC cells. Chinese hamster ovary (CHO) cell lines that stably express Glypican-3 on the cell surface were developed and used to determine the specificity of the Glypican-3 monoclonal antibodies by flow cytometry. CHO cells were transfected with expression plasmids containing full length cDNA encoding Glypican-3. Binding of the three anti 25 Glypican-3 monoclonal antibodies was assessed by incubating the transfected cells with each of the Glypican-3 antibodies at a concentration of 10 pig/ml. The cells were washed and binding was detected with a phycoerythrin-labeled anti-human IgG Ab (BD Biosciences). Secondary antibody alone was used as a negative control. The results are depicted in Figure 12. The Glypican-3 monoclonal antibodies 4A6, 1 1E7, and 16D10 30 bound to the CHO cell line transfected with Glypican-3 but not to the parental CHO cell lines. These data demonstrate the specificity of the monoclonal antibodies for Glypican 3. 78 MXI-347PC Epitope binning of anti glypican-3 monoclonal antibodies by Biacore Epitope binning was performed by Biacore analysis to determine whether the anti-Glypican-3 antibodies bind to overlapping epitopes. Antibodies that have overlapping epitopes will compete for binding to Glypican-3 whereas those with distinct 5 epitopes will not compete and will simultaneously bind to the antigen. Purified 4A6 and 16D 10 were immobilized at 4000 RU on a CM5 chip using a standard amine coupling protocol. Glypican-3-his (50 nM) was pre-incubated for at least 1 hour with the 4A6, 11E7, or 16D10 monoclonal antibodies prior to injection. The antibody concentrations were a two-fold dilution series starting at 400 nM. Antibody-antigen mixtures were 10 injected at a flow rate 5 pl/min for 5 minutes. Each of the 4A6, 1 1E7, and 16D10 monoclonal antibodies were able to compete for binding to the immobilized 4A6 and 16D 10 antibodies in a concentration-dependent manner. This shows that 4A6, 16D 10, and 1 1E7 have overlapping epitopes. 15 Example 4: Glypican-3 Antibodies Bind to Liver Cancer Tissue Anti-Glypican-3 monoclonal antibody 4A6 was shown to bind human liver cancer tissues. Biopsies from liver cancer patients were obtained and the antibodies used for immunohistochemistry staining (Cytomyx, MA). 5 pim tissues cores were used. After drying for 30 minutes on slides, the tissue sections were fixed with acetone at 20 room temperature for 5 minutes. Slides were rinsed in PBS and then blocked with serum-free protein and peroxidase blocker (Dako S2001, CO) and subsequently incubated with primary antibody complex at 5 pig/ml for 45 minutes at room temperature. Next, the slides were washed and incubated for 30 minutes with FITC conjugated secondary antibody (Jackson Immunoresearch Lab, 109-097-003) and 25 washed again with PBS and incubated with polymer HRP conjugates (Dako, CO, K4063) for 20 minutes. Chromogen (Dako K3464) was used as a substrate, resulting in brown staining. Slides were mounted in Faramount Aqueous Mounting Media (Dako, S3025). 4A6 was shown to bind specifically to liver tumor cells. As exemplified in Figure 13, the 4A6 monoclonal antibody specifically stains cancerous liver tissue but not 30 the surrounding normal tissue. When stained with the monoclonal antibodies, other organs exhibit negative or non-specific staining, which include uterus, lung, liver, kidney colon, cervix, breast, bone marrow, adrenal glands, cerebellum, cerebrum, esophagus, heart, prostate, placenta, pituitary, ovary, pancreas, mesothelia, salivary 79 MXI-347PC gland, tonsil, skin, small intestine, skeletal muscle, stomach, spleen, testis, thymus, and thyroid. The data demonstrates that anti-Gpc3 HuMab 4A6 recognizes Gcp3 expressed in liver tumors. 5 Example 5: Anti-Glypican-3 Antibody Activity The monoclonal antibodies 4A6, 11 E7, and 16D 10 were shown to kill Gcp3 positive Hep-G2 cells in the presence of natural killer T-cells using an antibody dependent cellular cytotoxicity ("ADCC") assay. Human effector cells were prepared from whole blood as follows. Human 10 peripheral blood mononuclear cells were purified from heparinized whole blood by standard Ficoll-paque separation. The cells are resuspended in RPMI 1640 media containing 10% FBS 200U/ml of human IL-2 (PeproTech, NJ) and incubated overnight at 37C. The following day, the cells are collected and washed twice in RPMI + 1% of BSA (assay media) and resuspended at 1x10e6/ml. 15 100 pil of target cells at 1x10e4/well in flat bottom 96-well plate were incubated in assay media overnight at 37C. Following the target cells washed twice in assay media, 1 00ul of assay media was added into each well and incubated with 50 ul of the effector cells and 50ul of either anti-Gpc3 antibodies or human IgGI as isotype control at final concentration of 1Oug/ml. The Gpc3+ Hep-G2 cell line was tested for antibody specific 20 ADCC to anti-Gpc3 antibodies using Takara LDH cytotoxicity detection kit (Roche, 04744 926001, Switzerland), a fluorescence emission analysis as follows. The target cell line, Hep-G2 was incubated with effector cells at a target to effector ratio of 1:50. After 18 hour incubation at 3 7C, 1 00ul of the supernatants were collected and transferred to a new flat bottom 96-well plate. 