AU775448B2 - Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers - Google Patents

Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers Download PDF

Info

Publication number
AU775448B2
AU775448B2 AU72432/00A AU7243200A AU775448B2 AU 775448 B2 AU775448 B2 AU 775448B2 AU 72432/00 A AU72432/00 A AU 72432/00A AU 7243200 A AU7243200 A AU 7243200A AU 775448 B2 AU775448 B2 AU 775448B2
Authority
AU
Australia
Prior art keywords
fragment
antigen
composition according
antigen binding
antibody
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.)
Ceased
Application number
AU72432/00A
Other versions
AU7243200A (en
Inventor
Michael D. Dan
Howard A. Kaplan
Pradip K. Maiti
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.)
Viventia Bio Inc
Original Assignee
Viventia Biotech Inc
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 AU33696/97A external-priority patent/AU725238B2/en
Application filed by Viventia Biotech Inc filed Critical Viventia Biotech Inc
Priority to AU72432/00A priority Critical patent/AU775448B2/en
Publication of AU7243200A publication Critical patent/AU7243200A/en
Assigned to VIVENTIA BIOTECH INC. reassignment VIVENTIA BIOTECH INC. Amend patent request/document other than specification (104) Assignors: NOVOPHARM BIOTECH INC.
Application granted granted Critical
Publication of AU775448B2 publication Critical patent/AU775448B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

1
AUSTRALIA
Patents Act 1990 DITVSIONAI. APPLICATION Regulation 3.2 Name of Applicant: Actual Inventor(s): Novopharm Biotech Inc.
DAN, Michael MAITI, Pradip and KAPLAN, Howard Address for Service: DAVIES COLLISON CAVE, Patent Attorneys, Level 3, 303 Coronation Drive, Milton, Queensland, 4064, Australia "Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers" Invention Title: Details of Parent Application No: 33696/97 The following statement is a full description of this invention, including the best method of performing it known to me/us: Q:\OpcrWpa\2363640 divisionai.355.c 20112/00 ANTIGEN BINDING FRAGMENTS THAT SPECIFICALLY DETECT CANCER CELLS, NUCLEOTIDES ENCODING THE FRAGMENTS, AND USE THEREOF FOR THE PROPHYLAXIS AND DETECTION OF CANCERS TECHNICAL FIELD This invention relates to antibodies specific to an antigen detected on neoplastic cells but not on normal cells. This antigen is termed herein the "C-antigen." The C-antigen is recognized by the human monoclonal antibody (Mab) termed "H I The invention encompasses a wide variety of antibodies, and functional derivatives thereof that retain the immunologic specificity of HI I and are termed herein The exemplary antibody, H 11. compositions comprising the H 1l, and hybridomas producing Hl 1 are included herein. The HI 1 V region polynucleotides and polypeptides encoded thereby and recombinant molecules containing these polynucleotides are also encompassed by the invention. Methods of use including therapeutic and diagnostic of the aC antibodies are also included in the invention.
BACKGROUND
ART
In spite of numerous advances in medical research, cancer remains the second leading cause of death in the United States. In the industrialized nations. roughly one in five persons will die of cancer. Traditional modes of clinical care, such as surgical resection, radiotherapy and chemotherapy, have a significant failure rate, especially for solid tumors. Failure occurs either because the initial tumor is unresponsive, or because of recurrence due to regrowth at the original site and/or metastases. Even in cancers such as breast cancer where the mortality rate has decreased, successful intervention relies on early detection of the cancerous cells. The etiology, diagnosis and ablation of cancer remain a 25 central focus for medical research and development.
Neoplasia resulting in benign tumors can usually be completely cured by removing the mass surgically. If a tumor becomes malignant, as manifested by invasion of surrounding tissue, it becomes much more difficult to eradicate. Once a malignant tumor metastasizes, it is much less likely to be eradicated.
The three major cancers, in terms of morbidity and mortality, are colon, breast and lung. New surgical procedures offer an increased survival rate for colon cancer. Improved screening methods increase the detection of breast cancer, allowing earlier, less aggressive tlIerapy. lNumerous studies have shown that early detection increases survival and treatment options. Lung cancer remains largely refractory to treatment.
Excluding basal cell carcinoma, there are over one million new cases of cancer per year in the United States alone, and cancer accounts for over one half million deaths per year in this country. In the world as a whole, the five most common cancers are those of lung, stomach, breast, colon/rectum, and uterine cervix, and the total number of new cases per year is over 6 million. Only about half the number of people who develop cancer die of it.
Melanoma is one of the human diseases for which there is an acute need of new therapeutic modalities. It is a particularly aggressive form of skin cancer, and occurs in increased frequency in individuals with regular unguarded sun exposure. In the early 15 disease phases. melanoma is characterized by proliferation at the dermal-epidermal junction, which soon invades adjacent tissue and metastasizes widely. Once it has metastasized, it is often impossible to extirpate and is consequently fatal. Worldwide, 70,000 patients are diagnosed with melanoma and it is responsible for 25,000 reported deaths each year. The American Cancer Society projects that by the year 2000. 1 out of every 75 Americans will be diagnosed with melanoma.
Neuroblastoma is a highly malignant tumor occurring during infancy and early childhood. Except for Wilm's tumor, it is the most common retroperitoneal tumor in children. This tumor metastasizes early, with widespread involvement of lymph nodes, liver, bone, lung, and marrow. While the primary tumor is resolvable by resection, the recurrence rate is high.
An estimated 178,100 new cases of lung cancer will be diagnosed in 1997, accounting for 13% of cancer diagnoses. An estimated 160,400 deaths due to lung cancer will occur in 1997 accounting for 29% of all cancer deaths. The one year survival rates for lung cancer have increased from 32% in 1973 to 41% in 1993, largely due to improvements in surgical techniques. The 5 year survival rate for all stages combined is only 14%. The survival rate is 48% for cases detected when the disease is still localized.
but only 15% of lung cancers are discovered that early.
Small cell lung cancer is the most malignant and fastest growing form of lung cancer and accounts for 20-25% of new cases of lung cancer. 60,000 cases will be diagnosed in the U.S. in 1996. The primary tumor is generally responsive to chemotherapy, but is followed by wide-spread metastasis. The median survival time at diagnosis is approximately 1 year, with a 5 year survival rate of 5-10%.
Breast cancer is one of the most common cancers and is the third leading cause of death from cancers in the United States with an annual incidence of about 180.200 new cases among women in the United States during 1997. About 1.400 new cases of breast cancer will be diagnosed in men in 1997. In industrialized nations, approximately one in eight women can expect to develop breast cancer. The overall mortality rate for breast cancer has remained unchanged since 1930. It has increased an average of 0.2% per year, but decreased in women under 65 years of age by an average of 0.3% per year.
15 Preliminary data suggest that breast cancer mortality may be beginning to decrease, probably as a result of increased diagnoses of localized cancer and carcinoma in situ. See Marchant (1994) Contemporary Management of Breast Disease II: Breast Cancer, in: Obstetrics and Gynecology Clinics of North America 21:555-560; and Colditz (1993) Cancer Suppl. 71:1480-1489. An estimated 44,190 deaths (43.900 women. 290 men) in 1997 will occur due to breast cancer. In women, it is the second major cause of cancer death after lung cancer. The five-year survival rate for localized breast cancer has increased from 72% in the 1940s to 97% today. If the cancer has spread regionally, however, the rate is 76%, and for women with distant metastases the rate is 20%. Survival after a diagnosis of breast cancer continues to decline beyond five years. Sixty-five percent of women diagnosed with breast cancer survive 10 years and 56% survive years.
Non-Hodgkin's B cell lymphomas are cancers of the immune system that are a expected to afflict approximately 225,000 patients in the United States in 1996. These cancers are diverse with respect to prognosis and treatment, and are generally classified into one of three grades. The median survival of the lowest grade is 6.6 years and the 4 higher grade cancers have much lower life expectancy. Virtually all non-Hodgkin's B cell lymphomas are incurable. New diagnoses of non-Hodgkins lymphomas have increased approximately 7% annually over the past decade, with 52.700 new diagnoses estimated for 1996. The increase is due in part to the increasing nrevalpnr-e of lymphomas i, the A patient population.
Colon and rectal cancer will account for an estimated 131,200 cases in 1997, including 94,100 of colon cancer and 37,100 of rectal cancer. Colorectal cancers account for about 9% of new cancer diagnoses. An estimated 54,900 deaths due to colorectal cancer will occur in 1997, accounting for about 10% of cancer deaths. Mortality rates for colorectal cancer have fallen 32% for women and 14% for men during the past 20 years.
reflecting decreasing incidence rates and increasing survival rates. However, the mortality rate in African American men continues to rise. The 1 and 5 year relative survival rates for patients with colon and rectal cancer are 82% and 61%, respectively. When colorectal cancers are detected in an early, localized stage, the 5 year survival rate is 91%; however, 15 only 37% of colorectal cancers are discovered at that stage. After the cancer has spread regionally to involve adjacent organs or lymph nodes, the rate drops to 63%. Survival :rates for persons with distant metastases is Survival continues to decline beyond years, and 50% survive 10 years.
In spite of the difficulties, effective cures using anticancer drugs (alone or in 20 combination with other treatments) have been devised for some formerly highly lethal cancers. Most notable among these are Hodgkin's lymphoma, testicular cancer, choriocarcinoma, and some leukemias and other cancers of childhood. For several of the i more common cancers, early diagnosis, appropriate surgery or local radiotherapy enables a large proportion of patients to recover.
25 Current methods of cancer treatment are relatively non-selective. Surgery removes the diseased tissue, radiotherapy shrinks solid tumors and chemotherapy kills rapidly dividing cells. Chemotherapy, in particular, results in numerous side effects, in some cases so severe to preclude the use of potentially effective drugs. Moreover, cancers often develop resistance to chemotherapeutic drugs.
I
Numerous efforts are being made to enhance the specificity of cancer therapy. For review, see Kohn and Liotta (1995) Cancer Res. 55:1856-1862. In particular, identification of cell surface antigens expressed exclusively or preferentially on certain tumors allows the formulation of more selective treatment strategi a, to these antigens have been used in immunothcrapy of several types of cancer.
The basic immunoglobulin (Ig) structural unit in vertebrate systems is composed of two identical light polypeptide chains (approximately 23 kDa), and two identical heavy chains (approximately 53 to 70 kDa). The four chains are joined by disulfide bonds in a configuration. At the base of the Y. the two H chains are bound by covalent disulfide linkages.
Figure 1 shows a schematic of an antibody structure. The L and H chains are each composed of a variable region at the N-terminus, and a constant region at the Cterminus. In the L chain, the V region (termed "VLJL") is composed of a V (VL) region connected through the joining region to the C region In the H chain, the V 15 region (VHDHJH) is composed of a variable (VH) region linked through a combination of S: the diversity (DH) region and the joining (JH) region to the C region The VLJL and VHDHJH regions of the L and H chains, respectively, are associated at the tips of the Y to form the antigen binding portion and determine antigen binding specificity.
The (CH) region defines the isotype, the class or subclass of antibody.
20 Antibodies of different isotypes differ significantly in their effector functions, such as the ability to activate complement, bind to specific receptors Fc receptors) present on a wide variety of cell types, cross mucosal and placental barriers, and form polymers of the basic four-chain IgG molecule.
Antibodies are categorized into "classes" according to the CH type utilized in the S 25 immunoglobulin molecule (IgM, IgG, IgD, IgE, or IgA). There are at least five types of CH genes (Cp, Cy, C8, Ce, and Ca), and some species have multiple CH subtypes Cy 1 Cy 2 Cy 3 and Cy 4 in humans). There are a total of nine CH genes in the haploid genome of humans, eight in mouse and rat, and several fewer in many other species. In contrast, there are normally only two types of L chain C regions kappa and lambda and only one of these C regions is present in a single L chain protein ~1 6 there is only one possible L chain C region for even' VLJL produced). Each H chain class can be associated with either of the L chain classes a Cly region can be present in the same antibody as either a K or L chain), although the C regions of the H and L chains within a particular class do not vary with antigen specificity an IgG antibody always has a Cy H chain C region regardless of the antigen specificity).
Each of the V, D, J, and C regions of the H and L chains are encoded by distinct genomic sequences. Antibody diversity is generated by recombination between the different and J1. gene segments in the H chain, and V, and JL gene segments in the L chain. The recombination of the different VH, DI, and genes is accomplished by DNA recombination during B cell differentiation. Briefly, the H chain sequence recombines first to generate a D 1 Jj complex, and then a second recombinatorial event produces a VFDHJii complex. A functional H chain is produced upon transcription followed by splicing of the RNA transcript. Production of a functional H chain triggers recombination in the L chain sequences to produce a rearranged VLJL region which in turn 15 forms a functional VLJLCL region, the functional L chain.
The value and potential of antibodies as diagnostic and therapeutic reagents has been long-recognized in the art. Unfortunately, the field has been hampered by the slow, tedious processes required to produce large quantities of an antibody of a desired specificity. The classical cell fusion techniques allowed for efficient production of Mabs by fusing the B cell producing the antibody with an immortalized cell line. The resulting cell line is a hybridoma cell line.
Antibodies and functional derivatives thereof have been used in a variety of clinical *o settings. For instance, digoxin-specific Fab antibody fragments were used to treat lifethreatening digitalis intoxication. Antibodies are becoming more routinely useful in 25 diagnostic techniques such as radioimmune diagnosis of colon cancers. Koda et al. (1995) Am. J. Gastroenterol. 90:1644. A number of uses of Mabs, previously thought to be untenable, have recently been put into practice. For instance, see Hall (1995) Science 279:915-916.
A number of autoantibodies (antibodies that recognize and bind to self antigens) are found in humans. Many of these are associated with particular diseases such as rheumatoid arthritis. systemic lupus erythematosus, myasthenia gravis. primary biliary cirrhosis. polymyositis. systemic vasculitis, idiopathic necrotizing and crescentic Sglomerulonephritis and amyotrophic lateral sclerosis. For review, see Shattner A AI I A:I A C S 6OO/ i Y I 7) llhInnizUil. LCII. It. J. J LIICI Uj 1a1LIL' UUL. A a lL (19O6/1987) immunw. an. 14:143-153. Oter autoantivouics arc naturally-occurrin.
Lutz and Wipp (1982) J. Immunol. 128:1965; and Guilbert et al. (1982) J. Immunol.
128:2779-2787. Recently. human autoantibodies to specific cancer antigens have been detected and. in some cases, are being produced by hybridoma technology. These antibodies have also been produced by active immunization. United States Patent No.
5.474.755. Originally, the human B cells were immortalized using Epstein-Barr Virus or mouse myelomas. For review, see Buck et al. (1984) "Monoclonal Antibodies" NY.
Plenum Press. More recent techniques have allowed immortalization without the use of this potentially harmful virus. See, U.S. Patent No. 4,618,477; and Glassy (1987) Cancer Res. 47:5181-5188. In most instances, the antibodies are specific for one, or in some instances, a few, cancer types. For instance, a Mab has been described that 15 specifically recognizes glioma cells but no other tumor or normal cells. These antibodies .i were used to image the glioma in the patient's brain. Fischer et al. (1991) hnmunobiol.
S.i Prot. Pep. VI Atassi, ed.) Plenum Press. NY. pp. 263-270. No antibody has been described that is capable of recognizing a wide range of tumors while failing to recognize, or only poorly recognize, normal, non-cancerous cells.
20 Recombinant genetic techniques have allowed cloning and expression of antibodies, functional fragments thereof and the antigens recognized. These engineered antibodies provide novel methods of production and treatment modalities. For instance, functional immunoglobulin fragments have been expressed in bacteria and transgenic tobacco seeds and plants. Skerra (1993) Curr. Opin. Immunol. 5:256-262; Fiedler and e*o 25 Conrad (1995) Bio/Technology 13:1090-1093; Zhang et al. (1993) Cancer Res. 55:3384- 3591; Ma et al. (1995) Science 268:916; and, for a review of synthetic antibodies, see Barbas (1995) Nature Med. 1:836-839.
SSeveral human Mabs against tumor associated antigens have been produced and characterized. The tumor associated antigens recognized by human Mabs include celi surface, cytoplasmic and nuclear antigens. Yoshikawa et al. (1989) Jpn. J. Cancer Res.
8 (Gann) 80:546-553: Yamaguchi et al. (1987) Proc. Natl. Acad. Sci. USA 84:2416-2420: Haspel et al. (1985) Cancer Res. 45:3951-3961; Cote et al. (1986) Proc. Nail. Acad. Sci.
USA 83:2959-2963: Glassy (1987) Cancer Res. 47:5181-5188; Borup-Christensen et al.
198 7) C7a. cr Dtec. e veint. Suppi. :207--2 15: Haspel ei ai. (i 985) Cancer Res.
45:3951-3961; Kan-Mitchell et al. (1989) Cancer Res. 49:4536-4541; Yoshikawa et al.
(1986) Jpn. J. Cancer Res. 77:1122-1133; and McKnight et al. (1990) Human Antibod Hybridomas 1:125-129.
Human Mabs have been used in cancer imaging. diagnosis and therapy. Olsson (1985) J. Nat. Cancer Inst. 75:397-404; Larrick and Bourla (1986) J. Biol. Resp. Mod.
5:379-393; McCabe et al. (1988) Cancer Res. 48:4348-4353; Research News (1993) Science 262:841; Ditzel et al. (1994) Cancer 73:858-863: and Alonso (1991) Am. J. Clin.
Oncol. 4:463-471. A recombinant single chain bispecific antibody has been reported that has high tumor cell toxicity. This molecule recognizes both the CD3 antigen of human T cells and EpCAM, which is associated with disseminated tumor cells in patients with 15 minimal residual colorectal cancer. Mack et al. (1995) Proc. Nail. Acad. Sci. USA 92:7021-7025.
Several murine monoclonal anti-GD2 antibodies were reported to suppress the growth of tumors of neuroectodermal origin in athymic (nu/nu) mice or cause remission in ,patients with metastatic melanoma. A human-mouse chimeric anti-GD2 antibody caused remission in patients with metastatic neuroblastoma. The mechanism of action of the antibodies is thought to involve antibody dependent cellular cytotoxicity (ADCC) or complement-mediated cytotoxicity (CMC). Clinical responses have been obtained by treating melanoma with Mabs against GM2, GD2 and GD3. Cheresh et al. (1985) Proc.
Natl. Acad. Sci. USA 82:5155-5159. Active immunization with a ganglioside vaccine 25 comprising GM2 produced anti-GM2 antibodies in 50/58 patients, who survived longer on average than patients without detectable anti-GM2 antibody.
Mabs to GD2 have also been found to react specifically with small cell lung carcinoma. Cheresh et al. (1986) Cancer Res. 46:5112-5118. Human Mabs specific for other cancers including lung, melanoma, stomach, squamous cell carcinoma, cervical carcinoma, and mammary carcinoma have also been produced. Murakami (1985) in Vitro Cell. Dev. Biol. 21:593; Schadendorf (1989) J Immunol. 142:1621-1625; Yoshikawa et al.
(1986) Jpn. J. Cancer Res. 77:1122-1133; Pickering and Misra (1984) Clin. Immunol.
Immunopathol. 32:253-260; Hagiwara and Sato (1983) Mol. Biol. Med. 1:245-252; and Schlom et al. (1980) Proc. Nail. Acad. Sci. USA 77:6841-6845. Human anti-cancer Mabs and the antigens they recognize have also been suggested for use in vaccines. See, e.g.
Finn et al. (1995) Immunol. Rev. 145:61-89. A human Mab to malignant brain tumors was used in a phase I clinical trial without adverse side effects. Matsumoto et al. (1994) The Clinical Report 28:118-126. Phase 11 trial results have been reported on combined treatment with murine Mab and colony stimulating factor in metastatic gastrointestinal cancer. Saleh ct al. (1995) Cancer Res. 55:4339-4346. A single chain immunotoxin has also been found to cure carcinomatous meningitis in a rat model. Pastan et al. (1995) Proc.
Nail. Acad. Sci. USA 92:2765-2769. Human Mabs that specifically recognize ovarian cancer cells have been shown to effectively image this cancer. Chaudhuri et al. (1994) Cancer 73: 1098-1104.
If there were a simple and reliable strategy for providing immune reactivity against an antigen common to these cancers rather than cancer-specific immunity, the clinical prospects of cancers in general would improve. All references cited herein are hereby incorporated by reference in their entirety.
DISCLOSURE OF THE INVENTION This invention encompasses compositions containing antigen binding fragments of an antibody where the antibody specifically recognizes the antigen recognized by an antibody comprising a H chain V region having the amino acid sequence of SEQ ID NO:2 .o and a L chain V region having the amino acid sequence of SEQ ID NO:4. Preferably, the antibody is H 11. The invention further encompasses antibodies comprising the H and L chain V regions of HI 1 (SEQ ID NOS:2 and 4, respectively). HI 1 specifically recognizes cancer cells from a wide variety of cancers but does not recognize normal, non-cancerous cells. By "does not recognize" is meant that noncancer cells are either not specifically bound to by HI 1 or are only poorly recognized by the antibody. The antibodies are designated aC and include HI 1 and any antibody with the "immunologic specificity" of
S
I
15 H1 I, that is. recognizing the antigen recognized by HI and that is specific for at least one type of cancer cell but does not recognize normal cells. These antigen binding fragments include, but are not limited to, whole native antibodies. exemplified by HIA; bipeci;ic r by -aI; isp e ci ic antibodies; chimeric antibodies; Fab, Fab'. single chain V region fragments (scFv) and fusion polypeptides.
The invention further encompasses HI 1 antibody fusion molecules comprising a polypeptide region with an antigenic, therapeutic, toxic or labeling molecule attached to the H chain C region. a single-chain VI--VL or V 1 -Vt V region, and polynucleotides encoding such polypeptides.
Also embodied in the invention are polypeptides having the immunologic specificity of H1 1, wherein the polypeptide comprises at least 5 consecutive amino acids from a V region of an uC antibody. The V region may be from a L chain or H chain. The consecutive amino acids preferably play a role in immunologic specificity, and may be from a CDR (Complementarity Determining Region of an antibody). Intact H I1, functionally active fragments of H 1, fusion proteins, chimeric antibodies, multiple antigen proteins, and other polypeptide derivatives of aC antibodies are included. Of special interest are single-chain V regions and fusion proteins.
The compounds and compositions of this invention may be used inter alia for detecting or treating a cancer; including therapy of such cancer, and prophylactic care, particularly for decreasing the risk of recurrence.
The invention further embodies cells and cell lines producing the aC antigen binding fragments.