1 00ul of Solution C was added to each 25 well and incubated at room temperature for 30 minutes. Absorbance of the samples at 490 nM was measured by SPECTRAMAX 340 PC (MTX Lab System, VA). The % lysis was determined by calculating the average absorbance of the triplicates and subtracting the background. As can be seen in Figure 14, the 4A6, 1 1E7, and 16D10 monoclonal antibodies 30 each cause specific lysis of the Hep-3b cells by natural killer T-cells as compared to the hIgGI isotype control antibody. 80 MXI-347PC Example 6: Anti-Glypican-3 Antibody Internalization The monoclonal antibodies 4A6, 1 1E7, and 16D10 were shown to be internalized by Hep-3b cells upon binding to the cells using a Hum-Zap assay. The Hum-ZAP assay showed internalization of the anti-Glypican-3 monoclonal antibodies through binding of 5 an anti-human IgG secondary antibody conjugated to the toxin saporin. (Advanced Targeting System, San Diego, CA, IT-22-100). First, 4A6, 1 1E7, and 16D10 were bound to the surface of the Hep-3B cells. Then, the Hum-ZAP antibodies were bound to the primary antibodies. Next, the primary antibody/Hum-ZAP complex was internalized by the cells. The entrance of Saporin into the cells resulted in protein synthesis 10 inhibition and eventual cell death. The Hum-ZAP assay was conducted as follows. Each of the cells was seeded at a density of 3x10 3 cells per well. The anti-Glypican-3 monoclonal antibodies or an isotype control human IgG were serially diluted then added to the cells. The Hum-ZAP was then added at a concentration of 2 pig/ml and the plates allowed to incubate for 96 15 hours. Cell viability in the plates was detected by CellTiter-Glo@ Luminescent Cell Viability Assay kit (Promega, G7571) and the plates were read at 490nM by a Luminomitor (Tuner BioSystems, Sunnyvale, CA). The data was analyzed by Prism (Graphpad). As can be seen in Figure 15, cell death was proportional to the concentration of 4A6, 1 1E7, and 16D10 monoclonal antibodies. Thus, the anti 20 Glypican-3 monoclonal antibodies were efficiently internalized by Hep-3b cells as compared to the hIgGI isotype control antibody. The monoclonal antibodies 4A6, 11 E7, and 16D 10 were also shown to be internalized by Hep-3b cells using an immunohistochemical staining assay. The cells, harvested with cell disassociation solution, were seeded at 104 cells per 100ul of 25 medium in each well of a 96-well plate and incubated with each monoclonal antibody at a concentration of 5ug/ml in FACS buffer (PBS + 5% FBS) for 30 minutes on ice. A human IgGI isotype control was used as a negative control. The cells were washed twice, re-suspended in medium (100ul per well) and then incubated with goat anti human secondary antibody conjugated with phycoerythrin (Jackson ImmunoResearch 30 Lab, PA) at a 1:100 dilution for 30 minutes. The cells were then washed with the media and either immediately imaged under a fluorescent microscope or incubated at 37' C for imaging at later time points. The images of cell morphology and immunofluorescence intensity of the stained cells were taken with a Nikon TE200 camera at 0, 30 or 60 81 MXI-347PC minutes as indicated in the Figure 16. The fluorescence was only observed in the cells stained with the 4A6, 1 1E7, and 16D10_antibodies. No fluorescence was detected with the IgGI control antibody. As can be seen in Figure 16, at 0 minutes after addition of the antibodies, the 5 cells are stained at the cell surface. After 30 minutes, the cells have begun internalizing the antibodies. After 60 minutes, the antibodies have been almost completely internalized by the cells. This shows that the human anti-Glypican-3 monoclonal antibodies are specifically internalized upon binding to Glypican-3-expressing HCC cells. 