Another embodiment of this invention is a polynucleotide comprising a sequence encoding a polypeptide with the immunologic specificity of H11, wherein the encoded polypeptide comprises at least 5 consecutive amino acids from a V region of HI 1. The V region may be from either the HI I L chain or H chain. The 5 consecutive amino acids preferably play a role in HI 1 immunologic reactivity, and may be from a CDR. The V region of HI 1 has been found to have a small region of homology to an antibody designated A6. Peptides comprised solely of this region of homology and lacking other
II
HI l-specific amino acid residues are specifically excluded from the claimed invention.
A6 is described in W0953574.
The invention also encompasses isolated polynucleotides of at least 20 consecutive nucleotides capable of forming a stable duplex with the HII L or H chain encoding sequences, but not with sequences for other previously described immunoglobulin molecules. Any of these polynucleotides may be in the form of cloning vectors, expression vectors, or transfected into host cells.
A further embodiment of this invention comprises prophylactic treatment of a cancer patient with at least one cC antigen binding fragment. Preferably, aC is fused to a therapeutic molecule to effect delivery of the therapeutic molecule to the cancer cell. The individual may have a clinically detectable tumor, or the tumor may have been previously treated and rendered undetectable. The method may be for palliating the disease, or for reducing the risk of recurrence.
A further embodiment of the invention is a kit for detection or quantitation of the 15 antigen recognized by aC (hereinafter, the "C-antigen") in a sample, comprising H 11 or a polypeptide of this invention in suitable packaging. Also embodied by the invention are :methods for detecting the C-antigen or cells expressing the C-antigen by employing a reagent or kit embodied in this invention.
*o S 20 BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 depicts a schematic of the general antibody structure.
Figure 2 depicts flow cytometric analysis of cells recognized by HI1.
.i *Figure 3 depicts flow cytometric analysis of cells recognized by H 1.
o *Figure 4 depicts flow cytometric analysis of cells recognized by HI 1. A is A-375 25 (melanoma), B is SKMG-1 (glioma), C is SK-BR-3 (breast adenocarcinoma), D is HT-29 (colon adenocarcinoma), E is MB-468 (breast carcinoma), and F is T47D (breast carcinoma).
SFigure 5 depicts binding of H 11 to tumor cell extracts. The light bars represent H 11 and the dark bars represent control antibody.
Figure 6 depicts binding of H 1 to tumor cell extracts.
12 Figure 7 depicts binding of H I to human tumor cell lines by cell-fixed ELISA.
The light bars are H 11 IgM and the dark bars are control IgM.
Figure 8 depicts a schematic of the expression vector pSJFI.
rigure 9 depicts ih; dueicriinaion of the antigenic similarities between Mab Hi 1 and H1 I-scFv by cell fixed ELISA. Reactivity was determined by rabbit antibody to HI 11 scFv.
Figure 10 depicts the relative fluorescence intensity of biotinylated HI 1-scFv (thick line) and BGA scFv (thin line) to lymphoma cells.
Figure 11 depicts the titration of the reactivity of biotinylated H 1 -scFv for the binding to lymphoma cells. RAMOS, Daudi. CA-46 and CCRF-CEM cells as determined by cell fixed ELISA. The antibody concentrations decrease from an initial 10 jg/mL (open bar) to 5 pg/mL. then 2.5 lg/mL and finally, 1.25 tg/mL (doubly cross-hatched line).
Figure 12 depicts the relative fluorescence intensity of H 1 -scFv and control scFv binding to tumor cell lines. A is A-375 (melanoma), B is SK-BR-3 (breast S 15 adenocarcinoma), C is HT-29 (colon adenocarcinoma). D is CA-46 (Burkitt's lymphoma), E is RAMOS (Burkitt's lymphoma), F is H9 (T cell lymphoma), and G is CCRF-CEM (acute lymphoblastoid lymphoma).
Figure 13 depicts the binding of 25 1I-HI 1-scFv to LSI74T cells.
Figure 14 depicts the binding of '"I-H11-scFv to A375.
Figure 15 depicts the mammalian expression vector pNB2 used to transfect and express recombinant H I -IgG.
Figure 16 depicts the mammalian expression vector pNB3 used to transfect and express recombinant H11 -IgG.
Figure 17 depicts the purification of I 5 I-H 11-scFv on P-2 minicolumn and analysis of 25 I-HI 1-scFv by paper chromatography in 85% methanol Figure 18 depicts the purification of 25 1-H 11 IgM on a Sephadex G-25 minicolumn and analysis of 25I-H 1 IgM by paper chromatography in 85% methanol i Figure 19 depicts the purification of'In-DTPA-Hll-scFv on Sephadex minicolumn and analysis of''In-DTPA-Hl 1-ScFv by ITLC-SG/0.1M citrate i 13 Figure 20 depicts in vivo binding of 'In-Hl 1-scFv to A375 xenograft tumors in nude mice.
BEST MODE FOR CARRYING OUT THE INVENTION This invention encompasses antigen binding fragments exemplified by a newly identified human Mab that recognizes specifically cancerous cells. This specificity extends only to cancer cells, and the antibody does not recognize non-cancerous cells. The exemplary antibody is designated HI 1 and the variable regions are encoded by SEQ ID NOS: I and 4 (SEQ ID NOS:3 and 6 being the complementary strands of I and 4, respectively) and recognizes the antigen designated "C-antigen." The specificity of HII includes, but is not limited to, glioblastoma, neuroblastoma. malignant melanoma, breast adenocarcinoma, lung adenocarcinoma, small cell lung carcinoma, colon adenocarcinoma and prostate adenocarcinoma.
As shown in the examples herein, HI 1 and HI l-scFv do not recognize non- S 15 cancerous cells from all normal tissues tested. H 11 and uC antigen binding fragments are .i therefore useful in palliating the clinical conditions related to a wide variety of cancers.
The invention comprises antigen binding fragments recognizing the antigen HI I is specific for (designated C antigen). The invention further comprises polypeptide derivatives of H 1 and methods for using these compositions in diagnosis. treatment, and manufacture of novel reagents. The invention further encompasses polynucleotides encoding aC, HI I and derivatives thereof. Methods of use thereof are also encompassed by the invention.
The invention further encompasses aC derivatives with immunologic specificity for the C-antigen. These derivatives comprise regions of the polypeptide sequence comprising part of the HI 1 VDJ junction. Also encompassed are regions spanning at least one, preferably 2, and more preferably 3 or more of the HI 1 CDR amino acid sequences.
The full sequences of the HI 1 L and H chain C regions have not been determined, but are expected to be identical or nearly identical to those of other human immunoglobulin molecules. Further, knowledge of the V region amino acid sequences allows subc!oning with any C region. Such subcloning techniques are well known in the art. The chimeric molecules produced by these cloning techniques are also encompassed by the invention.
Screening a commercial heptapeptide phage library with H I I 1gM and scFv anti body clones has shown a very strong consensus sequence at the N-tern-inus having the followving amino acid sequence: Phc-1-isArg-Tr-..5/TJr The results are shown in Table 1&gM H I I pannings sfFv H I1I pannings TABLE
I
Ml Phe-Hiis-ArgTyr-Ser-Leu-Pro M42 Phe-His-Arg-Tvr..Ser-Asp-Tyr M3 Phe-His-Arg-Tyr..Ser-Leu-Pro M4 Phc-His-Arg-Tyr..Ser..ProTh M7 Phe-HisArgTyrThrPro-Gly M8 Ph-i-r-y-e-e-r Ml 10 Phe-His-Arg-Tyr..Ser-Pr 0 jjT 1 S2 Ph -i-r-y-e-e -r Me-i-r-y-h-r-e The DNA sequences use multiple codons. indicating quite different phage origins.
For example. the Arg is coded by triplets CGx and AGx families. In addition. comparison of the H I I pentapeptide consensus with sequence databases showed homology to the S 100 family of Ca 2 binding proteins. The results are shown in Table 2.
I-
TABLE 2 H I pentapeptide Phe-His-Arg-Tyr-Ser/Thr S. griscus protein Phe-His-Arg-Tyr-Ser (amino acids 251-255) Peanut stunt virus Phe-His-Arg-Tyr-Ser (amino acids 540-544) Human calcyclin Phe-His-Lys-Tyr-Ser (amino acids 16-20) Cystic Fibrosis Ag Tyr-His-Lys-Tyr-Ser (amino acids 16-21) The consensus pentapeptide sequences described herein are encompassed by the present invention.
Certain compounds. compositions and methods described in this application relate generally to uC and derivatives thereof which are routinely generated by classical techniques of immunochemistry. This includes aC which has been coupled to another compound by chemical conjugation, or by mixing with an excipient or an adjuvant. The term antigen binding fragment includes any peptide that binds to the C antigen in a cancer cell-specific manner. Typically, these derivatives include such immunoglobulin fragments 10 as Fab. F(ab') 2 Fab', scFv (both monomers and polymeric forms) and isolated H and L chains. An antigen binding fragment retains the specificity of HI 1, although avidity and/or .affinity may be altered. Especially preferred is the H I-scFv described herein.
The antigen binding fragments (also termed "derivatives" herein) are typically generated by genetic engineering, although they may alternatively be obtained by other 1 5 methods and combinations of methods. This classification includes, but is not limited to, engineered peptide fragments and fusion peptides. Preferred compounds include polypeptide fragments of the H 1I CDRs, antibody fusion proteins comprising cytokine effector components, antibody fusion proteins comprising adjuvants or drugs, and singlechain V region proteins.
The invention further comprises polynucleotides encoding the HI 1 antibody V regions and derivatives thereof. These include isolated polynucleotide fragments, recombinant polynucleotides, and therapeutic plasmids and vectors, such as vaccinia vectors, comprising the polynucleotides. These polynucleotides are exemplified by SEQ ID NOS:1, 3, 4, 6, 13, 15, 16, and 18.
16 Pharmaceutical compositions and treatment modalities of this invention are suitable for eliciting an immune response against neoplasia. Human cancer patients, including, but not limited to, glioblastoma, melanoma. neuroblastoma, adenocarcinoma, glioma. soft tissue sarcoma, and various carcinomas (inciuding smaii ceil lung cancer) are especially appropriate subjects.
As H11 has been shown to recognize specifically a variety of carcinomas, it is particularly useful in diagnosis, imaging and treatment of carcinomas. Suitable carcinomas include any known in the field of oncology, including, but not limited to. astrocytoma, oligodendroglioma. ependymoma. mcdulloblastoma, primitive neural ectodermal tumor (PNET), pancreatic ductal adenocarcinoma, small and large cell lung adenocarcinomas.
squamous cell carcinoma, bronchoalveolarcarcinoma, epithelial adenocarcinoma, and liver metastases thereof, hepatoma, cholangiocarcinoma, breast tumors such as ductal and lobular adenocarcinoma, squamous and adenocarcinomas of the uterine cervix, uterine and ovarian epithelial carcinomas, prostatic adenocarcinomas, transitional squamous cell S 15 carcinoma of the bladder, B and T cell lymphomas (nodular and diffuse) plasmacytoma, acute and chronic leukemias, malignant melanoma, soft tissue sarcomas and leiomyosarcomas.
The subjects may have an advanced form of disease, in which case the treatment objective may include mitigation or reversal of disease progression, and amelioration of side effects. The subjects may have had a history of the condition, for which they have already been treated, in which case the objective will typically include a decrease or delay in the risk of recurrence.
Additionally, the antigen binding fragments of this invention can be used as diagnostic and imaging reagents. These applications are described in more detail in the sections that follow.
"H1 1" is an antibody obtained from the fusion of peripheral blood lymphocytes of S"a 64 year old male with a low grade glioma and fused to a human myeloma cell line to produce a hybridoma designated NBGM 1/H 11. The generation and characterization of HI i is described in Example 1. "aC" represents any antibody, or antigen binding fragment thereof, either monoclonal, polyclonal or derivative thereof that recognizes 17 specifically the C antigen and distinguishes between cancer and noncancer cells. tC includes H11.
"Immunologic activity" of aC refers to the ability to specifically bind C antigen.
Such binding may or may not elicit an immune response. A specific immune response may comprise antibody. B cells. T cells, and any combination thereof, and effector functions resulting therefrom. Included are the antibody-mediated functions ADCC and complement-mediated cytolysis (CDC). The T cell response includes T helper cell function, cytotoxic T cell function, inflammation/inducer T cell function, and T cell mediated suppression. A compound able to elicit a specific immune response according to any of these criteria is referred to as "immunogenic." aC "activity" or "function" refers to any of the immunologic activities of aC, or to any other biological activity ascribed to HI in this disclosure, including the role of HI 1 in the detection, amelioration or palliation of cancer.
The "V region" of HI 1 refers to the V region of the HI I L chain or the V region of the H 11 H chain, either alone or in combination. These V regions are depicted in SEQ ID NOS: 2 and 5; the DNA encoding these regions is depicted in SEQ ID NOS: 1 and 4, i* respectively.
9* GM-CSF. IL-2. and other biologically active molecules referred to herein are meant to include fragments and derivatives based on the respective parent molecule that have the same biologic or physiologic function.
The terms "polypeptide", "peptide" and "protein" are used interchangeably herein to refer to polymers of amino acid residues of any length. The polymer may be linear or branched, it may comprise modified amino acids or amino acid analogs, and it may be interrupted by chemical moieties other than amino acids. The terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling or bioactive component. Unless stated or implied otherwise, the term aC or H 11 includes any polypeptide monomer or polymer with H11 immunologic specificity, including the intact aC antibody, and smaller and larger functionally equivalent polypeptides.
I
18 A "fusion polypeptide" is a polypeptide comprising regions in a different position in the sequence than occurs in nature. The regions may normally exist in separate proteins and are brought together in the fusion polypeptide: they may normally exist in the same protein but arc placed in a new arrangement inhe fusion poiypeptidc: or they may be synthetically arranged. For instance, as described below, the invention encompasses recombinant proteins (and the polynucleotides encoding the proteins) that are comprised of a functional portion of aC and a toxin. Methods of making these fusion proteins are known in the art and are described for instance in W093/07286.
A "functionally equivalent fragment" of a aC polypeptide varies from the native sequence by any combination of additions. deletions, or substitutions while preserving at least one functional property of the fragment relevant to the context in which it is being used. A functionally equivalent fragment of a aC polynucleotide either encodes a polypeptide that is functionally equivalent to HI 1 when produced by an expression system.
or has similar hybridization specificity as a HI 1 polynucleotide when used in a hybridization assay. A functionally equivalent fragment of a aC polypeptide typically has one or more of the following properties: ability to bind C antigen; ability to bind at least o one type of cancer cell in a specific manner; and an ability to elicit an immune response with a similar antigen specificity as that elicited by HI 1.
A "polynucleotide" is a polymeric form of nucleotides of any length, which contain deoxyribonucleotides, ribonucleotides. and analogs in any combination analogs.
Polynucleotides may have any three-dimensional structure, and may perform any function, known or unknown. The term "polynucleotide" includes double- single-stranded, and triple-helical molecules. Unless otherwise specified or required, any embodiment of the invention described herein that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the double stranded form of either the DNA, RNA or hybrid molecules.
SThe following are non-limiting examples of polynucleotides: a gene or gene Sfragment, exons, introns, mRNA, tRNA, rRNA. ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers. A 19 polynucleotide may comprise modified nucleotides. such as methylated nucleotides and nucleotide analogs, uracyl. other sugars and linking groups such as fluororibose and thioate. and nucleotide branches. The sequence of nucleotides may be interrupted by non- I nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. Other types of modifications included in this definition are caps, substitution of one or more of the naturally occurring nucleotides with an analog, and introduction of means for attaching the polynucleotide to proteins, metal ions. labeling components, other polynucleotides. or a solid support.
The term "recombinant" polynucleotide means a polynucleotide of genomic, cDNA. semisynthetic. or synthetic origin which either does not occur in nature or is linked to another polynucleotide in a nonnatural arrangement.
A "vector" refers to a recombinant DNA or RNA plasmid or virus that comprises a heterologous polynucleotide to be delivered into a target cell, either in vitro or in vivo.
The heterologous polynucleotide may comprise a sequence of interest for purposes of therapy, and may optionally be in the form of an expression cassette. As used herein, a vector need not be capable of replication in the ultimate target cell or subject. The term includes cloning vectors for the replication of a polynucleotide, and expression vectors for translation of a polynucleotide encoding sequence. Also included are viral vectors, which comprise a polynucleotide encapsidated or enveloped in a viral particle.
A "cell line" or "cell culture" denotes bacterial, plant, insect or higher eukaryotic **cells grown or maintained in vitro. The descendants of a cell may not be completely identical (either morphologically, genotypically, or phenotypically) to the parent cell. A Mab may be produced by a hybridoma or other cell. Methods of making hybridomas, both murine and human, are known in the art. Particular methods of producing human hybridomas are described and referenced throughout the specification.
A "host cell" denotes a prokaryotic or eukaryotic cell that has been genetically altered, or is capable of being genetically altered by administration of an exogenous polynucleotide, such as a recombinant plasmid or vector. When referring to genetically altered cells, the term refers both to the originally altered cell, and to the progeny thereof.
"Heterologous" means derived from a genotypically distinct entity from the rest of the entity to which it is being compared. For example, a polynucleotide may be placed by genetic engineering techniques into a plasmid or vector derived from a different source, and is a heterologous polynucleotide. A promoter removed from its native coding sequence and operatively linked to a coding sequence other than the native sequence is a heterologous promoter.
A "signal peptide" or "leader sequence" is a short amino acid sequence that directs a newly synthesized protein through a cellular membrane, usually the endoplasmic reticulum in eukaryotic cells, and either the inner membrane or both inner and outer membranes of bacteria. Signal peptides are typically at the N-terminal portion of a polypeptide and are typically removed enzymatically between biosynthesis and secretion of the polypeptide from the cell. The signal peptide is not present in the secreted protein, only during protein production.
An "isolated" polynucleotide or polypeptide is one that is substantially free of the materials with which it is associated in its native environment. By substantially free is meant at least 50%, preferably at least 70%, more preferably at least 80%, and even more preferably at least 90% free of these materials.
A "stable duplex" of polynucleotides, or a "stable complex" formed between any two or more components in a biochemical reaction, refers to a duplex or complex that is sufficiently long-lasting to persist between the formation of the duplex or complex and subsequent detection, including any optional washing steps or other manipulation that may take place in the interim.
A "biological sample" encompasses a variety of sample types, including blood and other liquid samples of biological origin, solid tissue samples such as a biopsy specimens or tissue cultures, or cells derived therefrom and the progeny thereof. The definition also .o includes samples that have been manipulated in any way after their procurement, such as by treatment with reagents, solubilization, or enrichment for certain components, such as proteins or polynucleotides. The term encompasses various kinds of clinical samples obtained from any species, and also includes cells in culture, cell supernatants, and cell lysates. Particularly, for the purposes described herein, biological samples comprise tumor 1 21 tissue or tissue thought to be tumorous and are obtained for instance by surgical resection, biopsy, aspiration or any method known in the art.
An "immunogen" refers to composition for human or animal use. which is administered with lthe inllterioL of conferring to the ecCipient a uc gr ee specific immunologic reactivity against a particular antigen. The immunologic reactivity may be carried out by antibodies or cells (particularly B cells, plasma cells. T helper cells, and cytotoxic T lymphocytes, and their precursors) that are immunologically reactive against the target, or any combination thereof. For purposes of this invention, the target is primarily tumor-associated C antigen or a tumor-specific portion thereof. The immunologic reactivity may be desired for experimental purposes. for treatment of a particular condition, for the elimination of a particular substance, or for prophylaxis. An active immunogen is intended to elicit an immune response that persists in the absence of the vaccine components.
"Adjuvant" as used herein has several meanings, all of which will be clear depending on the context in which the term is used. In the context of a pharmaceutical preparation, an adjuvant is a chemical or biological agent given in combination with or recombinantly fused to an antigen to enhance immunogenicity of the antigen. In the context of cancer diagnosis or management, adjuvant refers to a class of cancer patients S"with no clinically detectable tumor mass, but who are suspected of being at risk of recurrence.
When referring to a type of cancer that normally manifests as a solid tumor, a "clinically detectable" tumor is one that is detectable on the basis of tumor mass; by such procedures as CAT scan, X-Ray, or palpation. Biochemical, histological or :immunologic findings alone may be insufficient to meet this definition.
As used herein, "treatment" refers to clinical intervention in an attempt to alter the S natural course of the individual or cell being treated, and may be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of the treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression. amelioration or palliation of the disease state, and remission or improved prognosis.
The "pathology" associated with a disease condition is any condition that compromises the well-being, normal physiology, or quality of life of the affected individual. This may involve, but is not limited to, destructive invasion of affected tissues into previously unaffected areas, growth at the expense of normal tissue function, irregular or suppressed biological activity, aggravation or suppression of an inflammatory or immunologic response, increased susceptibility to other pathogenic organisms or agents, and undesirable clinical symptoms such as pain, fever. nausea. fatigue, mood alterations.
I0 and such other features as may be determined by an attending physician.
An "effective amount" is an amount sufficient to effect a beneficial or desired clinical result. An effective amount can be administered in one or more doses. In terms of treatment, an effective amount is amount that is sufficient to palliate, ameliorate, stabilize, reverse or slow the progression of the disease, or otherwise reduce the pathological consequences of the disease. In terms of an adjuvant, an effective amount is one sufficient .i to enhance the immune response to the immunogen. The effective amount is generally determined by the physician on a case-by-case basis and is within the skill of one in the art.
Several factors are typically taken into account when determining an appropriate dosage.
c....These factors include age, sex and weight of the patient. the condition being treated, the severity of the condition and the form of the antibody being administered. For instance, the concentration of scFv need not be as high as that of native antibodies in order to be therapeutically effective.
An "individual", '"patient" or "subject" is a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, humans, farm animals, sport animals, and pets.
The practice of the present invention employs, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, "Molecular Cloning: A Laboratory Manual", second edition (Sambrook et al., 1989); "Oligonucleotide Synthesis" Gait, ed., 1984); "Animal Cell Culture" Freshney, ed.. 1987); "Methods in Enzymology" (Academic Press, Inc.); "Handbook of Experimental Immunology" Wei C.C. Blackwell, eds.); "Gene Transfer Vectors for Mammalian Ce!!s" Miller M.P. Ca!os, eds., 1987); "Current Protocols in Molecular Biology" Ausubel et al., eds., 1987); "PCR: The Polymerase Chain Reaction", (Mullis et al., eds., 1994); "Current Protocols in Immunology" Coligan et al.. eds.. 1991). These techniques are applicable to the production of the polynucleotides and polypeptides of the invention, and, as such, may be considered in making and practicing the invention. Particularly useful techniques for particular embodiments will be discussed in the sections that follow.