10 Example 7: Anti-Glypican-3 Antibody Stability Thermal stability The thermal stability of the anti-glypican-3 monoclonal antibodies was determined by calorimetric analysis of the melting temperatures for the 4A6, 1 1E7, and 15 16D 10 antibodies. Calorimetric measurements of melting temperatures (Tm) were performed on a VP-Capillary DSC differential scanning microcalorimeter platform that was combined with an autosampler (MicroCal LLC, Northampton, MA, USA). The sample cell volume was 0.144 mL. Denaturation data on the antibodies was obtained by heating the samples, at a concentration of 2.3 tM, from 30 to 95 0 C at a rate of 1 0 C/min 20 in phosphate-buffered saline (PBS) at pH 7.4. The same buffer was used in the reference cell to obtain the molar heat capacity by comparison. The observed thermograms were baseline corrected and normalized data analyzed based on a non-2 state model, using the software Origin v7.0. As shown in Table 2, 16D10 was observed to have the highest degree of thermal stability of the three anti-glypican-3 monoclonal 25 antibodies. Table 2. Differential scanning calorimetry: Anti-Glypican-3 Tml ( 0 C) Tm2 ( 0 C) Tm3 ( 0 C) 16D10 72.1 74.3 83.5 4A6 70.7 80.9 83.1 11E7 70.6 82.6 84.9 82 MXI-347PC Chemical Stability measured by fluorescence spectroscopy The stability of 4A6, 11 E7, and 16D 10 were compared by measuring the midpoint of chemical denaturation by fluorescence spectroscopy. Fluorescence measurements of chemical denaturation was performed on a SPEX Fluorolog 3.22 5 equipped with a Micromax plate reader (SPEX, Edison, NJ). The measurements were performed on antibody samples that had been equilibrated for 20 hours in 16 different concentrations of guanidinium hydrochloride in PBS buffer. The measurements were made in black, low volume, non-binding surface 384-well plates (Corning, Acton, MA) and required 1 piM of antibody in a well volume of 12 ptL. Fluorescence was excited at 10 280 nm and the emission spectra were measured between 320 and 400 nm. The scan speed was 1 second per nm and slits were set to a 5 nm bandpass. A buffer blank was performed using PBS and automatically subtracted from the data. Data was fitted to a two-state denaturation model using the GraphPad Prism software. As shown in Table 3, 16D 10 and 4A6 was observed to have similar stabilities but 11 E7 showed biphasic 15 unfolding. Table 3: Chemical denaturation determined by fluorescence spectroscopy Clone Unfolding Midpoint (M) 4A6 2.84 16D10 2.74 11 E7 biphasic All references cited in this specification, including without limitation all papers, publications, patents, patent applications, presentations, texts, reports, manuscripts, 20 brochures, books, internet postings, journal articles, periodicals, product fact sheets, and the like, one hereby incorporated by reference into this specification in their entireties. The discussion of the references herein is intended to merely summarize the assertions made by their authors and no admission is made that any reference constitutes prior art and Applicants' reserve the right to challenge the accuracy and pertinence of the cited 25 references. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit 30 or scope of the dependant claims. 83 MXI-347PC Table 4: Summary of sequence listing SEQ ID NO: SEQUENCE SEQ ID NO: SEQUENCE 1 VH CDR1 a.a. 4A6 25 VH n.t. 4A6 2 VH CDR1 a.a. 11E7 26 VH n.t. 11E7 3 VH CDR1 a.a. 16D10 27 VH n.t. 16D10 4 VH CDR2 a.a. 4A6 28 VK n.t. 4A6 5 VH CDR2 a.a. 11E7 29 VK n.t. 11E7 6 VH CDR2 a.a. 16D10 30 VK n.t. 16D10 7 VH CDR3 a.a. 4A6 31 VH 5-51 germline a.a. 8 VH CDR3 a.a. 1 1E7 32 JH JH4b germline a.a. 9 VH CDR3 a.a. 16D10 33 VK A27 germline a.a. 10 VK CDR1 a.a. 4A6 34 JK JK4 germline a.a. 11 VK CDR1 a.a. 11E7 35 JK JK1 germline a.a 12 VKCDR1 a.a. 16D10 36 Glypican-3 a.a. 13 VK CDR2 a.a. 4A6 14 VK CDR2 a.a. 11E7 15 VK CDR2 a.a. 16D10 16 VK CDR3 a.a. 4A6 17 VK CDR3 a.a. 11E7 18 VK CDR3 a.a. 16D10 19 VH a.a. 4A6 20 VH a.a. 11E7 21 VH a.a. 16D10 22 VK a.a. 4A6 23 VK a.a. 11E7 24 VK a.a. 16D10 The term "comprising" as used in this specification means "consisting at least in part of'. When interpreting each statement in this specification that includes the term 5 "comprising", features other than that or those prefaced by the term may also be present. Related terms such as "comprise" and "comprises" are to be interpreted in the same manner. In this specification where reference has been made to patent specifications, other 10 external documents, or other sources of information, this is generally for the purpose of providing a context for discussing the features of the invention. Unless specifically stated otherwise, reference to such external documents is not to be construed as an admission that such documents, or such sources of information, in any jurisdiction, are prior art, or form any part of the common general knowledge in the art. 15 84

Claims (15)

1. An isolated human monoclonal antibody, or an antigen-binding portion thereof, wherein the antibody binds to human Glypican-3 with a KD of 1x10-7 M or less. 5
2. The antibody, or antigen-binding portion thereof, of claim 1, wherein said antibody binds to human Glypican-3 with a KD of 5 x10- 9 M or less.
3. The antibody, or antigen-binding portion thereof, of claim 1, wherein human 10 Glypican-3 comprises a polypeptide having an amino acid sequence as set forth in SEQ ID NO:36 [Genbank Acc. No. NM_004484].
4. The antibody of claim 1, wherein the antibody is selected from the group consisting of an antibody comprising: 15 a) a heavy chain variable region CDR1 comprising SEQ ID NO:1; b) a heavy chain variable region CDR2 comprising SEQ ID NO:4; c) a heavy chain variable region CDR3 comprising SEQ ID NO:7; d) a light chain variable region CDR1 comprising SEQ ID NO:10; e) a light chain variable region CDR2 comprising SEQ ID NO:13; and 20 f) a light chain variable region CDR3 comprising SEQ ID NO:16; a) a heavy chain variable region CDR1 comprising SEQ ID NO:2; b) a heavy chain variable region CDR2 comprising SEQ ID NO:5; c) a heavy chain variable region CDR3 comprising SEQ ID NO:8; 25 d) a light chain variable region CDR1 comprising SEQ ID NO: 11; e) a light chain variable region CDR2 comprising SEQ ID NO:14; and f) a light chain variable region CDR3 comprising SEQ ID NO:17; and a) a heavy chain variable region CDR1 comprising SEQ ID NO:3; 30 b) a heavy chain variable region CDR2 comprising SEQ ID NO:6; c) a heavy chain variable region CDR3 comprising SEQ ID NO:9; d) a light chain variable region CDR1 comprising SEQ ID NO:12; e) a light chain variable region CDR2 comprising SEQ ID NO:15; and f) a light chain variable region CDR3 comprising SEQ ID NO: 18. 85 MXI-347PC
5. An isolated monoclonal antibody, or antigen binding portion thereof, selected from the group consisting of an antibody, or antigen binding portion thereof, comprising: 5 a) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 19; and/or a light chain variable region comprising the amino acid sequence of SEQ ID NO:22; b) a heavy chain variable region comprising the amino acid sequence of SEQ ID 10 NO:20 and/or a light chain variable region comprising the amino acid sequence of SEQ ID NO:23; and c) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO:21 and/or a light chain variable region comprising the amino acid sequence of SEQ 15 ID NO:24, wherein the antibody binds to human Glypican-3.
6. An isolated monoclonal antibody, or antigen binding portion thereof, which 20 competes for binding to Glypican-3 with the antibody of claim 4.
7. The antibody, or antigen-binding portion thereof, of any one of claims 4, 5 or 6, wherein the antibody, or antigen-binding portion is selected from the group consisting of a human antibody, a chimeric antibody, and a humanized antibody. 25
8. An immunoconjugate comprising the antibody, or antigen-binding portion thereof, of any one of the preceding claims, linked to a therapeutic agent.
9. A composition comprising the antibody, or antigen binding portion thereof, of 30 any one of claims 1, 4 or 5 or the immunoconjugate of claim 8 and a pharmaceutically acceptable carrier. 86 MXI-347PC
10. An isolated nucleic acid molecule selected from the group consisting of SEQ ID NOS:25, 26, 27, 28, 29 and 30.
11. An expression vector comprising the nucleic acid molecule of claim 10. 5
12. A host cell comprising the expression vector of claim 11.
13. A method of inhibiting growth of tumor cells expressing Glypican-3, comprising contacting the cells with the antibody, or antigen-binding portion thereof, of any one of 10 claims 1, 4 or 5, in an amount effective to inhibit growth of the tumor cells.
14. The method of claim 13, wherein the tumor cells are derived from liver tissue.
15. The method of claim 13, wherein the antibody, or antigen-binding portion 15 thereof, is an immunoconjugate. 87
AU2013257387A 2007-07-17 2013-11-11 Monoclonal antibodies against glypican-3 Abandoned AU2013257387A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013257387A AU2013257387A1 (en) 2007-07-17 2013-11-11 Monoclonal antibodies against glypican-3