The invention also encompasses aC conjugated to a chemically functional moiety.
Typically, the moiety is a label capable of producing a detectable signal. These conjugated aC are useful, for example, in detection systems such as quantitation of tumor burden, and imaging of metastatic foci and tumor imaging. Such labels are known in the art and include, but are not limited to, radioisotopes, enzymes, fluorescent compounds, chemiluminescent compounds, bioluminescent compounds substrate cofactors and inhibitors. See, for examples of patents teaching the use of such labels, U.S. Patent Nos.
3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366.241. The moieties may be covalently linked to aC. recombinantly linked, or conjugated to the aC through a secondary reagent, such as a second antibody, protein A. or a biotin-avidin complex.
S Other functional moieties include signal peptides, agents that enhance immunologic reactivity, agents that facilitate coupling to a solid support, vaccine carriers, bioresponse modifiers, paramagnetic labels and drugs. Signal peptides are described above and include prokaryotic and eukaryotic forms. Agents that enhance immunologic reactivity include, but are not limited to, bacterial superantigens. Agents that facilitate coupling to a solid support include, but are not limited to, biotin or avidin. Immunogen carriers include, but are not limited to, any physiologically acceptable buffers. Bioresponse modifiers include cytokines, particularly tumor necrosis factor (TNF), interleukin-2, interleukin-4, granulocyte macrophage colony stimulating factor and y interferons.
Suitable drug moieties include antineoplastic agents. These include, but are not limited to. radioisotopes, vinca alkaloids such as the vinblastine, vincristine and vindesine sulfates, adriamycin. bleomycin sulfate, carboplatin, cisplatin, cyclophosphamide, cyiarabine. dacarbaziie, daciinmuinycin, duainurubicin hydrochioride, doxorubicin hydrochloride, etoposide, fluorouracil, lomustine, mechlororethamine hydrochloride, melphalan, mercaptopurine, methotrexate, mitomycin. mitotane, pentostatin, pipobroman.
procarbaze hydrochloride, streptozotocin, taxol, thioguanine, and uracil mustard.
Immunotoxins. including single chain molecules, can be produced by recombinant means. Production of various immunotoxins is well-known in the art, and methods can be found, for example, in "Monoclonal Antibody-toxin Conjugates: Aiming the Magic Bullet." Thorpe et al. (1982) Monoclonal Antibodies in Clinical Medicine, Academic Press, pp. 168-190; Vitatta (1987) Science 238:1098-1104; and Winter and Milstein (1991) Nature 349:293-299. Suitable toxins include, but are not limited to, ricin, radionuclides, pokeweed antiviral protein, Pseudomonas exotoxin A, diphtheria toxin, ricin A chain, fungal toxins such as restrictocin and phospholipase enzymes. See, generally, "Chimeric Toxins," Olsnes and Pihl, Pharmac. Ther. 15:355-381 (1981); and "Monoclonal Antibodies for Cancer Detection and Therapy," eds. Baldwin and Byers, pp.
159-179, 224-266, Academic Press (1985).
The chemically functional moieties can be made recombinantly for instance by creating a fusion gene encoding the antigen binding fragment and functional regions from other genes enzymes). In the case of gene fusions, the two components are present "within the same polypeptide gene. Alternatively, the aC antigen binding fragments can be chemically bonded to the moiety by any of a variety of well known chemical procedures.
For example, when the moiety is a protein, the linkage may be by way of heterobifunctional cross linkers, SPDP, carbodiimide glutaraldehyde, or the like. The moieties may be covalently linked, or conjugated, through a secondary reagent, such as a second antibody, protein A, or a biotin-avidin complex. Paramagnetic moieties and the conjugation thereof to antibodies are well-known in the art. See, Miltenyi et al.
(1990) Cytometry 11:231-238.
El The aC antibody of this invention can be prepared in several ways. It is most conveniently obtained from cells engineered to express an antigen binding fragment containing SEQ ID NOS:I and 5 or other polynucleotides encoding aC binding fragments.
For example, the cells can be cultured in a suitable medium, and spent medium can be used as an antibody source. Optionally, matrix-coated channels or beads and cell co-cultures may be included to enhance growth of antibody-producing cells. For the production of large amounts of antibody, it is generally more convenient to obtain an ascites fluid. The method of raising ascites generally comprises injecting hybridoma cells into an immunologically naive histocompatible or immunotolerant mammal, especially a mouse.
The mammal may be primed for ascites production by prior administration of a suitable composition: Pristanc.
Alternatively, aC can be chemically synthesized using sequence data and other information provided in this disclosure, in conjunction with standard methods of protein synthesis. A suitable method is the solid-phase Merrifield technique. Automated peptide synthesizers are commercially available, such as those manufactured by Applied Biosystems, Inc. (Foster City, CA).
Sa C may also be obtained by employing routine recombinant methods such as described in Sambrook et al. (1989). For instance, using the amino acid and polynucleotide (SEQ ID NOS:1-6, and 13-18) sequences and information provided herein, a polynucleotide encoding either the aC H or L chain can be cloned into a suitable expression vector (which contains control sequences for transcription, such as a promoter).
The expression vector is in turn introduced into a host cell. The host cell is grown under suitable conditions such that the polynucleotide is transcribed and translated into a protein.
H and L chains of aC may be produced separately, and then combined by disulfide bond rearrangement. Alternatively, vectors with separate polynucleotides encoding each chain of aC, or a vector with a single polynucleotide encoding both chains as separate o *e transcripts, may be transfected into a single host cell which may then produce and assemble the entire molecule. Preferably, the host cell is derived from a higher eukaryote that can provide the normal carbohydrate complement of the molecule. The caC thus produced can be purified using standard techniques in the art. Polynucleotides encoding 26 aC for use in the production of cC can in turn be obtained from a hybridoma producing a aC antibody, or produced synthetically or recombinantly from the DNA sequences provided herein.
Another method of obtaining aC is to immunize suitable host animals with C antigen and to follow standard procedures for polyclonal or Mab production. Mabs thus produced can be "humanized" by methods known in the art. Examples of humanized antibodies are provided, for instance, in United States Patent Nos. 5,530,101 and 5.585,089.
"Humanized" antibodies are antibodies in which at least part of the sequence has been altered from its initial form to render it more like human immunoglobulins. In one version, the H chain and L chain C regions are replaced with human sequence. This is a fusion polypeptide comprising a HI I V region and a heterologous immunoglobulin C region. In another version, the CDR regions comprise H 11 amino acid sequences, while the V framework regions have also been converted human sequences. See, for example, EP 0329400. In a third version, V regions are humanized by designing consensus sequences of human and mouse V regions, and converting residues outside the CDRs that are different between the consensus sequences. The invention encompasses humanized Mabs.
In making humanized antibodies, the choice of framework residues can be critical in retaining high binding affinity. In principle, a framework sequence from any HuAb can serve as the template for CDR grafting; however, it has been demonstrated that straight CDR replacement into such a framework can lead to significant loss of binding affinity to the antigen. Glaser et al. (1992) J. Immunol. 149:2606; Tempest et al. (1992) Biotechnology 9:266; and Shalaby et al. (1992) J. Exp. Med 17:217. The more homologous a HuAb is to the original muAb, the less likely that the human framework will introduce distortions into the murine CDRs that could reduce affinity. Based on a sequence homology search against an antibody sequence database, the HuAb IC4 provides good framework homology to muM4TS.22, although other highly homologous HuAbs would be suitable as well, especially kappa L chains from human subgroup I or H chains from human subgroup III. Kabat et al. (1987). Various computer programs such as ElI ENCAD (Levitt et al. (1983) J. Mol. Biol. 168:595) are available to predict the ideal sequence for the V region. The invention thus encompasses HuAbs with different V regions. It is within the skill of one in the art to determine suitable V region sequences and to p izc tesc sequences. Methods fr obtaining antibodies with reduced Lll sLLuek1 IvlelUU Ug Ldl||ng immunogenicity are also described in U.S. Patent No. 5,270,202 and EP 699,755.
Methods of antibody production and isolation are well known in the art. See, for example, Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York. The HI 1 antibody is a human immunoglobulin of the IgM subclass, and may be isolated by any technique suitable for immunoglobulins of this isotypc. Purification methods may include salt precipitation (for example, with ammonium sulfate), ion exchange chromatography (for example, on a cationic or anionic exchange column run at neutral pH and eluted with step gradients of increasing ionic strength), gel filtration chromatography (including gel filtration HPLC), and chromatography on affinity resins such as protein A, protein G, hydroxyapatite. and antiimmunoglobulin. H 11 may also be purified on affinity columns comprising the C antigen; S for example, in the form of a purified Abl or Ab3. Preferably, HI1 is purified using Protein-A-CL-SepharoseTM 4B chromatography followed by chromatography on a DEAE- SepharoseTM 4B ion exchange column.
The invention also encompasses hybrid antibodies, in which one pair of H and L S 20 chains is obtained from a first antibody, while the other pair of H and L chains is obtained from a different second antibody. For purposes of this invention, one pair of L and H chains is from aC. In one example, each L-H chain pair binds different epitopes of the C antigen. Such hybrids may also be formed using humanized H or L chains.
Another aC contemplated by this invention is an antibody in which the H or L chain has been modified to provide additional properties. For instance, a change in amino acid sequence can result in reduced immunogenicity of the resultant polypeptide. The changes range from changing of one or more amino acids to the complete redesign of a Sregion such as a C region domain. Typical changes include, but are not limited to, those related to complement fixation, interaction with membrane receptors, and other effector functions. A recombinant antibody may also be designed to aid the specific delivery of a substance (such as a cytokine) to a tumor cell. Also encompassed by the invention are peptides in which various immunoglobulin domains have been placed in an order other than that which occurs in nature.
If aC is to be administered to an individual, it is preferably at least 80% pure, more preferably it is at least 90% pure, even more preferably it is at least 95% pure and free of pyrogens and other contaminants. In this context, the percent purity is calculated as a weight percent of the total protein content of the preparation, and does not include constituents which are deliberately added to the composition after the aC is purified.
The aC antibodies may be used for a number of purposes. These include eliciting an antibody response to produce aaC which can then be used to elicit a T cell response to aC or the C antigen and treating various types of cancer. These uses are elaborated more fully in a later section.
The invention encompasses polypeptide fragments of aC containing at least a portion of a V region of cC. Preferred fragments are those with the immunologic activity S 15 of HI 1. Also preferred are fragments which comprise amino acid sequences substantially different from other immunoglobulins, and fragments comprising a CDR. In one embodiment, the invention includes a polypeptide fragment of the aC H chain V region, comprising at least 25 consecutive amino acids, more preferably 30 consecutive amino acids of SEQ ID NO:2, or 5 consecutive amino acids of the CDR1 thereof, or at least 7 consecutive amino acids, preferably at least 9 consecutive amino acids of the CDR2 or CDR3 thereof. The invention also includes a polypeptide fragment of the aC L chain V region, comprising at least 25 consecutive amino acids, more preferably 30 consecutive amino acids of SEQ ID NO:5, or 7 consecutive amino acids of the CDR2 thereof, or at least 8 consecutive amino acids, preferably 10 consecutive amino acids of the CDR1 or CDR3 thereof.
The size of the aC polypeptides can be only the minimum size required to provide a desired function. The polypeptides can optionally comprise additional sequence, either native to aC, or from a heterologous source, as desired. aC peptides can contain only consecutive amino acids from a H 11 V region sequence that are not the same as the homologous region of A6. Polypeptides comprising 7 amino acids, more preferably about amino acids. more preferably about 15 amino acids, more preferably about 25 amino acids, more preferably about 50 amino acids, more preferably about 75 amino acids from the aC L or H chain V region are also included. Even more preferred are polypeptides comprising the entire aC L or H chain V region. Preferably the polypeptides are derived from H 11. Preferably, the polypeptides are the scFvs depicted in SEC ID NOS: 14 and 17.
The invention includes modified aC polypeptides which are functionally equivalent to HI 1, or have altered but measurable HI I immunologic activity. Modified polypeptides with improved HI 1 immunologic activity are preferred. Examples of modified polypeptides include those with conservative substitutions of amino acid residues, and one or more deletions or additions of amino acids which do not significantly deleteriously alter the immunologic activity.
One example of this is HI 1 polypeptides comprising one or more amino acid substitution in comparison with the prototype H 11 sequence. Substitutions can range from changing or modifying one or more amino acid residues to complete redesign of a region, such as the V region. Amino acid substitutions, if present, are preferably conservative substitutions that do not deleteriously affect folding or functional properties of the peptide.
Groups of functionally related amino acids within which conservative substitutions can be made are glycine/alanine; valine/isoleucine/leucine; asparagine/glutamine; aspartic acid/glutamic acid: serine/threonine/methionine; lysine/arginine: and 20 phenylalanine/tryosine/tryptophan. Polypeptides of this invention can be in glycosylated or unglycosylated form, can be modified post-translationally acetylation, and phosphorylation) or can be modified synthetically the attachment of a labeling group).
H11 polypeptide derivatives comprising both a H11 L chain and a H 11 H chain can 25 be formed as separate L and H chains and then assembled, or assembled in situ by an expression system for both chains. Such expression systems can be created by transfecting a suitable cell with a plasmid comprising separate transcribable regions for the L and H Schain, or by co-transfecting the same cell with plasmids for each chain. In a third method.
a suitable plasmid with a H chain encoding region is transfected into a H chain loss mutant.
H chain loss mutants can be obtained by treating approximately 2 x 107 HI I producing cells with fluorescein-labeled rabbit anti-mouse IgG (H chain specific, DAKO Corporation, Carpinteria, CA) according to the supplier's instruction. The stained and unstained cell populations are analyzed in a fluorescence-activated cell sorter. The unstained cells are collected in a sterilized tube and placed in 96-well plates with 1 cell/well by limiting dilution. The culture supernatants are then assayed by ELISA using goat anti-mouse IgG (H chain specific) and goat anti-mouse kappa. The clones with kappa-positive and IgG-negative phenotype are subcloned at least 3 times to obtain stable HI 11 mutants. mRNA from putative H chain loss mutant H 1I(H) clones can be isolated and the sequence of the L chain V region cDNA determined. Reverse PCR of the mRNA for the HI 1 VH is performed with 2 sets of and primers, used for cloning of HI l( 1 cDNA (Example A H chain loss mutant yields no detectable DNA band. Transfection of the cells proceeds with a suitable H chain plasmid.
Another aC derivative encompassed by this invention is an antibody in which the aC H or L chain has been modified to provide additional properties. For instance, a change in amino acid sequence can result in greater immunogenicity of the resultant polypeptide. The changes range from changing of one or more amino acids to the complete redesign of a region such as a C region domain. Changes contemplated affect complement fixation, interaction with membrane receptors, and other effector functions. A 20 recombinant HI 1 antibody can also be designed to aid the specific delivery of a substance (such as a lymphokine) to an effector cell. Also encompassed by the invention are proteins in which various immunoglobulin domains have been placed in an order other than that which occurs in nature.
The invention also encompasses single chain V region fragments ("scFv") of H 11.
25 Single chain V region fragments are made by linking L and/or H chain V regions by using a short linking peptide. Bird et al. (1988) Science 242:423-426. Any peptide having sufficient flexibility and length can be used as a linker in a scFv. Usually the linker is selected to have little to no immunogenicity. An example of a linking peptide is
(GGGGS)
3 which bridges approximately 3.5 nm between the carboxy terminus of one V region and the amino terminus of another V region. Other linker sequences can also be used, and can provide additional functions, such as a means for attaching a drug or a solid support.
All or any portion of the H or L chain can be used in any combination. Typically, the entire V regions are included in the scFv. For instance, the L chain V region can be linked to the H chain V region. Alternatively, a portion of the L chain V region can be linked to the H chain V region, or a portion thereof. Also contemplated are scFvs in which the H chain V region is from HI 1, and the L chain V region is from another immunoglobulin. It is also possible to construct a biphasic. scFv in which one component is a HI 1 polypeptide and another component is a different polypeptide, such as a T cell epitope.
The scFvs can be assembled in any order, for example, V--(linker)--VL or VL-(linker)-VH. For example, SEQ ID NOS:13 and 16 show H I -scFv2 constructs having the form VL-(linker)--VH. However, the construct shown in SEQ ID NO: 13 forms monomers, while the construct shown in SEQ ID NO: 16 forms dimers. There may be a difference in the level of expression of these two configurations in particular expression systems, in which case one of these forms may be preferred. Tandem scFvs can also be made, such as (X)-(linker)--(X)-linker)-(X), in which X are aC polypeptides, or combinations of aC polypeptides with other polypeptides. In another embodiment, single chain antibody polypeptides have no linker polypeptide. or just a short, inflexible 20 linker. Exemplary configurations include VL-VH and VH--VL. The linkage is too short to permit interaction between VL and VH within the chain, and the chains form homodimers with a VLNH antigen binding site at each end. Such molecules are referred to in the art as "diabodies".
ScFvs can be produced either recombinantly or synthetically. For synthetic .25 production of scFv, an automated synthesizer can be used. For recombinant production of scFv, a suitable plasmid containing a polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such as Escherichia coli, and the protein expressed by the polynucleotide can be isolated using standard protein purification techniques.
A particularly useful system for the production of scFvs is plasmid pET-22b(+) (Novagen, Madison.WI) in E. coli. pET-22b(+) contains a nickel ion binding domain consisting of 6 sequential histidine residues, which allows the expressed protein to be purified on a suitable affinity resin. Another example of a suitable vector is pcDNA3 (Invitrogen, San Diego, CA), described above.
Expression conditions should ensure that the scFv assumes functional and, preferably, optimal tertiary structure. Depending on the plasmid used (especially the activity of the promoter) and the host cell, it may be necessary to modulate the rate of production. For instance, use of a weaker promoter, or expression at lower temperatures, may be necessary to optimize production of properly folded scFv in prokaryotic systems: or, it may be preferably to express scFv in eukaryotic cells.
Preferred scFv comprise at least 10 consecutive amino acids of SEQ. ID NO:2 and at least 10 consecutive amino acids of SEQ. ID NO:5, especially wherein the amino acids of SEQ. ID NO:2 and the amino acids of SEQ. ID NO:5 are joined by a linker polypeptide 15 of 5 to 20 amino acids, or comprising the L chain V region and the H chain V region of HI .1.
The invention also encompasses polymeric forms ofacC polypeptides. containing a plurality of aC polypeptides. One embodiment is a linear polymer of aC polypeptides, optionally conjugated to carrier. These linear polymers can comprise multiple copies of a 20 single uC polypeptide, or combinations of different aC polypeptides, and can have tandem aC polypeptides, or aC polypeptides separated by other amino acid sequences. Another embodiment is aC multiple antigen peptides (MAPs). MAPs have a small immunologically inert core having radially branching lysine dendrites, onto which a number of aC polypeptides are covalently attached. See for instance, Posnett et al. (1988) 25 J. Biol. Chem. 263:1719-1725; and Tam (1989) Meth. Enz. 168:7-15. The result is a large macromolecule having a high molar ratio of aC polypeptides to core. MAPs are efficient immunogens and useful antigens for immunoassays. The core for creating an aC MAP can be made by standard peptide synthesis techniques, or obtained commercially, from Quality Controlled Biochemicals, Inc., Hopkinton, MA. A typical core matrix is made up of three levels of lysine and eight amino acids.
33 When using uC polypeptides as immunogens, preferably the polypeptides are delivered in conjunction with a carrier. Any carrier can be used which is not harmful to the host. Suitable carriers are typically large, slowly metabolized macromolecules such as proteins; poiysaccharides (such as iatex functionaiized Sepharose, agarose, ceiiuiose, cellulose beads and the like); polymeric amino acids (such as polyglutamic acid, polylysine, and the like); amino acid copolymers; and inactive virus particles or attenuated bacteria, such as Salmonella. Especially useful carrier proteins are serum albumins, keyhole limpet hemacyanin (KLH), certain Ig molecules, thyroglobulin. ovalbumin, and tetanus toxoid. KLH is especially preferred.
tiC polypeptides of the invention can be identified in a number of ways. For example. the V regions of the L and H chains can be screened by preparing a series of short polypeptides that together span the entire V region amino acid sequence. Using a series of polypeptides of 20 or 50 amino acids in length, each aC V region can be surveyed for useful functional properties. It is also possible to carry out a computer analysis of a protein sequence to identify potentially interesting polypeptides, such as those that bear the shape of D2, or those involved in idiotype-anti-idiotype contact.
SThe invention further encompasses various adaptations of aC described in this section combined in various fashions to yield other aC polypeptides with desirable properties. For instance. aC polypeptides with modified amino acid residues can be 20 comprised in a MAP. In another example, a aC scFv is fused to a cytokine, such as IL-2.
All such combinations are contemplated in this invention.
The polypeptides of this invention can be made by any suitable procedure, including proteolysis of the aC antibody, by recombinant methods or by chemical synthesis. These methods are known in the art and need not be described in detail herein.
25 Examples of proteolytic enzymes include, but are not limited to, trypsin, chymotrypsin, pepsin, papain, V8 protease, subtilisin, plasmin, and thrombin. Intact aC can be incubated with one or more proteinases simultaneously or sequentially. Alternatively, or in addition, Sintact antibody can be treated with disulfide reducing agents. Peptides can then be separated from each other by techniques known in the art including, but not limited to, gel filtration chromatography, gel electrophoresis, and reverse-phase HPLC.
34 aC polypeptides can also be made by expression from a polynucleotide encoding the peptide according to the information provided elsewhere in this application, in a suitable expression system. Typically, polynucleotides encoding a aC polypeptide are ligated into an expression vector under control of a suitable promoter and used to genetically alter the intended host cell. Both eukaryotic and prokaryotic host systems can be used. The polypeptide is then isolated from lysed cells or from the culture medium and purified to the extent needed for its intended use. Examples of prokaryotic host cells appropriate for use with this invention include E. coli. Examples of eukaryotic host cells include avian. insect, plant, and animal cells including, but not limited to, COS7, HeLa, and CHO cells.
In certain applications, such as when a HI 1 polypeptide is expressed in a suitable storage medium such as a plant seed, the H 1I polypeptide can be used without purification. Fiedler et al. (1995) Biotechnology 13:1090-1093. For most applications, it is generally preferable that the polypeptide is at least partially purified from other cellular constituents. Preferably, the polypeptide is at least about 50% pure as a weight percent of total protein. More preferably, the protein is at least about 50-75% pure. For clinical use, the polypeptide is preferably at least about 80% pure.
The invention also encompasses methods of detecting C antigen in a biological sample. The methods include obtaining a biological sample, contacting the sample with 20 aC under conditions that allow antibody antigen binding and detecting binding, if any, of the antibody to the antigen.
The invention also encompasses methods of detecting anti-H 11 or anti-aC in a biological sample. Anti-aC is detectable whenever it cross-reacts with H II. Anti-aC with this activity can spontaneously arise during the course of a tumor-associated disease. Anti- S 25 aC with this activity is especially likely in individuals who have received a course of therapy with aC. These methods are applicable in a clinical setting, for example, for S monitoring antibody levels in an individual, as well as an industrial setting, as in commercial production of anti-H 1 or anti-aC.
The assay methods entail contacting any anti-H I or anti-aC target antibody in the sample with a HI 1 antibody or polypeptide under conditions suitable to allow the formation of a stable complex between the target and HI 1, and detecting any stable complex formed. The sample is suitably prepared before conducting the assay, optionally by enriching for antibody concentration. When using intact murine aC. it is generally preferable to deIplee the sampl e any i-mouse imm unolcbu!in ;ctivity that ma. be present. Anti-mouse immunoglobulin antibody can be removed from a sample, for example. by precipitation with normal mouse IgG or adsorption with a mouse Ig adsorbent.
Binding of anti-mouse immunoglobulin antibody, particularly that specific for the Fc region, can be minimized by judicious choice of the reagents of the assay. or Fab fragments of murine aC and other reagents such as humanized aC or HI 1. with fewer mouse determinants are appropriate.
After the sample is suitably prepared, it is mixed with a excess aC under conditions that permit formation of a complex between aC and any target antibody that may be present. The amount of complex is then determined, and compared with complexes formed with standard samples containing known amounts of target antibody in the range expected. Complex formation can be observed by any method known in the art such as immunoprecipitation or nephelometry, but it is generally more sensitive to employ a reagent labeled with such labels as radioisotopes such as '251. enzymes such as peroxidase and P-galactosidase, or fluorochromes such as fluorescein.
The invention provides various polynucleotides encoding the antibody H 11 or 20 fragments of H 1. based on the polynucleotide sequences provided herein (SEQ ID NOS:I and Various embodiments are described in this section, comprising a number of different combinations of the H 11 H or L chain V region sequences. In general, a H 11 polynucleotide of this invention encodes at least one feature that is unique to the HI 1 molecule (in comparison with other immunoglobulins and, in particular, the A6 antibody).
25 Preferably, this feature is related in some way to an immunologic reactivity of HI 1.
The invention encompasses polynucleotides encoding a portion of the Hi 1 L chain V region, comprising at least about 70 consecutive nucleotides, preferably at least about consecutive nucleotides, more preferably at least about 100 consecutive nucleotides, even more preferably at least about 150 nucleotides of SEQ ID NO:4. The invention also encompasses a polynucleotide encoding a portion of the HI L chain V region, comprising at least about 25 consecutive nucleotides, preferably at least about 30 consecutive nucleotides, and even more preferably at least about 35 consecutive nucleotides of the CDRI encoding sequence thereof. The invention also encompasses a polynucleotide encoding a portion of the HI! L chain V reion, comprising at least about 20 consecutive nucleotides, preferably at least about 25 consecutive nucleotides, and even more preferably at least about 35 consecutive nucleotides of the CDR2 or CDR3 encoding sequence thereof.
The invention also encompasses polynucleotides encoding a portion of the H 1 H chain V region, comprising at least about 70 consecutive nucleotides, preferably at least about 80 consecutive nuclcotides, more preferably at least about 100 consecutive nucleotides, even more preferably at least about 150 nucleotides of SEQ ID NO:1. The invention also encompasses a polynucleotide encoding a portion of the HI 1 L chain V region, comprising 15 consecutive nucleotides of the CDR1 encoding sequence thereof.
The invention also encompasses a polynucleotide encoding a portion of the HI 1 L chain V region, comprising at least about 20 consecutive nucleotides, preferably at least about consecutive nucleotides, and even more preferably at least about 35 consecutive nucleotides of the CDR2 or CDR3 coding sequence thereof.
The invention includes isolated H11 polynucleotides encoding a polypeptide Shaving immunologic activity of HI 1, wherein the polypeptide encodes at least 5 amino S 20 acids of a V L chain of HI 1 as depicted in SEQ. ID NO:5. The invention also includes isolated H 1 polynucleotides encoding a polypeptide having immunologic activity of H 11, wherein the polynucleotide encodes at least 5 amino acids of a V H chain of H 1 as depicted in SEQ. ID NO:2. The polynucleotide sequence can be similar to those depicted in SEQ. ID NO: 1 or SEQ ID NO:4 with changes designed to optimize codon usage, 25 stability, facilitate cloning, or any other purpose. It is within the skill of one in the art, given the amino acid sequence in SEQ ID NO:2 or SEQ ID NO:5, to design such polynucleotides. Preferred polynucleotides encode at least five amino acids of a H11
CDR.
The invention also encompasses polynucleotides encoding for functionally equivalent variants and derivatives of HI 1 and functionally equivalent fragments thereof which may enhance, decrease or not significantly affect properties of the polypeptides encoded thereby. These functionally equivalent variants, derivatives, and fragments display the ability to specifically recognize C antigen. For instance, changes in a DNA sequence ihai du not ihaiige the encoded amino acid sequence, as weii as those that result in conservative substitutions of amino acid residues, one or a few amino acid deletions or additions, and substitution of amino acid residues by amino acid analogs are those which will not significantly affect properties of the encoded polypeptide. Conservative amino acid substitutions are glycine/alanine; valine/isoleucine/leucine; asparagine/glutamine; aspartic acid/glutamic acid: serine/threonine/methionine; lysine/arginine; and phenylalanine!tyrosineitryptophan.
The polynucleotides of the invention may comprise additional sequences. such as additional encoding sequences within the same transcription unit, controlling elements such as promoters, ribosome binding sites, and polyadenylation sites, additional transcription units under control of the same or a different promoter, sequences that permit S 15 cloning, expression, and transformation of a host cell, and any such construct as may be desirable to provide embodiments of this invention.
S. The invention encompasses a polynucleotide of at least about 15 consecutive nucleotides, preferably at least about 20 nucleotides, more preferably at least about 00. consecutive nucleotides. more preferably at least about 35 consecutive nucleotides, more 20 preferably at least about 50 consecutive nucleotides, even more preferably at least about nucleotides, still more preferably at least about 100 nucleotides, still more preferably at S• least about 200 nucleotides, and even more preferably at least about 300 nucleotides that forms a stable hybrid with a polynucleotide encoding the L chain or H chain V region of H 1, but not with other immunoglobulin encoding regions known at the time of filing of 25 this application. Any set of conditions may be used for this test, as long as at least one set of conditions exist wherein the test polynucleotide demonstrates the required specificity.
SPreferably, the H 11 encoding sequences to which the test polynucleotide binds are those shown in SEQ. ID NOS: and 4. Since the known immunoglobulin sequences fall into a hierarchy of similarity with that of H 1, the test may be performed by comparing the hybridization of the test polynucleotide with the H 11 sequence with the hybridization with the most closely related sequences. Preferred is a panel of about 10 of the most closely related sequences to SEQ. ID NO:1. 3, 4 or Hybridization reactions can be performed under conditions of different "si'lingelcy". LCondiions that increase siringency of a hybridization reaction are well known. See, for example, Sambrook and Maniatis. Examples of relevant conditions include (in order of increasing stringency): incubation temperatures of 25 0 C, 37 0 C, 50 0
C
and 68 0 C; buffer concentrations of 10 x SSC, 6 x SSC, 1 x SSC, 0.1 x SSC (where SSC is 0.15 M NaCI and 15 mM citrate buffer) and their equivalent using other buffer systems; formamide concentrations of 25%. 50%, and 75%: incubation times from 5 minutes to 24 hours: 1, 2, or more washing steps: wash incubation times of 1, 2, or 15 minutes: and wash solutions of 6 x SSC, 1 x SSC, 0.1 x SSC, or deionized water.
Useful HI 1 polynucleotides encoding fragments of HI 1 may be identified by generating polynucleotide fragments (based on SEQ ID NO: 1 or SEQ ID NO:4, for example) and testing the polypeptides encoded thereby for a function of interest.
Alternatively, the polypeptide fragment encoded by a particular polynucleotide can be prepared and tested for a function of interest. Alternatively, given a uC polypeptide with i desirable properties, polynucleotides can be designed that encode the polypeptide.
*.o Included in all these embodiments are polynucleotides with encoding regions for HI 1 polymers, fusion proteins, humanized immrunoglobulins, single-chain V regions, and 20 other particular polypeptides of interest. These polypeptides are described above.
The invention also provides polynucleotides covalently linked with a detectable label. Such polynucleotides are useful, for example, as probes for detection of related nucleotide sequences.
The polynucleotides of this invention can be obtained using chemical synthesis, 25 recombinant cloning methods. PCR, or any combination thereof. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art can use the sequence data provided herein to obtain a desired polynucleotide by employing a DNA synthesizer or ordering from a commercial service.
Alternatively, aC polynucleotide sequences can be obtained from a aC antibody producing cell line, aC cloning vector, or aC expression vector. RNA or DNA encoding the desired sequence can be isolated, amplified, and processed by standard recombinant techniques. Such techniques include digestion with restriction nucleases, and amplification by polymerase chain reaction (PCR), or a suitable combination thereof. PCR tecnologUy s describedCU in L.S. P atlnt Nos. 4,683,195, 4,800,159, t,54,0 and 4,683,202, as well as PCR. The Polymerase Chain Reaction, Mullis et al. eds., Birkauswer Press. Boston (1994).
Polynucleotides comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification. Polynucleotides can be introduced into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis. transfection. f-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome. Amplified DNA can be isolated from the host cell by standard methods. See, Sambrook et al. (1989). RNA can also be obtained from transformed host cell, or it can be obtained directly from the DNA by using a DNA-dependent RNA polymerase.
The present invention further encompasses a variety of vectors comprising a HI 1 polynucleotide. These vectors can be used for expression of recombinant polypeptides are also a source of HI 1 polynucleotides. Cloning vectors can be used to obtain replicate copies of the HI 1 polynucleotides they contain, or as a means of storing the polynucleotides in a depository for future recovery. Expression vectors (and host cells containing these expression vectors) can be used to obtain polypeptides produced from the polynucleotides they contain. They can also be used where it is desirable to express HI 1 in an individual and thus have intact cells capable of synthesizing the polypeptide. such as in gene therapy. Suitable cloning and expression vectors include any known in the art, those for use in bacterial, mammalian, yeast and insect expression systems. Specific vectors and suitable host cells are known in the art and are not described in detail herein.
See e.g. Gacesa and Ramji, Vectors, John Wiley Sons (1994).
Cloning and expression vectors typically contain a selectable marker (for example, a gene encoding a protein necessary for the survival or growth of a host cell transformed 0** 0 0 0 00 9.0.
0. 0 00 00** 00.0 0 000* *0 0 00 y 0 0 with the vector), although such a marker gene can be carried on another polynucleotide sequence co-introduced into the host cell. Only those host cells into which a selectable gene has been introduced will grow under selective conditions. Typical selection genes eithcr: conero resistance to antibiotics or oher oxinis, amnpicillin, neomycin.
methotrexate; complement auxotrophic deficiencies; or supply critical nutrients not available from complex media. The choice of the proper marker gene will depend on the host cell. and appropriate genes for different hosts are known in the art. Vectors also typically contain a replication system recognized by the host.
Suitable cloning vectors can be constructed according to standard techniques, or selected from a large number of cloning vectors available in the art. While the cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, or may carry marker genes. Suitable examples include plasmids and bacterial viruses, pUC18, mpl8, mp19, pBR322. pMB9, ColE1, pCR1, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28. These and other cloning vectors are available from commercial vendors such as BioRad, Stratagene. and Invitrogen.
Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide encoding a aC polypeptide of interest. The polynucleotide encoding aC polypeptide is operatively linked to suitable transcriptional controlling elements, such as 20 promoters, enhancers and terminators. For expression translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons. These controlling elements (transcriptional and translational) can be derived from the H11 gene, or heterologous derived from i other genes or other organisms). A polynucleotide sequence encoding a signal peptide can 25 also be included to allow a aC polypeptide to cross or lodge in cell membranes or be secreted from the cell. A number of expression vectors suitable for expression in eukaryotic cells including yeast, avian, and mammalian cells are known in the art. One example of an expression vector is pcDNA3 (Invitrogen, San Diego, CA), in which transcription is driven by the cytomegalovirus (CMV) early promoter/enhancer. This vector also contains recognition sites for multiple restriction enzymes for insertion of an 20 uC polynucleotide of interest. Another example of an expression vector (system) is the baculovirus/insect system.
Also encompassed by the invention are expression systems suitable for use in antibody-targeted gene therapy comprising a aC polynucleotide. Suitable .systems are described for instance by Brown et al. (1994) Virol. 198:477-488; and Mivamura et al.
(1994) Proc. Nail. Acad. Sci. USA 91:8507-8511.
The vectors containing the polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate. DEAE-dextran. or other substances: microprojectile bombardment: lipofection: and infection (where the vector is an infectious agent, such as vaccinia virus, which is discussed below). The choice of introducing vectors or aC polynucleotides will often depend on features of the host cell.
Once introduced into a suitable host cell, expression of a aC polypeptide can be determined using any assay known in the art. For example, presence of aC polypeptide can be detected by RIA or ELISA of the culture supernatant (if the HI 1 polypeptide is secreted) or cell lysates.
A particularly useful expression vector for HI polynucleotides is a vaccinia virus comprised of a HI 1 polynucleotide sequence, which can also be used in vaccine preparations. Moss (1991) Science 252:1662-1667. To introduce polynucleotide sequences encoding H 11 polypeptide, including HI 1 polypeptide fragments, into vaccinia.
the polynucleotide sequence of interest is first inserted into a plasmid containing a vaccinia virus promoter with flanking sequences homologous to vaccinia DNA not required for replication. Plasmid-containing cells are then infected with vaccinia, which leads to a low level of homologous recombination between plasmid and virus, with resultant transfer of the vaccinia promoter and H11 polypeptide-encoding polynucleotide sequence into the vaccinia virus genome. Typically, the HI 1 polynucleotide is inserted into the viral TK (thymidine kinase) gene. Insertion into the TK site attenuates the virus more than 10,000 fold compared to wild type. Flexner et al. (1980) Vaccine 88 (Cold Spring Harbor Laboratory), pp. 179-184. Recombinant virus is identified by the TK" phenotype.
Preferably, expression of the H 11 polynucleotide is under the control of the vaccinia 42 early/late promoter (7.5 whereby the resultant HI I polypeptides can be expressed in infected cells throughout the life cycle of the virus. However, other promoters known in the art can be used, such as pH6, or synthetic promoters. Expression of the HI 1 polypeptide occurs in cells infected with the recombinant vaccinia or individuals immunized with the live recombinant vaccinia virus. Any one of several strains of vaccinia can be used, including, but not limited to, WR. ALVAC. and NYVAC.
A vector of this invention can contain one or more polynucleotides encoding a uC polypeptide. It can also contain polynucleotide sequences encoding other polypeptides that enhance, facilitate, or modulate the desired result, such as lymphokines, including, but not limited to. IL-2. IL-4. GM-CSF. TNF-a, and IFN-y. A preferred lymphokine is GM- CSF. Preferred GM-CSF constructs are those which have been deleted for the AU-rich elements from the 3' untranslated regions and sequences in the 5' untranslated region that are capable of forming a hairpin loop. Also embodied in this invention are vaccinia vectors encoding for recombinant aC variants, such as scFvs, chimeras, and polymers.
Other embodiments of this invention are host cells transformed with aC polynucleotides and vectors comprising aC polynucleotide sequences, as described above.
Both prokaryotic and eukaryotic host cells may be used. Prokaryotic hosts include bacterial cells, for example E. coli and Mycobacteria. Among eukaryotic hosts are yeast.
insect. avian, plant and mammalian cells. Host systems are known in the art and need not 20 be described in detail herein. Examples of mammalian host cells include CHO and NSO, obtainable from the European Collection of Cell Cultures (England). Transfection of NSO cells with a plasmid, for example, which is driven by a CMV promoter, followed by amplification of this plasmid in using glutamine synthetase provides a useful system for protein production. Cockett et al. (1990) Bio/Technology 8:662-667.
The host cells of this invention can be used, inter alia, as repositories of aC polynucleotides, or as vehicles for production of aC polynucleotides and polypeptides.
They may also be used as vehicles for in vivo expression of aC polypeptides. The HI 1 polynucleotides of this invention can be used in expression systems to produce H 11 polypeptides, intact HI 1. or recombinant forms of HI 1, such as are described below.
I;
The polynucleotides of this invention have several uses. They are useful, for example, in expression systems for the production of aC. They are also useful as hybridization probes to assay for the presence of aC polynucleotide or related sequences in a sample using methods well known to those in the art. Further, the polynucleotides are also useful as primers to effect amplification of desired polynucleotides. The polynucleotides of this invention are also useful in pharmaceutical compositions including vaccines and for gene therapy.
The polynucleotides can also be used as hybridization probes for detection ofaC encoding sequences. Suitable samples include cells transformed ex vivo for use in gene therapy. In one illustration. DNA or RNA is extracted from a sample. and optionally run on a gel and/or digested with restriction endonucleases. The processed sample polynucleotide is typically transferred to a medium suitable for washing. The sample polynucleotide is then contacted with the H 11 polynucleotide probe under conditions that permit a stable duplex to form if the sample contains a matching aC sequence. Any stable duplexes formed are detected by any suitable means. For example, the aC polynucleotide probe can be supplied in labeled form, and label remaining with the sample after washing will directly reflect the amount of stable duplex formed. In a second illustration, hybridization is performed in situ. A suitably prepared tissue sample is overlaid with a labeled probe to indicate the location aC encoding sequences.
A short aC polynucleotide can also be used as a primer for a PCR reaction, particularly to amplify a longer sequence comprising a region hybridizing with the primer.
This can be conducted preparatively, in order to produce polynucleotide for further genetic manipulation. It can also be conducted analytically, to determine whether a aC encoding polynucleotide is present, for example, in a sample of diagnostic interest.
Another use of the polynucleotides is in vaccines and gene therapy. The general principle is to administer the polynucleotide so that it either promotes or attenuates the expression of the polypeptide encoded therein. Thus, the present invention includes methods of inducing an immune response and methods of treatment comprising administration of an effective amount aC polynucleotides to an individual. In these methods, a aC polynucleotide encoding a aC polypeptide is administered to an individual,
S
MS either directly or via cells transfected with the aC polynucleotide. Preferably, the uC polynucleotide is in the form of a circular plasmid. preferably in a supercoiled configuration. Preferably, the aC polynucleotide is replicated inside a cell. Thus. the aC polynucleotide is operatively linked to a suitable promoter, such as a heterologous promoter that is intrinsically active in cells of the target tissue type. Preferably, once in cell nuclei, plasmids persist as circular non-replicating episomal molecules. In vitro mutation can be carried out with plasmid constructs to encode, for example, molecules with greater affinity and/or avidity.
To determine whether plasmids containing uC polynucleotides are capable of aC expression in eukaryotic cells, cells such as COS-7. CHO, or HeLa can be transfected with the plasmids. Expression ofuC is then determined by immunoassay; for example, by Western blot. Smaller aC polypeptides can be detected, for example, by constructing the plasmid so that the resultant aC polypeptide is fused with a tag, such as a target epitope or enzyme label. Further characterization of the expressed aC polypeptide can be achieved i 15 by purifying the peptide and then conducting one of the functional assays described herein.
In one mode of gene therapy, the polynucleotides of this invention are used for genetically altering cells ex vivo. In this strategy, cells removed from a donor or obtained from a cell line are transfected or transduced with vectors encoding a aC polypeptide. and then administered to a recipient. Suitable cells for transfection include peripheral blood S 20 mononuclear cells.
In another mode of gene therapy, the polynucleotides of this invention are used for genetically altering cells in vivo. The purpose includes, but is not limited to, treating various types of cancer.
aC polypeptides can be characterized in several ways. For instance, a aC 25 polypeptide may be tested for its ability to bind specifically to cancer cells, for its ability to specifically inhibit the binding between cancer cells and intact HI 1. A aC polypeptide can also react with anti-CDR3 polypeptides. aC polypeptides can also be tested for their Sability to palliate or ameliorate neoplastic disease, such as carcinomas. It is understood that only one of these properties need be present in order for a polypeptide to come within the scope of this invention, although preferably more than one of these properties is present.
The ability ofa uC polypcptide to bind cancer cells or antigenic fractions thereof can be tested by immunoassay. Any form of direct binding assay is suitable. In one such assay, the cancer cell or the putative aC polypeptide is labeled. Suitable labels include radioisotopes such as "5l. enzymes such as peroxidase. fluorescent labels such as fluorescein. and chemiluminescent labels. Typically, the other binding partner is insolubilized (for example, by coating onto a microtiter plate) to facilitate washing. After combining the labeled component with the insolubilized component. the solid phase is washed and the amount of bound label is determined. Another such assay is a sandwich assay, in which the putative aC polypeptide is captured by a first anti-immunoglobulin on a solid phase and developed with aC antibody. In either of these examples, the extent of binding of aC is directly related to the amount of label bound to the solid phase.
To conduct the inhibition assays, the putative uC polypeptide is titered for its ability to decrease the binding of HI 1 to cancer cells. Either of the binding pairs in the reaction to be inhibited is labeled, while the other is typically insolubilized in order to facilitate washing. The putative aC polypeptide is typically mixed with the labeled component, and then the mixture is combined with the solid phase. Polypeptides with the characteristics of H I will proportionately decrease the amount of label attached to the 20 solid phase, compared with control polypeptides. This test may be more sensitive than measuring direct binding, because lower affinity interaction between aC and C antigen may be too weak to form a stable bond, but adequate to interfere with the binding of another ligand-receptor pair when present at sufficient concentration.
The present invention encompasses pharmaceutical compositions and immunogenic 25 compositions containing aC either alone or in combination. Such pharmaceutical compositions and vaccines are useful for eliciting an immune response and treating
C*
•neoplastic diseases, either alone or in conjunction with other forms of therapy, such as chemotherapy or radiotherapy.
The preparation of pharmaceutical compositions that contain aC antibody, or a polynucleotide or a polypeptide derivative thereof as an active ingredient is conducted in 46 accordance with generally accepted procedures for the preparation of pharmaceutical preparations. See, for example. Remington's Pharmaceutical Sciences 18th Edition (1990), E.W. Martin ed., Mack Publishing Co., PA. Depending on the intended use and mode of ad.inistr it my be esirable to process ihe active ingredient further in the preparation of pharmaceutical compositions. Appropriate processing may include sterilizing, mixing with appropriate non-toxic and non-interfering components, dividing into dose units, and enclosing in a delivery device.
Liquid pharmaceutically acceptable compositions can, for example, be prepared by dissolving or dispersing a polypeptide embodied herein in a liquid excipient, such as water.
saline, aqueous dextrose, glycerol. or ethanol. The composition can also contain other medicinal agents, pharmaceutical agents. adjuvants, carriers, and auxiliary substances such as wetting or emulsifying agents, and pH buffering agents.
Pharmaceutical compositions of the present invention are administered by a mode appropriate for the form of composition. Typical routes include subcutaneous, intramuscular, intraperitoneal, intradermal, oral, intranasal. and intrapulmonary by aerosol). Pharmaceutical compositions of this invention for human use are typically administered by a parenteral route, most typically intracutaneous, subcutaneous, or intramuscular.
Pharmaceutical compositions for oral. intranasal, or topical administration can be 20 supplied in solid, semi-solid or liquid forms, including tablets, capsules, powders, liquids.
and suspensions. Compositions for injection can be supplied as liquid solutions or suspensions, as emulsions, or as solid forms suitable for dissolution or suspension in liquid prior to injection. For administration via the respiratory tract, a preferred composition is one that provides a solid, powder, or liquid aerosol when used with an appropriate 25 aerosolizer device. Although not required, pharmaceutical compositions are preferably supplied in unit dosage form suitable for administration of a precise amount. Also contemplated by this invention are slow release or sustained release forms, whereby a relatively consistent level of the active compound are provided over an extended period.
Compositions embodied in this invention can be assessed for their ability to recognize specifically a neoplasia. Accordingly, test compounds are prepared as a suitable
I
pharmaceutical composition and administered to test subjects. Initial studies are preferably done in small animals such as mice or rabbits, optionally next in non-human primates and then ultimately in humans. Immunogenicity is preferably tested in individuals without a previous antibody response. A test composition in an appropriate dose is administered on an appropriate treatment schedule. It may be appropriate to compare different doses and schedules within the predicted range. Such testing is within the skill of one in the art.
Compositions of this invention are particularly suitable for administration to humans with a neoplastic disease. Especially relevant are melanoma. neuroblastoma, glioma. sarcoma, lymphoma, and small cell lung cancer.
1 0 Also included in this invention are methods for treating cancer. The methods comprise administering an amount of a pharmaceutical composition containing aC effective to achieve the desired effect, be it palliation of an existing tumor mass or prevention of recurrence. For treatment of cancer, the amount of a pharmaceutical composition administered is an amount effective in producing the desired effect. An effective amount can be provided in one or a series of administrations.
The effective amount of aC antigen binding fragments to be administered will depend upon several factors, such as the route of administration, the condition of the individual, and the desired objective. The term "therapeutically effective" means that the amount of antigen binding fragment used is of sufficient quantity to ameliorate the cancer.
.i 20 "Ameliorate" denotes a lessening of the detrimental effect of the cancer on the individual.
Typically, if administered directly, the amount per administration is about 10 Alg to 20 mg, preferably 250 pg to 10 mg, more preferably 300 pg to 5 mg, even more preferably 500 jig to 2.5 mg. Administrations are typically conducted on a weekly or biweekly basis until a desired, measurable parameter is detected, such as diminution of disease symptoms.
o .25 Administration can then be continued on a less frequent basis, such as biweekly or S"monthly, as appropriate.
~The various compositions of this invention can be used alone, or in conjunction with other active agents that promote the desired objective, or provide a desirable adjunct therapy. Suitable active agents include the anti-neoplastic drugs and bioresponse modifiers described above and effector cells such as those described by Douillard et al. (1986) Hybridonas (Supp. 1:5139).
When used for immunotherapy, aC can be unlabeled or labeled with a therapeutic gent as described above. These aents ca n be coupled either directly or indirectly to the polypeptides of the invention. One example of indirect coupling is by use of a spacer moiety. These spacer moieties, in turn, can be either insoluble or soluble (Diener et al.
(1986) Science 231:148) and can be selected to enable drug release from aC at the target site. Alternatively, an aC and a therapeutic agent can be translated, synthesized, ligated or otherwise produced as a single molecule which has both aC and therapeutic agent functions. Examples of therapeutic agents which can be coupled to aC for immunotherapy include, but are not limited to, bioresponse modifiers, drugs, radioisotopes, lectins, and toxins. Bioresponse modifiers include lymphokines which include, but are not limited to, tumor necrosis factor, interleukins 1, 2, and 3, lymphotoxin, macrophage activating factor, migration inhibition factor, colony stimulating factor, and interferon. Interferons with which aC can be labeled include u-interferon, P-interferon, and y-interferon (IFN-y) and their subtypes.
In using radioisotopically conjugated aC for immunotherapy, certain isotypes may i be more preferable than others depending on such factors as leukocyte distribution as well as isotype stability and emission. If desired, the malignant cell distribution can be 20 evaluated by the in vivo diagnostic techniques described below. Depending on the malignancy, some emitters may be preferable to others. In general, alpha and beta particleemitting radioisotopes are preferred in immunotherapy. For example, if an animal has solid tumor foci, as in a carcinoma, a high energy beta emitter capable of penetrating i several millimeters of tissue, such as 90 Y, may be preferable. On the other hand, if the i 25 malignancy consists of simple target cells, as in the case of leukemia, a short range, high energy alpha emitter, such as 2 12 Bi, may be preferable. Radioisotopes which can be bound to the antigen binding fragments of the invention for therapeutic purposes include, but are not limited to, 1 25 131 9, Cu 2 12 Bi 211At, 22b, 47 Sc, and "Re.
Lectins are proteins, usually isolated from plant material, which bind to specific sugar moieties. Many lectins are also able to agglutinate cells and stimulate lymphocytes.
S 15 However. ricin is a toxic lectin which has been used immunotherapeutically. This is preferably accomplished by binding the alpha-peptide chain of ricin, which is responsible for toxicity, to the antibody molecule to enable site specific delivery of the toxic effect.
Toxins are poisonous substances produced by plants, animals, or microorganisms that, in sufficient dose, are often lethal. Diphtheria toxin is a substance produced by Corynebacterium diphtheria which can be used therapeutically. This toxin consists of an alpha and beta subunit which under proper conditions can be separated. The toxic A chain component can be bound to an antibody and used for site specific delivery to a neoplastic cell.
Thus, for example. aC can be used in combination with alpha-interferon. This treatment modality enhances Mab targeting of melanomas by increasing the expression of Mab reactive antigen by the melanoma cells. Greiner et al. (1987) Science 235:895.
Alternatively, aC could be used, for example, in combination with IFN-y to thereby activate and increase the expression of Fc receptors by effector cells which, in turn, results in an enhanced binding of the antigen binding fragments to the effector cell and killing of target malignant cells. Those of skill in the art will be able to select from the various biological response modifiers to create a desired effector function which enhances the efficacy of aC.
When uC is used in combination with various therapeutic agents. such as those described herein, the administration of both usually occurs substantially contemporaneously. The term "substantially contemporaneously" means that they are administered reasonably close together with respect to time. Usually, it is preferred to administer the therapeutic agent before aC. For example, the therapeutic agent can be administered I to 6 days before aC. The administration of the therapeutic agent can be daily, or at any other suitable interval, depending upon such factors, for example, as the nature of the malignancy, the condition of the patient and half-life of the agent.
Using aC, it is possible to design combination therapies. It may be desirable to administer a therapeutic agent, or agents, prior to the administration of aC in combination with effector cells and the same, or different, therapeutic agent or agents. For example, 3 e 25 patients can be treated by first administering IFN-y and interleukin-2 (11-2) daily for 3 to days, and on day 5 administering aC in combination with effector cells. IFN-y, and 11-2.
The present invention also encompasses the use of liposomes with membrane bound aC to specifically deliver the liposome to the area of the tumor or neoplastic cells expressing C antigen. These liposomes can be produced such that they contain, in addition to aC, such immunotherapeutic agents as those described above which would then be released at the site of malignancy. Wolff et al. (1984) Biochem. Biophys. Acta 802:259.
Another such delivery system described by Brown et al. (1994) Virology 198:477-488; and Miyamura et al. (1994) Proc. Nail. Acad Sci. USA 91:8507-8511 utilizes chimeric parvovirus B19 capsids for presentation of the antigen binding fragments. Such chimeric systems are encompassed for use in the claimed methods.
The dosage ranges for the administration of aC are those large enough to produce the desired effect in which the symptoms of the malignant disease are ameliorated without causing undue side effects such as unwanted cross-reactions. anaphylactic reactions, and the like. Generally, the dosage will vary with the patient's age, condition, sex and extent of the disease and can be determined by one of skill in the art. The dosage can be adjusted by the individual physician in the event of any complication. Dosage can vary from about 0.1 mg/kg to about 2000 mg/kg, preferably about 0.1 mg/kg to about 500 mg/kg, in one or more dose administrations daily, for one or several days. Generally, when aC are administered conjugated with therapeutic agents, lower dosages, comparable to those used for in vivo immunodiagnostic imaging, can be used.
Therapeutic compositions of aC can be administered by injection or by gradual perfusion. The aC antigen binding fragments can be administered intravenously, intraperitoneally, intramuscularly, subcutaneously, intracavity, intrathecally or transdermally, alone or in combination with effector cells.
Another method of administration is intralesionally, for instance by direct injection directly into the tumor. Intralesional administration of various forms of immunotherapy to cancer patients does not cause the toxicity seen with systemic administration of immunologic agents. Fletcher et al. (1987) Lymphokine Res. 6:45; Rabinowich et al.
see.
0.00 0000 90640 99** 0 9.
(1987) Cancer Res. 47:173; Rosenberg et al. (1989) Science 233:1318; and Pizz et al.
(1984) Int. J. Cancer 34:359.
uC is particularly suitable for use in treating and imaging brain cancer. When the site of delivery is the brain, the therapeutic agent must be capable of being delivered to the brain. The blood-brain barrier limits the uptake of many therapeutic agents into the brain and spinal cord from the general circulation. Molecules which cross the blood-brain barrier use two main mechanisms: free diffusion; and facilitated transport. Because of the presence of the blood-brain barrier, attaining beneficial concentrations of a given therapeutic agent in the CNS may require the use of specific drug delivery strategies.
Delivery of therapeutic agents to the CNS can be achieved by several methods.
One method relies on neurosurgical techniques. In the case of gravely ill patients, surgical intervention is warranted despite its attendant risks. For instance, therapeutic agents can be delivered by direct physical introduction into the CNS, such as intraventricular, intralesional, or intrathecal injection. Intraventricular injection can be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Methods of introduction are also provided by rechargeable or biodegradable devices. Another approach is the disruption of the blood-brain barrier by substances which increase the permeability of the blood-brain barrier. Examples include intra-arterial infusion of poorly diffusible agents such as mannitol, pharmaceuticals which 20 increase cerebrovascular permeability such as etoposide, or vasoactive agents such as leukotrienes. Neuwelt and Rappoport (1984) Fed Proc. 43:214-219; Baba et al. (1991) J Cereb. Blood Flow Metab. 11:638-643; and Gennuso et al. (1993) Cancer Invest. 11:638- 643.
Further, it may be desirable to administer the compositions locally to the area in 25 need of treatment: this can be achieved by, for example, local infusion during surgery, by injection, by means of a catheter, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as silastic membranes, or fibers. A suitable such membrane is Gliadel@ provided by Guilford Pharmaceuticals Inc.
Another method involves pharmacological techniques such as modification or selection of the cC to provide an analog which will cross the blood-brain barrier.
Examples include increasing the hydrophobicity of the molecule, decreasing net charge or molecular weight of the molecule, or modifying the molecule, such as to resemble one normally transported across the blood-brain barrier. Levin (1980) J. Med Chem. 23:682- 684; Pardridge (1991) in: Peptide Drug Delivery to the Brain: and Kostis et al. (1994) J.
Clin. Pharmacol. 34:989-996.
Encapsulation of aC in a hydrophobic environment such as liposomes is also effective in delivering drugs to the CNS. For example, WO 91/04014 describes a liposomal delivery system in which the drug is encapsulated within liposomes to which molecules have been added that are normally transported across the blood-brain barrier.
Another method of formulating aC to pass through the blood-brain barrier is encapsulation in cyclodextrin. Any suitable cyclodextrin which passes through the bloodbrain barrier can be employed, including, but not limited to, P-cyclodextrin, y-cyclodextrin S 15 and derivatives thereof. See generally, U.S. Patent Nos. 5,017.566, 5,002,935 and 4,983,586. Such compositions can also include a glycerol derivative as described by U.S.
Patent No. 5,153,179.
'Yet another method takes advantage of physiological techniques such as conjugation of aC to a transportable agent to yield a new chimeric transportable aC. For 20 example, vasoactive intestinal peptide analog (VIPa) exerts its vasoactive effects only after conjugation to a Mab to the specific carrier molecule transferrin receptor, which facilitates the uptake of the VIPa-Mab conjugate through the blood-brain barrier. Pardridge (1991); and Bickel et al. (1993) Proc. Natl. Acad. Sci. USA 90:2618-2622. Several other specific transport systems have been identified, these include, but are not limited to, those for 25 transferring insulin, or insulin-like growth factors I and II. Other suitable, non-specific carriers include, but are not limited to, pyridinium, fatty acids, inositol, cholesterol, and glucose derivatives. Certain prodrugs have been described whereby, upon entering the central nervous system, the drug is cleaved from the carrier to release the active drug. U.S.
Patent No. 5,017,566.
:20 5* Suitable subjects include those who are suspected of being at risk of a pathological effect of any neoplasia. particularly carcinoma, are suitable for treatment with the pharmaceutical compositions of this invention. Those with a history of cancer are especially suitable. Suitable human subjects for therapy comprise two groups, which can be distinguished by clinical criteria. Patients with "advanced disease" or "high tumor burden" are those who bear a clinically measurable tumor. A clinically measurable tumor is one that can be detected on the basis of tumor mass by palpation, CAT scan, or X- Ray; positive biochemical or histopathological markers on their own may be insufficient to identify this population). A pharmaceutical composition embodied in this invention is administered to these patients to elicit an lanti-tumor response. with the objective of palliating their condition. Ideally, reduction in tumor mass occurs as a result, but any clinical improvement constitutes a benefit. Clinical improvement includes decreased risk or rate of progression or reduction in pathological consequences of the tumor.
A second group of suitable subjects is known in the art as the "adjuvant group".
These are individuals who have had a history of cancer, but have been responsive to another mode of therapy. The prior therapy may have included, but is not restricted to, surgical resection, radiotherapy, and traditional chemotherapy. As a result, these individuals have no clinically measurable tumor. However. they are suspected of being at risk for progression of the disease, either near the original tumor site, or by metastases.
This group can be further subdivided into high-risk and low-risk individuals. The subdivision is made on the basis of features observed before or after the initial treatment.
These features are known in the clinical arts, and are suitably defined for each different cancer. Features typical of high risk subgroups are those in which the tumor has invaded neighboring tissues, or who show involvement of lymph nodes.
Another suitable group of subjects is those with a genetic predisposition to cancer but who have not yet evidenced clinical signs of cancer. For instance, women testing positive for a genetic mutation associated with breast cancer, but still of childbearing age, may wish to receive tC treatment prophylactically to prevent the occurrence of cancer until it is suitable to perform preventive surgery.
go S *o oo oo.o..
A pharmaceutical composition embodied in this invention is administered to patients in the adjuvant group, or in either of these subgroups, in order to elicit an anticancer response. Ideally, the composition delays recurrence of the cancer, or even better.
reduces the risk of recurrence improves the cure rate). Such parameters mav be determined in comparison with other patient populations and other modes of therapy.
Of course, crossovers between these two patient groups occur, and the pharmaceutical compositions of this invention can be administered at any time that is appropriate. For example, aC therapy can be conducted before or during traditional therapy of a patient with high tumor burden, and continued after the tumor becomes clinically undetectable. uC therapy can be continued in a patient who initially fell in the adjuvant group, but is showing signs of recurrence. The attending physician has the discretion to determine how or when the compositions of this invention are to be used.
Various compounds and compositions of this invention have other clinical indications, of which the following section provides only a survey.
One indication is the treatment of cells ex vivo. This may be desirable for experimental purposes, or for treatment of an individual with a neoplastic disease. In one example. aC is administered to a culture of cells, such as peripheral blood cells obtained from a donor, or a suitable cell line. About 0.5 to 2 pg/mL of H 1 is an effective dose for 0 this purpose. In a second example, donor cells are genetically altered with an expression 20 vector of this invention, to provide for ongoing secretion of aC after administration of the cells to the recipient.
The present invention further encompasses methods for in vivo detection of cancer cells. A diagnostically effective amount of detectably labeled aC is given to the subject in need of tumor imaging. The term "diagnostically effective" means that the amount of 25 detectably labeled aC is administered in sufficient quantity to enable detection of the neoplasia.
The concentration of detectably labeled aC which is administered should be ,i sufficient such that the binding to those cells having C antigen is detectable compared to the background. Further, it is desirable that the detectably labeled aC be rapidly cleared from the circulatory system in order to give the best target-to-background signal ratio.
As a rule. the dosage of detectably labeled aC for in vivo diagnosis is somewhat patient-specific and depends on such factors as age, sex, and extent of disease. The dosage of aC can vary from about 0.01 mg/m 2 to about 500 mg/m-, preferably 0.1 mg/m 2 to about 200 mg/m most preferably about 0.1 mg/m 2 to about 10 mg/m 2 Such dosages may vary, Sjur exampie, depending on number of injections given, tumor burden, and other factors known to those of skill in the art. For instance, tumors have been labeled in vivo using cyanine-conjugated Mabs. Ballou et al. (1995) Cancer hnmunol. Immunother. 41:257-263.
For in vivo diagnostic imaging, the type of detection instrument available is a major factor in selecting a given radioisotope. The radioisotope chosen must have a type of decay which is detectable for a given type of instrument. Still another important factor in selecting a radioisotope for in vivo diagnosis is that the half-life of the radioisotope be long enough so that it is still detectable at the time of maximum uptake by the target, but short enough so that deleterious radiation with respect to the individual is minimized. Ideally, a radioisotope used for in vivo imaging lacks a particle emission, but produces a large number of photons in the 140-250 keV range, to be readily detected by conventional gamma cameras. For imaging, doses of 'In-HI 1-scFv (for instance. 2 mg of scFv labeled with 5 mCi of' 'Indium) the range administered is about 0.01 mg to 20 mg, more Spreferably about 0.1 -10 mg and even more preferably about 1-5 mg per patient.
For in vivo diagnosis. radioisotopes can be bound to aC either directly or indirectly S. 20 by using an intermediate functional group. Intermediate functional groups which are often used to bind metallic ion radioisotopes to immunoglobulins are the bifunctional chelating agents such as diethylene triaminepentacetic acid (DTPA) and ethylenediaminetetraacetic acid (EDTA) and similar molecules. Typical examples of metallic ions which can be bound to aC are In, 9Ru, 6Ga, 6 Ga, 7As, 9Zr, 0Y, and :25 aC can also be labeled with a paramagnetic isotope for purposes of in vivo diagnosis, as in magnetic resonance imaging (MRI) or electron spin resonance (ESR). In general, any conventional method for visualizing diagnostic imaging can be utilized.
Usually, gamma and positron emitting radioisotopes are used for camera imaging and paramagnetic isotopes for MRI. Elements which are particularly useful in such techniques
S
S.
20
*S
S
555.
25 include 15 7 Gd, 5 5 Mn, 162Dy, 52Cr and 56Fe. aC can also he labeled with a fluorescent dye for the purpose of in vivo diagnosis.
aC can also be used to detect neoplasias using in viro assays. Samples are taken from the patient and subject to any suitable immunoassay with aC to detect the presence of C antigen. This is particularly useful in detecting lymphomas and leukemias where the tumor cells bearing C antigen are circulating in the patient's bloodstream.
t(xC can also be used to monitor the course of amelioration of malignancy in an individual. Thus, by measuring the increase or decrease in the number of cells expressing C antigen or changes in the concentration of C antigen present in various body fluids, it is possible to determine whether a particular therapeutic regimen aimed at ameliorating the malignancy is effective.
The present invention encompasses kits containing aC. Diagnostic procedures using aC can be performed by diagnostic laboratories, experimental laboratories, practitioners, or private individuals. The clinical sample is optionally pre-treated for enrichment of the target being tested for. The user then applies a reagent contained in the kit in order to detect the changed level or alteration in the diagnostic component.
Each kit comprises aC used for detecting C antigen in the sample. Optionally, the reagent may be conjugated with a label to permit detection of any complex formed with the target in the sample. In another option, a second reagent is provided that is capable of combining with the first reagent after it has found its target and thereby supplying the detectable label. For example, labeled anti-mouse IgG may be provided as a secondary reagent for use with intact aC. Labeled avidin may be provided as a secondary reagent when the primary reagent has been conjugated with biotin.
The kits can be employed to test a variety of biological samples, including both liquid samples, cell suspensions and tissue samples. Suitable assays using aC that can be supplied in kit form include those described herein. Each reagent is supplied in a solid form or dissolved/suspended in a liquid buffer suitable for inventory storage, and later for exchange or addition into the reaction medium when the test is performed. Suitable packaging is provided. The kit can optionally provide additional components that are useful in the procedure. These optional components include, but are not limited to, 57 buffers. capture reagents, developing reagents, labels, reacting surfaces, means for detection, control samples, instructions, and interpretive information.
The foregoing description provides, inter alia, detailed methods for preparing H1 1, along with HI 1 encoding polynucleotides. HI 1 polypeptide fragments. and other Sderivatives. A prracttioner of ordinary tski!! in the art can "ractice embodiments oflthi invention by referring to the sequence data for H 1i, which is provided herein. The following examples are provided to illustrate but not limit the claimed invention.
EXAMPLE 1 Method of obtaining Mab HI 1 Mab NBGMI/HI I is a human monoclonal IgM antibody reactive against the following human tumor tissues and corresponding tumor cell lines: glioma. malignant melanoma, colon adenocarcinoma and breast adenocarcinoma. In vitro characterization of i Mab NBGMI/H I is shown in Example 2.
o* 15 Fusion of HI 1 was accomplished by fusing 8 x 10 6 peripheral blood lymphocytes obtained from a 64 year old male with a low grade glioma with the TM-H2-SP2 human myeloma cell line. The TM-H2-SP2 cell line is the immunoglobulin nonsecreting subline of the IgG(K) parental cell line TM-H2, a hypoxanthine guanine phosphoribosyltransferase (EC 2.4.2.8)-deficient derivative of an unknown human myeloma-like line selected in 0.8% methylcellulose for its resistance to 6-thioguanine (6 gg/mL) and failure to grow in hypoxanthine-aminopterin-thymidine medium. The karyotype of TM-H2-SP2 is 46±2.
XX.
The resultant viable hybridoma cells were split among 40 microwells at a density of 2 x 10 cells/mL and 0.2 mL/well. The frequency of outgrowth from fusion HI 1 was 12 of 25 40 potential hybridoma-containing wells. Outgrowth resulting from sustained growth is defined as prolonged growth with culture expansion for periods longer than 3 months; instances of hybridoma growth failure occurring later than 3 months post-fusion were not observed.
58 Screening of hybridoma clones was performed by antigen-capture enzyme-linked immunosorbent assay (ELISA) in microtiter plates using polyclonal anti-human IgM or IgG as coating antigen. A hybridoma culture supernatant was positive if the measured optical density value exceeded the mean background level of a control culture supernatant by reater than iwo standard deviations.
Selection of hybridoma clone NBGMI/HI 1 was performed by cell-fixed ELISA.
Culture supernatants from 6 microtiter wells. which tested high for IgM or IgG secretion, were screened against previously attached and fixed human tumor cell lines: Glioblastoma (SKMG-1 and D-54MG); melanoma (A-375): and colon adenocarcinoma (SK-CO-I). A hybridoma supernatant was considered to be positive if the measured O.D. value exceeded the mean background level of control culture supernatants by greater than two standard deviations. Mab produced by hybridoma NBGM1/H11 continues to be reactive against these tumor cell lines. The "H 11" antibodies are IgM(),.
Characterization of the hybridoma NBGM I/H 11 seed bank was performed by Microbiological Associates (Rockville, MD). The cells tested negative for bacterial .I and fungal contamination, mycoplasma contamination. HIV-1 and HIV-2 antigens and HTLV-1 and HTLV-2 antigens.
The methods used for the characterization of Mab NBGM 1/H 11 include: antigencapture ELISA. antigen ELISA. cell-fixed ELISA. flow cytometry, immunoperoxidase staining of human tumor cell lines and immunohistochemistry of human tumor and normal tissues (see following examples).
Binding characteristics of this human Mab to human tumor cell lines as determined by flow cytometry, immunoperoxidase staining, cell-fixed ELISA and antigen ELISA tumor cell freeze-thaw extracts) are presented below.
25 EXAMPLE 2 Binding of Mab HI I to Human Glioblastoma (SKMG-I) and Melanoma (A375) Cell Lines by Flow Cvtometric Analysis In order to determine the binding of Mab HI 1 to tumor cells, tumor cells growing in T-flasks were detached by incubation with PBS-EDTA. Cells were collected by low speed centrifugation, washed with ice-cold PBS-l% FBS. centrifuged and the supernatant aspirated. The cell pellet was resuspended in culture medium spiked with one of the following: a control human melanoma IgM; hybridoma NBGM/HI 1 culture supernatant; or PBS containing purified Mab H I1; and incubated on ice for 30 minutes. After incubation, the cells were collected by centrifugation. washed by resuspension in PBS-FBS and centrifuged. The cell pellet was then incubated for 30 min. with FITC-conjugated goat anti-human IgM. After incubation, the cells were washed with PBS-FBS. Finally, the cells were resuspended in PBS-FBS propidium iodide (PI) was added and the cells washed.
PI-positive and FITC-positive cells were analyzed by flow cytometry.
The results of the flow cytometric analyses are shown in Figs. 2. 3 and 4. These results indicate that crude and purified forms of Mab HI 1 bind to a cell surface-associated antigen(s) expressed on live human tumor cell lines, including glioblastoma. melanoma, breast adenocarcinoma and colon adenocarcinoma.
EXAMPLE 3 15 Binding of Mab HI I to Freeze-Thaw Extracts of Human Tumor Cell Lines by ELISA Analysis In order to determine the ability ofHI1 to bind specifically to human tumor antigen(s), ELISA plates were coated with human tumor cell extracts prepared by repeated freezing and thawing of glioblastoma (SKMG-1), breast adenocarcinoma (BT-20. MB-468 and MB-453), colon adenocarcinoma (SK-CO-1 and HT-29) cells.
The coated ELISA plates were incubated for 16-18 hours at 2-8C. The plates were blocked with PBS-3% BSA for 1 hr at room temperature. Then the plates were incubated with either Mab HI I in PBS or control IgM in PBS or culture medium for 2 hrs at room temperature. The plates were washed and incubated with biotinylated anti-human IgM 25 followed by incubation with biotinylated anti-human IgM followed by incubation with streptavidin-conjugated alkaline phosphatase for 1 hr. After washing, p-nitrophenyl phosphate substrate was added to each plate and, after incubation, the plates were read at 405 nm in an ELISA plate reader.
The binding of Mab HI 1 to the tumor cell extracts is shown in Figs. 5 and 6. These results indicate that Mab H 1 binds to tumor cell extracts prepared from glioblastoma, breast adenocarcinoma and colon adenocarcinoma cells in a dose-dependent manner.
EXAMPI. 4 Binding of Mab HI 1 to Human Tumor Cells Determined by Immunoperoxidase Staining In order to determine immunorcactivity of HI 1. the following experiment was performed. Tumor cells were grown in 24-well plates on coverslips for 48-96 hrs. The cells were washed with PBS. fixed with formaldehyde and incubated with 5% normal goat serum on PBS for 30 min. After washing, the cells were incubated for 2 hrs with either hybridoma NBGM1/HI 1 culture supcrnatants or purified Mab HI1 (10 uJg/mL) in PBS or culture medium spiked with control human myeloma IgM (10 pg/mL) for 2 hrs. The cells were then washed and incubated with anti-human IgM conjugated to HRP. Finally, the cells were washed, incubated with DAB substrate to visualize Mab HI 1 binding, counter- 15 stained with hematoxylin and mounted in GVA.
The results of the immunoreactivity of Mab H 11 are shown in Table 3 where reactivity is indicated as negative weak positive positive strong positive These results indicate that, as determined by immunoperoxidase staining, the epitope recognized by Mab HI 1 is expressed by a number of different types of human tumor cells and cell lines.
TAI3LE3 CELL LINES/TYPE OF TUMOR
REACTIV'ITY
Control 1gM Mab HI 1 HUMAN GLIOBLASTOMA SKMG I
I
U-1 18 MG U-87 MG HUMNAN MAIGINANT MrLNO A A-375 SK-MEL-S HUMAN COLON ADENOCARCINOMA SK-CO-I HUMAN BREAST ADENOCARCINOMA MG-468 MB-453 BT-474 HUMAN KIDNEY ADENOCARCINOMA SW-839
I
HUMAN OSTEOGENIC SARCOMA SAOS-2 HUMAN OVARY ADENOCARCrNOMA SK-OV-3 Bindina of Mob H I I to Human Tumor ell Lines Determined by Cell-Fixed-ELISA The binding of HlII to human tumor cells and cell lines was also determined by cellfixed ELISA. Growing tumor cells were detached from the T-flask surface by incubating with EDTA-PBS. Cells were collected by centrifugation, washed with PBS, resuspended in culture medium, counted, and 50 gl of cell suspension containing 5,000-10,000 cells placed in each well of 96-well ELISA plates. After allowing the cells to attach to the plates, the culture superniatants were removed and the plates were blocked with PBS-BSA. The cells were then incubated with different concentrations (1 -20 gig/mL) of either Mab H I I or control human 62 myeloma IgM for 2 hrs. After incubation, the plates were washed, incubated with biotinconjugated goat anti-human IgM. washed again and incubated with streptavidin-conjugated alkaline phosphatase. Finally, the plates were washed, incubated with p-nitrophenyl phosphate substrate and read at 405 nm in an ELISA plate reader.
Results of the reactivity of Mab HI I to human tumor cell lines by cell-fixed ELISA are shown in Table 4 and Figure 7. In Table 4. Control IgM 10 pg/mL and HI1 10 j.g/mL were used for testing the reactivity, and values are given as absorbance at 405 nm standard deviation. These results indicate that: 1) Mab HI 1 reacts strongly with glioblastoma cells (SKMG-I). even at a low concentration of 1 gg/mL, whereas control IgM at 20 gg/mL does not react with SKMG-I cells: and 2) Mab H 11 recognizes the tumor antigen(s) present on numerous tumor cell lines (breast adenocarcinoma. colon adenocarcinoma, malignant melanoma, neuroblastoma, glioblastoma. lung adenocarcinoma, small cell lung carcinoma and prostate adenocarcinoma). The degree for Mab reactivity varies both with the type of cancer and the tumor cell lines. The reactivity of Mab H 1 for cancer and tumor cells was between 15 three and ten times greater than that of the control IgM.
e
*S
*o 63 TABLE 4 Cell lines/Tumor Type Reactivity at 405 nm) Control 1gM Mab IIl Human Glioblastoma SKMG- I D-54-NMG U-87MG Neu ro blastom a
SK-N-SH
SK-N-MC
Malignant Melanoma SK-MEL-28 Breast adenocarcmnoma MB-453 MB-468 SK-BR-3 T47 D BT-4 74 be p cc C.
C
cc..
C.
C.
C.
C
C C p* Lung adlenocarcinoma SW-900 SK-LU- I A-427 Small cell lung carcinoma NCI-H69 NCI..H82 Colon adenocarcinoma SK-Co- I HT-29 Prostate adenocarcinoma PC-3 DU- 145 Kidney adenocarcinoma SW-83 9 Osteogenic sarcoma SAOS-2 U-2 OS Bladder cell carcinoma T-24 Ovarian adenocarcinoma SKOOV-3 Larynx carcinoma HEP -2 Normal human fibroblast GM-8333 0.21 ±0.01 0.13 0.02 0.13 0.02 0.14 0.02 0.17 0.03 0.18 ±0.03 0.19 0.03 0.68 0.18 0.60 0.03 0.60 0.03 0.58 0.01 0.57 0.02 0.61 0.03 0.20 0.02 0.19 0.02 0.22 0.01 0.25 0.04 0.20 0.09 0.27 0.03 0.37 0.02 0.17 0.01 0.15 0.01 0.2 ±0.01 0.24 ±0.02 0.13 ±0.04 0.13 ±0.01 0.12 ±0.01 0.25 ±0.01 0. 13 *0.0 1 0.95 0.06 0.43 0.07 0.60 0.01 0.96 0.06 1.00 0.05 1.42 0.04 1.79 0.05 2.85 0. 14 2.39 0.10 2.14 0.13 2.13 0.04 2.07 0.13 2.20 0. 17 0.68 0.10 0.57 0.07 0.88 0.07 1.42 0.20 1.16 0.13 0.98 ±0.11 1.78 0.20 0.60 0.01 0.52 0.01 1.43 0.01 1.22 0.07 1.93 0.05 1.25 0.03 1.14 0.02 1.25 ±+0.01 0.39 0.01 64 EXAMPLE 6 Immunoanatomic Distribution and Immunopathologic Analysis of HI 1 Immunohistochemistry was used to determine HI 1 specificity for micro-anatomical detail and heterogeneity in tissues and tumors. Limitations of this technique include possible false negative results due to low levels of expression of the molecule under study, as well as false positive results (cross-reactivity) due to antibody-binding to similar epitopes or epitopes shared by other antigens. To address these limitations, this study was carried out at the highest concentration of antibody that did not show non-specific binding.
This allowed for detection of all levels of cross-reactivity in different tissues. Also, fixation analysis established the best combination of antigenic staining intensity and morphological preservation. The present example presents results obtained from IMPATH Inc., New York. retained to study the cellular specificity and antigen expression of H 1I, on a selected panel of crvostat-cut frozen sections of normal and tumor tissues. The study Sused an indirect immunoperoxidase technique.
15 Histologically normal human tissues were obtained from surgical and autopsy specimens. These fresh tissues were embedded in OCT (Miles Laboratories, Inc., Naperville, IL) in cryomolds. snap-frozen in isopentane, cooled by liquid nitrogen. The tissues from IMPATH's frozen tissue bank were cut at 5 microns, placed on poly-L-lysine coated slides, air-dried, and stored at 20 HI 1, received on wet ice and stored at 2-8 0 C, was supplied non-biotinylated at a concentration of 200 .g/mL, total volume of 3.0 mL. A human myeloma IgM (Pierce Cat.
#31146), also supplied by Novopharm, was used as the negative control. Both antibodies o were diluted in phosphate buffered saline to the same working concentrations dictated by titration analysis of antibody H 1. The peroxidase-labeled secondary antibody was a goat 25 anti-human IgM (American Qualex, San Clemente, CA, lot #A112PN) diluted in PBS to 1:500.
Immunoperoxidase Techniques: Immunohistochemical studies were performed using an indirect immunoperoxidase method. The cryostat cut sections were removed from the -70 0 C freezer, air-dried and fixed according to the fixation protocol (fixation details, provided below). Tissue sections were blocked for 10 minutes with 5% normal goat serum diluted in PBS, then incubated with the primary antibody overnight at 4°C.
Slides were washed in PBS, followed by a wash with 0.5% Tween/PBS solution, then PBS again. Endogenous peroxidase activity was blocked with a 30 minute 3% hydrogen peroxidc/methanol incubation, followed by 3 washes of PBS. The sections were then incubated with goat anti-human IgM (peroxidase-labeled) secondary antibody for minutes, at room temperature, and washed in PBS as described above.
The peroxidase reaction was visualized by incubating tissue sections for minutes with 3.3-diaminobenzidine-tetrahydrochloridc (DAB) (Sigma Chemical Co., St.
Louis. MO). Tissue sections were thoroughly washed. counterstained with a modified Harris hematoxylin (Fisher Scientific. Fairlawn, NJ) dehydrated through graded alcohols, cleared in xylene. and coverslipped. Tissues that demonstrated high levels of background staining with the negative control antibody were repeated with more extensive washing.
Human breast carcinoma (F95-036). supplied by IMPATH, was the positive control for HI 1. Negative controls substituted the primary test antibody with purified human myeloma IgM.
The purpose of the fixation analysis was to establish the conditions which provide i" the optimal combination of antigenic staining intensity and morphological preservation.
The positive control tissue was tested with five fixation protocols. including no fixation.
20 The fixation protocols tested were 10% neutral buffered formalin (23-25 0 acetone (2methyl/acetone (1:1 V/V, 2-8 0 C) and 95% ethanol (23-25 0 For this study, S neutral buffered formalin (NBF) gave optimal results for H11.
Using 10% NBF as the fixative, serial antibody dilutions (20.0 tg/mL to 0.1 Stg/mL) were tested on the positive control, human breast carcinoma. A concentration of 25 10.0 ug/mL of antibody HI 1 gave optimal results-maximum staining intensity without significant background staining of the negative control.
The results obtained are depicted in Tables 5 and 6. Table 5 depicts H11 reactivity on normal tissues and Table 6 depicts HI 1 reactivity on human tumors.
66
TABLES
Tissue Adrenal 13ladder Bone Marrow Brain Breast Cervix Esophag~us Eye Heart Kidnev Laree Intestine Liver Lung Lymph Node Muscle Ovary Pancreas Parotid Pituitary Prostate Skin Small intestine Spinal cord Spleen Stomach Testis Thymus Thyroid Tonsil Uterus
WBC
Tested Positive/TFotal 0/3 0/3) 1/3 0/3) 0/3 0/3 0/3) 0/3 0/3 0/3 0/3 0/3 0/3 0/3 0/3 0/2 0/3 0/3 0/I 0/3 0/3 0/3 0/3 0/3 0/3) 0/3 0/3 0/3 1/3 0/3 0/3 Range of Reactivity (03+ 0 0 1+ 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
I+
0 0 67 TABLE 6 Tumor Breast carcinoma Colon carcinoma Glioma Gastric carcinoma Lung adenocarcinoma Lung squamous carcinoma Lung small cell carcinoma Lymphoma Melanoma Ovarian carcinoma Prostate carcinoma Sarcoma Tested Positive/Total 2/3 3/3 4/6 3/3 3/4 3/3 1/2 8/8 3/3 3/3 3/3 0/3 of Tumor Cells Staining 30-90 40-70 30-90 30-50 10-70 10-95 30 10-95 20-95 20-30 20-95 0 Range of Reactivity 1-3+ 1-2+ 1-2+ 1-2+ 1-2+ 1-3+ 1+ 1-3+ 1-2+ 1-3+ 1-2+ 0 9* The results obtained indicate that weak to strong reactivity was observed in over 70% of the positive control sample. The antigen recognized by HI 1 has a restricted pattern of distribution. HI 1 was largely unreactive with normal human tissues tested in the IMPATH system. All simple epithelial cells, as well as the stratified epithelia and squamous epithelia of different organs were found to be unreactive. Reactivity was also not seen in neuroectodermal cells, including those in the brain, spinal cord and peripheral nerves. Mesenchymal elements such as skeletal and smooth muscle cells, fibroblasts. and endothelial cells were negative. Tissues of lymphoid origin including bone marrow, lymph node, spleen, and thymus were largely unreactive with antibody HI 1.
Weak reactivity was observed in rare cells in one specimen of bone marrow and in the germinal centers of one of three specimens of tonsil tested.
Positive immunoreactivity was observed in almost all specimens of tumor tested including breast, colon, glioma, gastric, lung (adeno, squamous, and small cell), lymphoma, melanoma, ovarian, and prostate. Reactivity was seen in 10% to greater than of the tumor cells present in these specimens; staining intensity ranged from weak to strong Antibody H11 was, however, unreactive with all three specimens of sarcoma tested. Some but not all normal counterparts of the tumor cells, when present in the specimens, were reactive with H 11. A few normal cells present in breast, gastric and 68 prostate carcinoma were reactive with antibody HI 1. The large granular cells that were reactive with antibody HI 1 are believed to be inflammatory cells of the eosinophile-mast cell lineage.
In summary, antibody HI 1 is largely unreactivc with normal human tissues with the exception of some normal tissues present in tumors. The HI I antibody detects an antigen that is expressed in almost all of the tumors tested.
EXAMPLE 7 1I I1 Cloning. Expression and immunologic reactivity In order to determine the ability of HI -scFv antibody fragments to bind specifically to cancer cells, the following experiments were performed.
The single chain antibody constructs were made by the following procedure.
Primers specific to the 5' and 3' ends of the HI 1 kappa and mu V regions were synthesized on an Applied Biosystems DNA synthesizer. All the primers contained a restriction endonuclease site for cloning. Primers 5 and 6 also contained additional nucleotides that encode a (SGGGG) 3 linker. The primers used are listed in Table 7 where the restriction endonuclease site introduced is underlined.
*o *o S* 69 TABLE 7 Primer Sequence Site Introduced 1. TATGAAGACACCAGGCCGATATTGTGTTGACGCAG Bbsl (SEQ ID NO:7) 2. TATCCGGATGCAGCCACAGTTCGTTT (SEQ ID NO:8) BspEl 3. TATTCGGACAGGTGCAGCTGGTGGAG (SEQ ID NO:9) BspEl 4. TATGGATCCTGAGGAGACGGTGACCGT (SEQ ID NO:10) BamHI TATATATCCGGAGGTGGTGGATCAGGTGGAGGTGGCTC BspEl CCAGGTGCAGCTGGTGGAGTCT(SEQ ID NO:11) 6. ACCTCCGGAACCGCCACCGCCAGAGACAGATGGTGCA BspEl GCCACATTC (SEQ ID NO:12) PCR reactions were carried out using primers 1 and 2 for the kappa dimer, primers 3 and 4 for the mu dimer, primers I and 6 for the kappa monomer and primers 4 and 5 for the mu monomer. The PCR fragments were then purified and digested with their respective restriction endonucleases. The coding nucleotides are depicted in SEQ ID NOS: 13 and 16 and the complementary nucleotides are depicted in SEQ ID NOS: 15 and ,i 18. respectively.
The expression vector pSJFI containing a ribosome binding site, OmpA signal peptide sequence, c-myc (9E10) detection tag and histidine tail (See Figure 8) was prepared by cutting with Bbsl and BamHl. The monomer and dimer constructs were assembled by ligating the respective kappa and mu fragments into pSJFI and transforming them into competent TG1 E. coli. Resulting colonies were screened by colony PCR and restriction endonuclease digests to confirm the correct size inserts and the sequences were 15 verified by dideoxy fluorescent sequencing.
Transformed TGI containing either the HI 1 monomer or dimer expression plasmid were shaken at 26 0 C for 24 hours followed by the addition IPTG to a final concentration of 0.1 pM. The cells were incubated for a further 16 hours and then harvested by centrifugation. The periplasmic proteins containing the HI 1 antibody were released by treatment with sucrose buffer (25% sucrose. 1 mM EDTA. 10 mM Tris pH 8.0) followed by ice cold shock buffer (10 mM Tris 8.0, 0.5 mM MgCI,). Expression was verified by polyacrylamide gel electrophoresis and Western Blotting. The antibody was purified using anickel-charged column (Pharmacia HiTran rhePlat1 n mn. rn e .id antibody che *atinaz tf'hmfl' t1 uju ii'u was eluted with an increasing gradient ofimidazolc. The purified antibody was dialyzed against PBS/0.02% sodium azide and concentrated to 0.5 mg/mL.
The antigenic similarities between Mab HI 1 and HI 1-scFv were also determined by cell fixed ELISA. ELISA plates coated with A375 cells were incubated with Mab H11.
control IgM. HI 1-scFv or control BGA-scFv followed by incubation with rabbit antihuman IgM antibody or rabbit anti-scFv antibody as appropriate. The detection was by goat anti-rabbit IgG-horse radish peroxidase followed by substrate. The results, shown in Figure 9, demonstrate a high affinity of both H 1 IgM and H 1 -scFv, and a low affinity of both the control IgM and BGA-SL-6.
In order to determine the specificity of biotinylated HI l-scFv relative to a biotinylated control scFv, the following experiment was performed. Human tumor cells were fixed to ELISA plates and incubated with either biotinylated HI 1-scFv or S. biotinylated BGA scFv (control) as described above.
Biotinylated HI 1-scFv also demonstrated a much greater affinity (between 8- and 50-fold) for tumor cell lines than the control in cell fixed ELISA. Data corresponding to a 20 concentration of 2.5 pg/mL of HI 1-scFv or BGA scFv is shown in Table 8 and Figure Figure 11 illustrates the portion of Table 8 related to the titration of reactivity of S biotinylated H1 l-scFv for the binding to lymphoma cells Daudi, Ramos, CA-46 and .CCRF-CEM cells. At every concentration tested (1.25 to 10 Ag/mL), H 1-scFv demonstrated a high affinity for lymphoma cells, but BGA scFv did not.
ol° TABLE 8 Tumor Cell Lines Reactivity at 450 nm SD) Biotinviated BGA scFv, Biotin-HlI I-scFv Human Glioblastoma SKMG-1 0.01 0.01 0.56 0.04 U- 118MG 0.01 0.02 0.47 0.03 D-54MG 0.01 0.00 0.50 0.02 Neu ro biastoma SK-N-MC 0.01 0.00 0.50 ±0.02 Malignant Melanoma 0.02 0.01 0.61 ±0.04 A-375 0. 12 0.03 0.97 ±0.03 SK-MEL-28 0.02 0.00 0.71 ±0.04 Bircast adenocarcinoma T47D 0.02 0.00 0.64 0.03 MB-468 0.01 0.00 0.65 0.01 SK-BR-3 0.01 0.00 0.58 0.02 0.01 0.00 0.54 0.06 BT-474 0.01 0.00 0.60 0.01 Lung adenocarcinoma SW-9000 0.01 0.00 0.41 0.02 SK-LU- 1 0.01 0.00 0.45 0.03 A-427 0.01 0.00 0.40 0.05 Colon adenocarcinoma SK-Co-1 0.01 0.00 0.56 0.01 HT-29 0.01 0.00 0.53 0.06 LS17T 0.01 0.00 0.57 0.02 Osteogenic sarcoma SAOS-2 0.02 0.00 0.88 0.06 U-2 OS 0.02 ±+0.00 0.93 0.01 Bladder cell carcinoma T-24 0.02 0.01 0.97 0.05 Ovarian adenocarcinoma SK-OV-3 0.01 0.00 0.77 0.02 Larynx carcinoma HEP-2 0.02 0.00 0.08 0.04 Prostate carcinoma DU- 145 0.01 0.00 0.42 0.02 PC-3 0.01 0.00 0.36 ±0.01 Tumor Cell Lines Reactivity at 450 nm SD) Biotinylated BGA scFv Biotin-HI 1-scFv Small cell lung carcinoma NCI-H82 0.01 0.00 0.44 i 0.02 NCI-69 0.01 0.00 0.44 0.01 Lymphoma cell lines Chronic myclogenous leukemia K-562 0.02 0.00 0.65 0.00 Acute lymphoblastic Icukemia CEM 0.04 0.00 1.4 0.03 Burkitt Lymphoma CA-46 0.02 0.00 1.2 0.02 RAMOS 0.04 0.00 1.3 0.02 DAUDI 0.02 0.00 1.38 0.01 In order to verify the specificity of biotinylated HI 1-scFv for cancerous cells, the following experiment was performed. Malignant and normal tissue specimens were prepared and incubated with biotinylated H 11-scFv as described above.
The HI 1-scFv was used to stain sections of tumor and normal tissues. The results are depicted in Table 8 for normal tissues and Table 9 for tumor tissues.
Figure 12 depicts the relative fluorescence intensity of H1 l-scFv and control scFv to tumor cell lines.
The data in Table 9 demonstrate that biotinylated H 11-scFv generally does not react to normal tissues. Almost all of the normal tissues tested demonstrated no measurable reactivity, with only a weakly positive signal generated by normal pancreas and peripheral nerve tissues. In Table 9, -ve indicates no measurable activity and indicates weakly positive activity.
p p. .z TABLE 9 Normal Tissues Reactivity of Biotinviated H II-scFv Mg/mL) Cortex -ye DI easi -ye Colon -ve Heart -ye Liver -ye Lymph node -ye Prostate -ye Thyroid -ye Adrenal -ye Cerebellum -ve Lung -vc Pancreas +1- Peripheral Nerve Skin -ye Spleen -ye Smooth muscle -ye Stomach -ye aThymus -ye 74 TABLE Tissue Type Number of Percentage of Range of positives/ Total Tumor Cells Reactivity (0 -3) samples tested staining Breast carcinoma 27/31 40/60 1-3+ Colon carcinoma 23/26 80/100 1-3+ Melanoma 13/14 50/70 1-3+ Prostate carcinoma 17/20 20/70 1-2+ Cervix squamous 22/24 nd 1-2+ cell carcinoma Cervix 9/9 nd 1-2+ adenocarcinoma Kaposi Sarcoma 7/8 nd 1-2+ Benign Colon 0/2 0 0 nd: not determined The results presented in Table 10 indicate that positive staining was found in most breast (27/31) and colon (23/26) and prostate (17/20) carcinoma samples tested. Positive staining was found at 25 j.g/mL concentration of H 1 -scFv. Although the staining was predominantly detected in tumor cells, various degrees of reactivity were also found on stroma and adjacent tissue. The HI 1-scFv was also tested for its specificity for normal tissue. The results obtained are presented in Table 11 which summarizes the immunohistochemistry staining of H1 I-scFv with normal human tissue sections.
p TABLE 11
TB
1 Isue HI -scFv (25 gg/mL) 3B1 scFv (25 tg/mL) Adrenal Cerebellum Cortex Colon Breast Kidney Aorta Heart Liver Lung Lymph node Pancreas Pituitary Prostate Peripheral nerve Skin Spleen Small intestine Stomach St. muscle Thymus Thyroid S94-7474-2 (Colon Car. control) -(focal -(sweat gland -4- -(focal -(sweat gland 0**4 *44e 4 EXAMPLE 8 Reactivity of HI l-scFv to Live Tumor Cells as Determined by Flow Cvtometrv In order to test the reactivity of HI I-scFv to live tumor cells, cells from tumor cell lines were prepared for flow cytometry as described above in Example 2. Tumor cells were incubated with either biotinylated HI 1-scFv or control biotinylated scFv as described above at a protein concentration of 100 pg/mL or 200 pg/mL. The reactivity was determined as the mean fluorescence and positive cells. A biotinylated HI11-scFv was prepared as described above and at a protein concentration of either 100 ulg/mL or 200 ug/mL. The mean fluorescence and positive cells are shown in Table 12 where is biotinylated 3131 as control scFv; is biotinylated BGA SL-6 as control scFv; **is PBS FCS as control: and is Biotin-5B31 as control scFv.
Table 12 Mean Fluorescence I Posilive Cells Cell Line Protein Biotinylated JBiotinylated Biotinviated Biorinylated conc'n 313 1 scFv 4 HIl I-scFv 3B3I ScFv H11 l-scFv (pglmL) SK-BR-3 200 149 233 11 36 (Breast adcnocarcinoma) MB-468 200 144 156 9 11 (Breast adenocarcinoma) A-375 200 I 207 10 (Melanoma) A-375 200 161* 235 28 76 (Melanoma) LS-174T (Colon adeno 200 182 233 24 37 carcinoma) HT-29 (Colon adeno- 200 141 179 12 18 carcinoma) SKMG-1 100 148** 206 9 31 (Glioma) 100 189** 224 14 27 H9 100 185*** 145 20 13 (T cell Lymphoma) WI-38 (Human 100 293*** 2 55 26 diploid lung cells) 125 1 labeled H I1I -scFv demonstrated an affinity of binding (Ka) of 3 x 10 L/mol for LS I74T cells (specific binding shown in Figure 13). 1n-H II-scFv demonstrated an affinity of binding (Ka) of 3.6 x 10~ L/mol for A-3 75 cells and 1.4 x 10 9L/mol for SKMG I cells. The results are depicted in Figure 14. There were approximately 24,000 binding sites/cell for A-375 cells and approximately 5000 binding sites/cell for SKMG1.
4* *4 4 .4 4 4 *44* 4 4*4* 4* 4.
*4 .4 These results indicate that purified forms of Mab H I I bind to a cell surfaceassociated antigen(s) expressed on breast adenocarcinoma (SK-BR-3), glioblastomra (SKMG-l), and melanoma (A-375) lymphoma cell lines.
In order to further test the reactivity of biotinylated H I I -scFv to live lymphoma cells. cells from tumor cell lines were prepared and incubated with biotinylated H I Il-scFv at a protein concentration of either 100 pg/ml, or 200 l.±g/mL and analyzed by flow cytometry as described above in Example 2.The mean fluorescence and positive cells were measured by flow cytometry. The control for scFv binding was biotinylated BGA scFv. Results are showvn in Table 13.
Table 13 CELL SAM1PLES CONC. MEAN FLUORESCENCE POSITWE LINE (pg/mL) FLUORESCENCE INCREASE CELLS Burkittfs PBS 123 Lymphoma BIOTIN- 200 126 9' BGA scFv 100 133 14 CA-46 BIOT'N- 200 262 108 76 H I I-scFv 100 221 66 58 T cell PBS 150 6 lymphoma BIOTIN- 200 155 8 BGA scFv 100 149 7 H9 BIOTIN- 200 186 20 13 H II -scFv 100 171 15 9 Acute PBS 151 8 lymphoblast B[OTIN- 200 171 14 oid leukemia BGA scFv 100 159 BIOT[N- 200 231 35 34 CCRF-CEM H I I1-scFv 100 242 52 38 Burkitt's PBS 151 Lymphoma BIOTIN- 200 174 BGA scFv 100 169, 13 RAMOS BIOTIN- 200 423 j143 H I I-scFv 100 316 j87 67 0 0e *0 *0 0 0a* 000* a a *0 a.
000* a a ~.9a* 0 a a a.
a 00* 78 FXAMPT F 0 4;Bindinii, of Biotinvlated H I I -scFv to Human Tumor Cells Determined by Immunop~eroxidase Stainin In order to determine iminunoreactiv'ity of H I Il-scFv, the following experiment was performed. Tumor cells were grown in T-flasks and cytospins were prepared and incubated with biotinylated H I I -scFv or PBS to determine binding.
The results of the immunoreactivitv ofl 1-11-scFv are shown in Table 14 wvhere reacuivitv is indicated as negative weak positive positive or These results indicate that, as determined by immunoperoxidase staining, the epitope recognized by Mab H-II I is expressed by a number of different types of human tumor cells and cell lines.
Table 14 CELL LINES/TYPE OF TUMOR
REACTIVITY
PBS H! ISCF,, (P gh'mL) HUMAN GLIOBLA STOMA SKMG I U-87 MG HUMAN MALIGNANT MELANOMA A-375 +4 HUMAN COLON ADENOCARCINOMA SK-CO-1- HT-29 1 74T HfUMAN BREAST ADENOCARCTNOMA SK-BR-3 HUMAN LYMPHOMA CELL LINES U-937 Histocytic Lymphoma H9 T Cell Lvmphoma CEM Acute Lymphoblastoid leukemia MOLT-3 Acute Lymphoblastoid leukemia Promyelocytic lekei KG- I Acute myelogenous leukemia K-562 Chronic myelogenous leukemia GASTRIC CARCINOMA KATO III +4 HUMAN OSTEOGENIC SARCOMA SAOS-2 HUMAN OVARY ADENOCARCINOMA SK-OV-3 BLADDER CELL CARCINOMA :T-24 LARYNX CARCINOMA *Hep-2+ .EXAMPLE Reactivity of Recornbinantlv Produced HI I 19-GI H I I IgG I was produced in Chinese Hamster Ovary (CHO) cells as follows.
Several vectors containing cDNAs encoding light and heavy chain sequences of H I11 were prepared. The orientation, DNA inserts and antibiotic selection criteria of these constructs are shown in Table 15 where CMV is cytomegalovirus; DHFR is dihydrofolate reductase; HC is heavy chain and LC is light chain.
Table vector DNA insert HC LC promoter orientation antibiotic amplif promoter selection ppNB 1 cDNA heavy and CMV* HC- clockwise neomycin DHFR light chains LC- anticlockwise pNB2 cDNA heavy and CMV HC clockwise zeocin DHFR light chains LC- clockwise pNB3 cDNA heavy and CMV HC clockwise zeocin DHFR light chains LC anticlockwise These expression vectors have separate insertional sites for the sequences encoding the light and heavy antibody chains. A high level of constitutive expression of both the heavy and light chains is directed by the cytomegalovirus immediate early (CMV) enhancer/promoter. A chimeric intron comprising the 5' donor site of the first intron of the 10 human P-globin and the 3' acceptor site from the intron of an immunoglobulin gene (heavy chain variable region) is located downstream from the promoter which has frequently been shown to enhance gene expression levels. Polyadenylation of mRNAs are provided by the poladenylation signal from the simian virus 40 The plasmids also contain the gene encoding dihydrofolate reductase (DHFR) and 15 can thus be grown in Chinese hamster ovary (CHO) DHFR deficient cells. Amplification using methotrexate, a folate analogue and potent DHFR inhibitor, results in amplification of the DHFR gene and its flanking sequences (namely, the light and heavy chains of the antibody in the construct). A stepwise increase in methotrexate (from about 0.01 nM to about 800 nM) concentration can produce very high levels of protein from the target 20 gene(s). The constructs also contain a gene which confers antibiotic resistance was a selectable marker, either neomycin or zeomycin is used. The vectors are shown in Figs. and 16.
81 Results of flow-cytometry analysis of recombinantly produced HI 1 IgGI are shown in Table 16 and illustrate that H11 IgGI which binds to an antigen on SK-BR-3 breast carcinoma cells can be produced in CHO cells.
Table 16 Cell Lines/I.D. Conc. of'IgGI in Mean Increase of MF samples (mg/L) Fluorescence (MF) above IgGI control PBS 156 Control IgGI 5 169 1129/pNB1 2 1722 1233 /pNB 2 184 9 KL-13 pNB2 3.3 192 14 KL-14 pNB2 3.3 186 Sb2 pNB3 4.0 224 33 S3sB3 pNB3 3.9 200 18 EXAMPLE 11 S H11 Binding to Cancer Cell Lines The binding affinities of HI1 IgM and HI l-scFv for various human cancer cell 10 lines were determined by labeling HI 1 antibodies with either radioactive iodine or radioactive indium. 25I-H 1 -scFv was prepared with specific activities of 7, 20 or 150 LCi/pg and 2I-H11 IgM with 0.6 pCi/pg were obtained. In addition, "In-H 11 -scFv having a specific activity.of 13 and 38 pCi/tg was prepared as described in Example 12.
The scFv 3B 1, which does not recognize the C-antigen, was used as a control to indicate 15 non-specific binding and was labeled with 150 pCi/lg.
12I-H11-scFv was purified using a P-2 minicolumn and analyzed by paper chromatography in 85% methanol as shown in Figure 17. 12SI-H 1 IgM was purified using a Sephadex G-50 mini-column (Pharmacia) and analyzed by paper chromotography in methanol as shown in Figure 18. A Sephadex G-50 column was used to purify Iln- H 1 1 scFv which was then analyzed by ITLC-SG in 0.1 M Citrate as shown in Figure 19.
Results of H 1 binding are shown in Figures 13 and 14. Figure 13 shows the specific binding of 12I-H1 l-scFv to LS 174T human colon cancer cells. Figure 14 shows the total binding of "'In-HI -scFv to A375 cells.
The results obtained indicate that HI 1 binds specifically to both LT174T and human melanoma cells. HI 1 also binds, but with lower affinity, to the breast cancer cell line.
EXAMPLE 12 Tumor Imaging with Indium-DTPA-H Il1-scFv H 11-scFv was conjugated with the bycyclic anhydride of diethylenetriaminepentaacetic acid (DTPA) at a molar ratio of 10:1 (DTPA:H 11-scFv) resulting in a substitution level of 2 moles of DTPA per mole of HI1-scFv. DTPA-H 11scFv was purified from excess DTPA on a Sephadex G-25 (Pharmacia) mini-column and reconcentrated to 10 mg/mL using a Centricon-30 microconcentrator (Amicon). The DTPA-H 11-scFv was radiolabeled to a specific activity of 25 mCi/mg with "'Indium acetate. Unincorporated "'In was removed using a Sephadex G-25 minicolumn. The Indium acetate was prepared from Indium chloride (Nordion) and I M acetate buffer at pH 6.0. The radiochemical purity of the final '"In-DTAP-HI 1-scFv was greater than 99% as measured by thin layer silica gel chromatography in 100 mM sodium citrate pH 20 5.0. Figure 19 shows the purification and TLC.
A female nude mouse with an existing subcutaneous A375 melanoma xenograft on the right lateral side and a subcutaneous HT-29 human colon cancer xenograft in the midabdominal region was injected intravenously in the tail vein with 100 pCi of 'In-DTAP- H11-scFv. The mouse was immediately placed under the gamma camera (Siemans 25 ZL3700) interfaced with a GE Star 4000i computer and a dynamic acquisition was obtained for 120 minutes, for a total of 480 frames of 15 seconds each. The frames were then combined into 12 x 10 minute images. The A375 tumor was visible on the right lateral side of the mouse as early as 30 minutes post-injection.
Region-of-interest analysis of the two tumors showed that the A375 tumor accumulated radioactivity throughout the 120 minute study, whereas the HT-29 tumor accumulated radioactivity for the first hour and then the radioactivity concentration remained relatively constant. Figure 20 shows 12 frames and the two arrows on the bottom right hand frame, taken at 120 minutes, show the accumulation of radioactivity in the two tumors. The narrow arrow points to the A375 tumor, and the broad arrow points to the I-IT-29 tumor. Normal tissues visible on the images include the heart, liver, kidneys and bladder. The heart is visible due to circulating amounts of radioactivity, and the kidneys and bladder are visible due to renal elimination of "'In-DTAP-HI l-scFv. The small amount of liver uptake may be due to blood flow to the liver or to partial binding of ''In-DTAP-H 1l-scFv to the liver.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity and understanding, it will be apparent to those skilled in the art that certain changes and modifications may be practiced. Therefore, the description and examples should not be construed as limiting the scope of the invention, which is delineated by the appended claims.
15 Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers.
The reference to any prior art in this specification is not, and should not be taken 20 as, an acknowledgement or any form of suggestion that that prior art forms part of the common general knowledge in Australia.
o*o 84 SEQUENCE LISTING GENERAL INFORMATION: APPLICANT: Dan, Michael D.
Maiti. Pradip K.
Kaplan, Howard A.
(ii) TITLE OF INVENTION: ANTIGEN BINDING FRAGMENTS H11, THAT SPECIFICALLY DETECT CANCER CELLS, NUCLEOTIDES ENCODING THE FRAGMENTS, AND USE THEREOF FOR THE PROPHYLAXIS AND DETECTION OF CANCERS (iii) NUMBER OF SEQUENCES: 18 (iv) CORRESPONDENCE ADDRESS: ADDRESSEE: Morrison Foerster STREET: 755 Page Mill Road CITY: Palo Alto STATE: CA COUNTRY: USA ZIP: 94304-1018 COMPUTER READABLE FORM: MEDIUM TYPE: Floppy disk COMPUTER: IBM PC compatible OPERATING SYSTEM: PC-DOS/MS-DOS SOFTWARE: PatentIn Release Version #1.30 (vi) CURRENT APPLICATION DATA: APPLICATION NUMBER: FILING DATE:
CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION: NAME: Lehnhardt, Susan K.
REGISTRATION NUMBER: 33,943 REFERENCE/DOCKET NUMBER: 31608-20001.20 (ix) TELECOMMUNICATION INFORMATION: TELEPHONE: (415) 813-5600 TELEFAX: (415) 494-0792 TELEX: 706141 INFORMATION FOR SEQ ID NO:1: SEQUENCE CHARACTERISTICS: LENGTH: 543 base pairs TYPE: nucleic acid STRANDEDNESS: double TOPOLOGY: linear (ix) FEATURE: NAME/KEY: CDS LOCATION: 1..543
I
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:i: CAAGCTATTT AGGTGACACT ATAGAATACT CAAGCTATGC ATCCAACGCG TTGGGAGCTC
TCCCATATGG
AGAGGACTCA
ATCCAGTGTC
CTGAGACTCT
CGCCAGGCTC
AAATACTACG
ACGGTGTATC
GAAAGACAGA
GGA
(2j I NFORMI
TCGACCTGCA
CCATGGAGTT
AGGTGCAGCT
CCTGTGCAGC
TAGGCAAGGG
CAGACTCCGT
TAAAAATGAA
GCCTGCTGGG
GGCGGCCGCA
TGGGCTGAGC
GGTGGAGTCT
CTCTGGATTC
GCTGGAGTGG
GAAGGGGCGA
CAGGCTGAGA
TGACTATGAC
CTAGTGATTT
TGGGTTTTCC
GGGGGAGGCG
CCC--TCAGAA
GTGGCAGTTA
TTCACCATCT
AC7GAGGACA
CACTACTACG
CAAGCTTCAT
TCGTTGCTCT
TGGTCCAGCC
GCTTTGCTAT
TATCATATGA
CCAGAGACAC
CGGCTGTCTT
GNTTGGACGC
CACTGAACAC
TTTAAGAGGT
TGGGAGGTCC
GCACTGGGTC
TGGAAGCACT
TTCCAAGAAC
TTACTTGTGC
TTGGGGAAAG
120 180 240 300 360 420 480 540 543 LTION FOR SEQ ID NO:2: SEQUENCE CHARACTERISTICS: LENGTH: 179 amino acids TYPE: amino acid STRANJDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2: C Gin Ala Ile Val Thr Leu Asn Thr Gin Ala Met His Pro Thr Gin Ala Arg Trp is Glu Leu Ser His Met Val Asp Leu Ala Ala Leu Ser Phe Ile Trp Vai Phe Thr Giu His Arg Gly 40 Leu Leu Thr Met Glu Phe Cys Val Ile Ser Gly Leu Ser Gin Val Gin Leu Val Ala so Leu Vai Leu 55 Gly Arg Gly Ile Gin Gly Giu Ser Gly Val Val Gin Leu Pro 75 Arg Arg Ser Leu Arg So Ser Cys Ala Ala Gly Phe Pro Phe 90 Gly Lys Gly Leu 105 Ser Phe Ala Met His Val Ile Trp Vai Arg Gin 100 Ala Leu Giu Trp Val Ala 110 Ser Tyr Asp Gly Ser Thr Lys Tyr Tyr Ala Asp Ser Val Lys Gly Arg 115 120 125 Phe Thr lie Ser Arg Asp Thr Ser Lys Asn Thr Val Tyr Leu Lys Met 130 135 140 Asn Arg Leu Arg Thr Glu Asp Thr Ala Val Phe Tyr Leu Cys Glu Arg 145 150 155 160 Gin Ser Leu Leu Gly Asp Tyr Asp His Tyr Tyr Gly Leu Asp Ala Trp 165 170 175 Gly Lys Gly INFORMATION FOR SEQ ID NO:3: SEQUENCE CHARACTERISTICS: LENGTH: 543 base pairs TYPE: nucleic acid STRANDEDNESS: double TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3: TCCCTTTCCC CAAGCGTCCA ANCCGTAGTA GTGGTCATAG
TCACCCAGCA
TTCGCACAG TAAAAGACAG CCGTGTCCTC AGTTCTCAGC
CTGTTCATTT
CGTGTTCTTG GAAGTGTCTC TGGAGATGGT GAATCGCCCC
TTCACGGAGT
TTTAGTGCTT CCATCATATG ATATAACTGC CACCCACTCC
AGCCCCTTGC
GCGGACCCAG TGCATAGCAA AGCTTCTGAj., GGGGAATCCA
GAGGCTGCAC
CAGGGACCTC CCAGGCTGGA CCACGCCTCC CCCAGACTCC
ACCAGCTGCA
GATACCTCTT AAAAGAGCAA CGAGGAAAAC CCAGCTCAGC
CCAAACTCCA
TCTGTGTTCA GTGATGAAGC TTGAAATCAC TAGTGCGGCC GCC TGCAGGT GGAGAGCTCC CAACGCGTTG GATGCATAGC TTGAGTATTC
TATAGTGTCA
TTG
INFORMATION FOR SEQ ID NO:4: SEQUENCE CHARACTERISTICS: LENGTH: 450 base pairs TYPE: nucleic acid STRANDEDNESS: double TOPOLOGY: linear
GGCTCTGTCT
TTAGATACAC
CTGCGTAGTA
CTAGAGCCTG
AGGAGAGTCT
CCTGACACTG
TGGTGAGTCC
CGACCATATG
CCTAAATAGC
220 180 240 300 360 420 480 540 (ix) FEATURE: NAME/KEY: CDS LOCATION: 1. .450 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4: CTCGAGATGG ACATGGAGTT CCAGGCGCAG CTTCTCTTCC TCCTGCTACT CTGGCTCCCA GATATCACCG GAGATATTGT GTTGACGCAG TCTCCAGGCA CCCTGTCTTT GTCTCCAGGG GAAAGAGCCA CCCTCTCCTG CAGGGCCAGT CAGAGTGTTA GTAGCAGCTA CTTAGCCTGG TACCAGCAGA AACCTGGCCA GGCTCCCAGG CTCCTCATCT ATGGTGCATC CACCAGGGCC ACTGGCATGC CAGACAGGTC CAGTGGCAGT GGGTCCGGGA CAGACTTCAC TCTCACCATC AGTAGACTGG AGCCTGAAGA T TTT GCAGTG TATTACTGTC AGCAGTATGG TAGCTCACCT CAGACACCTC AGATCACTTT CGGCGGAGGG ACCAAGGTGG AGATCAAACG AACTGTGGCT GCACCATCTG TCTTCATCTT CCCGCCATCT INFORMATION FOR SEQ ID (iJ) SEQUENCE CHARACTERISTICS: LENGTH: 150 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID Leu Glu Met Asp Met Glu Phe Gin Ala Gin Leu Leu Phe Leu Leu Leu 120 180 240 300 360 420 450 5= 5a 1 Leu Gly Trp Leu Pro Asp Ile Thr Thr Leu Ser Leu Ser Pro 35 Ala Ser 50 Pro Gly Thr Gly Gin Ser Val Ser Ser 5 Gln Ala Pro Arg Leu 70 Met Pro Asp Arg Phe 85 Thr Ile Ser Arg Leu Gly Asp Ile 25 Gly Glu Arg 40 Ser Tyr Leu Leu Ile Tyr Ser Gly Ser 90 Val Leu Thr Ala Thr Leu Ala Trp Tyr Gln Ser Ser Pro Cys Arg Gln Gin Lys Gly Ala Ser Thr Arg Gly Thr Leu Giu Pro Giu Asp Pro Gin Thr Pro 120 Ser Gly Thr Asp Phe Phe Ala Val Tyr Tyr 110 Gin Ile Thr Phe Gly 125 Cys Gln Gin Tyr Gly Ser Ser 115 88 Gly Gly Thr Lys Val Glu Ile Lys Arg Thr Val Ala Ala Pro Ser Val 130 135 140 Phe Ile Phe Pro Pro Ser 14515 INFORMATION FOR SEQ ID NO:6: SEQUENCE CHARACTERISTICS: LENGTH: 450 base pairs TYPE: nucleic acid STRANDEDNESS: double TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6: AGATGGCGGG AAGATGAAGA CAGAT GGTGC AGCCACAGTT CGTTGATCT CCACCTTGGT CCCTCCGCCG AAAGTGATCT GAGGTGTCTG AGGTGAGC-TA CCATACTGCT GACAGTAATA 120 CACTGCAAAA TCTTCAGGCT CCAGTCTACT GATGGTGAGA GTGAAGTCTG TCCCGGACCC 180 ACTGCCACTG AACCTGTCTG GCATGCCAGT GGCCCTGGTG GATGCACCAT AGATGAGGAG 240 CCTGGGAGCC TGGCCAGGI-I TCTGCTGGTA CCAGGCTAAG TAGCTGCTAC TAACACTCTG 300 ACTGGCCCTG CAGGAGAGGG TGGCTCTTTC CCCTGGAGAC AAAGACAGGG TGCCTGGAGA 360 :CTGCGTCAAC ACAATATCTC CGGTGATATC TGGGAGCCAG AGTAGCAGGA GGAAGAGAAG 420 *CTGCGCCTGG AACTCCATGT CCATCTCGAG 450 INFORMATION FOR SEQ ID NO:7: SEQUENCE CHARACTERISTICS: LENGTH: 34 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7: S.TATGAAGACA CCAGGCCGAT ATTGTGTTGA CGCA 34 INFORMATION FOR SEQ ID NO:S: SEQUENCE
CHARACTERISTICS:
LENGTH: 26 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:B: 89 TATCCGGATG CAGCCACAGT TCGTTT 26 INFORMATION FOR SEQ ID NO:9: SEQUENCE CHARACTERISTICS: LENGTH: 26 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: TATTCGGACA GGTGCAGCTG GTGGAG 26 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 27 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID TATGGATCCT GAGGAGACGG TGACCGT 27 INFORMATION FOR SEQ ID NO:11: SEQUENCE CHARACTERISTICS: LENGTH: 60 base pairs 0o TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:ll: TATATATCCG GAGGTGGTGG ATCAGGTGGA GGTGGCTCCC AGGTGCAGCT GGTGGAGTCT INFORMATION FOR SEQ ID NO:12: g SEQUENCE CHARACTERISTICS: LENGTH: 46 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12: ACCTCCGGAA CCGCCACCGC CAGAGACAGA TGGTGCAGCC ACATTC 46 !NFOJRMAT-ON FOR SEQ ID NO:i13: SEQUENCE
CHARACTERISTICS:
LENGTH: 918 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ix) FEATURE: NAME/KEY:
CDS
LOCATION: join(1. .906, 913. .918) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1]:
GAA
Giu 1 TTC ATG AAA AAA ACC GCT ATC GCG ATC GCA GTT GCA CTG Phe Met Lys Lys Thr Ala Ile Ala Ile Ala Val Ala Leu GCT GGT Ala Gly TTC GCT ACC Phe Ala Thr ACC CTG TCT Thr Leu Ser 35 GCG CAG GCC GAT Ala Gln Ala Asp ATT GTG TTG ACG Ile Vai Leu Thr AGA GCC ACC CTC Arg Ala Thr Leu CAG TCT CCA GGC Gin Ser Pro Gly TTG TCT CCA GGG Leu Ser Pro Gly TGC AGG GCC CYs Arg Ala AGT CAG Ser Gln s0 AGT GTT AGT AGC Ser Val Ser Ser
AGC
Ser 55 TAC TTA GCC TGG Tyr Leu Ala Trp
TAC
Tyr CAG CAG AAA CCT Gin Gin Lys Pro
GC
Gly 65 CAG GCT CCC AGG Gin Ala Pro Arg CTC ATC TAT GGT Leu Ile Tyr Gly TCC ACC AGG GCC Ser Thr Arg Ala
ACT
Thr 144 192 240 268 336 384 GOC ATO CCA GAC Gly Met Pro Asp
AGG
Arg 85 TTC AGT GGC AGT Phe Ser Gly Ser
GGG
Gly 90 TCC GGG ACA GAC Ser Gly Thr Asp TTC ACT Phe Thr CTC ACC ATC Leu Thr Ile
ACT
Ser 100 AGA CTG GAG CCT Arg Leu Giu Pro
GAA
Giu 105 GAT TTT GCA GTG Asp Phe Ala Val TAT TAC TGT Tyr Tyr Cys CAG CAG TAT GGT AGC TCA CCT CAG ACA CCT CAG ATC ACT TTC GGC GGA Gin Gin Tyr Giy Ser Ser Pro Gin Thr Pro Gin Ile Thr Phe Gly Gly 115 120 1)C GGG ACC Gly Thr 130 AAG GTG GAG ATC AAA CGA ACT GTG GCT GCA Lys Val Giu Ile Lys Arg Thr Val Ala Ala 135 140 CCA TCT GTC TCT Pro Ser Val Ser GGC GGT GGC OGT TCC GGA GGT GGT GGA TCA GGT GGA GOT GOC TCC CAG Gly G1ly Oly Gay Ser Gly Gay Gay Gly Ser'Gly Gly Gly Oly Ser Gin 14S 150 155 160 GTG CAG CTG GTG GAG TCT GGG GGA GGC GTG GTC CAG CCT GGG AGG TCC Val Gin Leu Val Oiu Ser Gly Oly Gly Vq- a O1 n.1 Inh rc C Iy A rg 165 170 175 480 528 CTO AGA CTC Leu Arg Leu ATO CAC TG Met His TrD 195
TCC
Ser 180 TOT GCA GCC TCT Cys Ala Ala Ser
GGA
Oly 185 TTC CCC TTC AGA Phe Pro Phe Arg AGC TTT GCT Ser Phe Ala 190 TOG OTO GCA Trp Val Ala 576 624 GTC CGC CAG OCT Val Arg Gin Ala
CTA
Leu 200 GGC AAG GGG CTO Gly Lys Gay Leu
GAG
Glu 205 O TT ATA Val lie 210 TCA TAT OAT OGA Ser Tyr Asp Gly
AOC
Ser 215 ACT AAA TAC TAC Thr Lys Tyr Tyr
OCA
Ala 220 GAC TCC OTO AAG Asp Ser Val Lys
GC
Oly 225 COA TTC ACC ATC Arg Phe Thr Ile
TCC
Ser 230 AGA GAC ACT TCC Arg Asp Thr Ser
AAG
Lys 235 AAC ACO OTO TAT Asn Thin Val Tyr
CTA
Leu 672 720 768 AAA ATO AAC AGC Lys Met Asn Ser
CTG
Leu 245 AGA ACT GAG GAC Aing Thin Giu Asp
ACG
Thn 250 OCT OTC TAT TAC Ala Val Tyr Tyr TOT OCO CYs Ala 255 AGA OAT CAG Arg Asp Gln OTC TOG GOC Val Trp Gly 275
AOC
Ser 260 CTG TTG GOT GAC Leu Leu Oly Asp
TAT
Tyr 265 GAC CAC TAC TAC Asp His Tyr Tyr GOT TTO GAC Gay Leu Asp 270 TCC GAA CAA Ser Giu Gin AAA 000 ACC ACG Lys Gly Thr Thr
GTC
Val 280 ACC OTC TCC TCA Thr Val Ser Ser
GGA
Gly 285 816 864 906 918 AAA CTO Lys Leu 290 ATC AOC GAA OAA Ile Ser Glu Glu
GAT
Asp 295 CTG AAC CAT CAC Leu Asn His His CAT CAC CAT His His His 300 TAGTGA AAO CTT Lys Leu INFORMATION FOR SEQ ID NO:14: SEQUENCE
CHARACTERISTICS:
LENGTH: 304 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14: Glu Phe Met Lys Lys Thr Ala Ile Al Phe Thr Ser Gly Gly Leu Gin Gly Gly 145 Vai Leu Met Val Gly.
225 Lys Arg Ali Lei Glr sc Gin Met Thr Gin Thr 130 Gly Gin Arg His Ile 210 Arg Met Asp Thr 1 Ser Ser Ala Pro Ile Tvr 115 Lys Gly Leu Leu Trp 195 Ser Phe Asn Gin Val Leu Va1 Pro Asp Ser 100 Gly Vai Gly Val Ser 180 Val Tyr Thr Ser i Ser 260 Ala Ser Sex Arg Arg Arg Ser Glu Ser Glu 165 Cys Arg Asp Ile Leu 245 eu Gin Pro Ser Leu 70 Phe Leu Ser Ile Gly 150 Ser Ala Gin Gly Ser 230 Arg Leu Al G13 Sel Leu Ser Glu Pro Lys 135 Gly Gly Ala Ala Ser 215 Arg rhr 3 1y a Asp t Glu 40 Tyr Ile Gly Pro Gln 120 Arg Gly Gly I Ser Leu 200 Thr I Asp Glu I Asp 'I 2 11 2! Arc Lei Tyr Ser Glu 105 Thr Thr Gly Gly Gly 185 31 y .ys rhr ~sp :yr !65 a Ile Val Ala Ala Gly Gly 90 Asp Pro Val Ser Val 170 Phe Lys Tyr Ser I Thr 1 250 Asp F Ala Leu Thr Trp Ala 75 Ser Phe Gln Ala Gly 155 al Pro 31y ['yr 4 ys ~35 la Iis Val Thr Leu Tvr Ser Gly Ala Ile Ala 140 Gly Gin Phe Leu Ala 220 Asn Val Tyr Al Glr Sex Gir Thr Thr Val Thr 125 Pro Gly Pro Arg Glu 205 Asp Thr Tyr Tyr a Leu x Ser Cys Gin Arg Asp Tyr 110 Phe Ser Gly Gly Ser 190 Trp Ser Val Tyr Gly I 270 Ala Pro Arg Lys Ala Phe Tyr Gly Val Se- Arg 175 Phe Jal lal yr -ys jeu Gly Gly Ala Pro Thr Thr Cys Gly Ser Gin 160 Ser Ala Ala Lys Leu 240 Ala Asp Val Trp Gly Lys Gly Thr Thr Val Thr Val Ser Se Gly Se Glu Gin 275 280 285 Lys Leu Ile Ser Glu Glu Asp Leu Asn His His His His His Lys Leu 290 295 300 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 918 base pairs TYPE: nucleic acid STR.ANDEDNESS: single TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID AAGC TTT CAC TAATGGTGAT GGTGATGGTT CAGAT CTT CT TCGCTGATCA GTT TTT GTTC GGATCCTGAG GAGACGGTGA GTCATAGTCA CCCAACAGGC TCTCAGGCTG TTCATTTTTA TCGGCCCTTC ACGGAGTCTG CCACTCCAGC CCCTTGCCTA GAATCCAGAG GCTGCACAGG AGACTCCACC AGCTGCACCT ACCGCCAGAG ACAGATGGTG *GAAAGTGATC TGAGGTGTCT ATCTTCAGGC TCCAGTCTAC GAACCTGTCT GGCATGCCAG CTGGCCAGGT TTCTGC TGGT *GCAGGAGAGG GTGCCTCTr CACAATATCG GCCTGCGCAA
CCGTGGTCCC
TCTGATCTCT
GATACACCGT
CGTAGTATTT
GAGCCTGGCG
AGAGTCTCAG
GGGAGCCACC
CAGCCACAGT
GAGGTGAGCT
TGATGGTGAG
TGGCCCTGGT
ACCAGGCTAA
CCCCTGGAGA
TTTGCCCCAG
CGCACAGTAA
GTTCTTGGAA
AGTGCTTCCA
GACCCAGTGC
GGACCTCCCA
TCCACCTGAT
TCGTTTGATC
ACCATACTGC
AGTGAAGTCT
GGATGCACCA
GTAGCTGCTA
CAAAGACAGG
ACGTCCAAAC
TAGACAGCCG
GTGTCTCTGG
TCATATGATA
ATAGCAAAGC
GGCTGGACCA
CCACCACCTC
TCCACCTTGG
TGACAGTAAT
GTCCCGGACC
TAGATGAGGA.
CTAACACTCT
GTGCCTGGAG
CGTAGTAGTG
TGTCCTCAGT
AGATGGTGAA
TAACTGCCAC
TTCTGAAGGG
CGCCTCCCCC
CGGAACCGCC
TCCC TCCGCC
ACACTGCAAA
CACTGCCACT
GCCTGGGAGC
GACTGGCCCT
ACTGCGTCAA
120 180 240 300 360 420 480 540 600 660 720 780 840 900 CGGTAGCGAA ACCAGCCAGT GCAACTGCGA TCGCGATAGC a.
GGTTTTTTTC ATGAATTC INFORMATION FOR SEQ ID NO:16: SEQUENCE CHARACTERISTICS: LENGTH: 867 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ix) FEATURE: NAME/KEY: CDS LOCATION: join(i. .855, 862. .867) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:16: GAA TTC ATC AAA AAA AC Glu Phe Met Lys Lys Thr GCT ATC CCG -7TC CCA CTT- Ala Ile Ala Ile Ala Val GCA CTG GCT GGT Ala Leu Ala Giy TTC GCT ACC Phe Ala Thr ACC CTG TCT Thr Leu Ser GTT GCG CAG GCC GAT ATT GTG Val Ala Gin Ala Asp Ile Val 25 TTG ACG CAG Leu Thr Gin TCT CCA GGC Ser Pro Gly TGC AGG GCC Cys Arg Ala 96 144 TT-G TCT CCA GGG Leu Ser Pro Gly
GAA
Glu AGA GCC ACC CTC Arg Ala Thr Leu AGT CAG Ser Gin AGT GTT AGT AGC Ser Vai Ser Ser
AGC
Ser TAC TTA GCC TGG Tyr Leu*Ala Trp CAG CAG AAA CCT Gin Gin Lys Pro
GGC
Gly CAG GCT CCC AGG Gin Ala Pro Arg
CTC
Leu 70 C"?rC ATC TAT GG-T Leu Ile Tyr Gly
GCA
Ala TCC ACC AGG GCC Ser Thr Arg Ala 240 288 GGC ATG CCA GAC Gly Met Pro Asp
AGG
Arg TTC AGT GGC AGT Phe Ser Gly Ser TCC GGG ACA GAC Ser Gly Thr Asp TTC ACT Phe Thr CTC ACC ATC Leu Thr Ile CAG CAG TAT Gin Gin Tyr 115
AGT
Ser 100 AGA CTG GAG CCT Arg Leu Giu Pro GAA GAT Glu Asp 105 TTT GCA GTG Phe Ala Val TAT TAC TGT Tyr Tyr Cys 110 TTC GGC GGA Phe Gly Gly 336 GGT AGC TCA CCT Gly Ser Ser Pro
CAG
Gin 120 ACA CCT CAG ATC Thr Pro Gin Ile
ACT
Thr 125 GGG ACC Gly Thr 130 AAG GTG GAG ATC Lys Val Giu Ile
AAA
Lys 135 CGA ACT GTG GCT Arg Thr Val Ala
GCA
Ala 140 TCC GGA CAG GTG Ser Gly Gin Val CTG GTG GAG TCT Leu Val Giu Ser
GG
Giy 150 GGA GGC GTG GTC Gly Gly Vai Val
CAG
Gin 155 CCT GGG AGG TCC Pro Gly Arg Ser AGA CTC TCC TGT Arg Leu Ser Cys
GCA
Ala 165 GCC TCT GGA TTC Ala Ser Gly Phe TTC AGA AGC TTT Phe Arg Ser Phe GCT ATG Ala Met 175 CAC TGG GTC His Trp Val
CGC
Arg 180 CAG GCT CTA GGC Gin Ala Leu Gly
AAG
Lys 185 GGG CTG GAG TGG Gly Leu Glu Trp GTG GCA GTT Val Ala Val 190 ATA TCA TAT GAT lie Ser Tvr Aso 195 GGA AGC ACT Gly Ser Thr
AAA
Lys 200 TAC TAC GCA GAC Tyr Tyr Ala Asp
TCC
Ser 205 GTG AAG GGC Val Lys Giy CGA TTC Arg Phe 210 ACC ATC TCC Thr Ile Ser AGA GAC ACT TCC AAG Arg Asp 215 Thr Ser Lys AAC ACG Asn Thr GTG TAT CTA AAA Val Tyr Leu Lys 672
ATG
Met 225 AAC AGC CTG AGA Asn Ser Leu Arg
ACT
Thr 230 GAG GAC ACG GCT Giu Asp Thr Ala
GTC
Val 235 TAT TAC TGT GCG Tyr Tyr Cys Ala
AGA
Arg 240 GAT CAG AGC CTG Asp Gin Ser Leu
TTG
Leu 245 GGT GAC TAT GAC Gly Asp Tyr Asp
CAC
His 250 TAC TAC GGT TTG Tyr Tyr Gly Leu GAC GTC Asp Val 255 TGG GGC AAA Trp Gly Lys
GGG
Glv 260 ACC ACO C-TC ACC Thr Thr Val Thr
GTC
Val1 265 TCC TCA GGA T-CC Ser Ser Gly Ser GAA CAA AAA Giu Gin Lys 270 TAGTGA AAG Lys CTG ATC AGC GAA GAA GAT CTG AAC Leu Ile Ser Glu Glu Asp Leu Asn 275 280 CAT CAC CAT CAC His His His His
CAT
His 285 864 867 S S
S.
S
*fr* S.
S
.5 *5
S
.5 INFORMATION FOR SEQ ID NO:i7: SEQUENCE CHARACTERISTICS: LENGTH: 287 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:i7: Giu Phe Met Lys Lys Thr Aia Ile Ala Ile 1 5 10 Phe Ala Thr Val Ala Gin Aia Asp Ile Val 20 Ala Vai Aia Leu Ala Gly Leu Thr Gin Ser Pro Gly Thr Leu Ser Leu Ser Pro Giy Giu Arg Ala Thr Leu Ser Cys Arg Ala 40 Ser Gin Ser Val Ser Ser Ser Tyr Leu Ala Trp Tyr Gin Gin Lys Pro 50 55 Gin A1la Pro Arg Leu Leu Ile T'yr Gly Ala Ser Thr Arg Aia Giy met Leu Thr Gin Gin Gly Thr 130 