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/959,845 2007-07-17
AU2008275985A AU2008275985B2 (en) 2007-07-17 2008-07-17 Monoclonal antibodies against Glypican-3
AU2013257387A AU2013257387A1 (en) 2007-07-17 2013-11-11 Monoclonal antibodies against glypican-3

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2008275985A Division AU2008275985B2 (en) 2007-07-17 2008-07-17 Monoclonal antibodies against Glypican-3

Publications (1)

Publication Number Publication Date
AU2013257387A1 true AU2013257387A1 (en) 2013-12-19

Family

ID=49759714

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013257387A Abandoned AU2013257387A1 (en) 2007-07-17 2013-11-11 Monoclonal antibodies against glypican-3

Country Status (1)

Country Link
AU (1) AU2013257387A1 (en)

Similar Documents

Publication Publication Date Title
US9217033B2 (en) Monoclonal antibodies against Glypican-3
US20190315881A1 (en) Human monoclonal antibodies to fucosyl-gm1 and methods for using anti-fucosyl-gm1 antibodies
AU2006321841B2 (en) Human monoclonal antibodies to protein tyrosine kinase 7 ( PTK7 ) and their use
AU2006294663B2 (en) Human monoclonal antibodies to CD70
AU2012241084B2 (en) Human monoclonal antibodies to fucosyl-GM1 and methods for using anti-fucosyl-GM1
AU2013257387A1 (en) Monoclonal antibodies against glypican-3
AU2013202283A1 (en) Human monoclonal antibodies to fucosyl-GM1 and methods for using anti-fucosyl GM1

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted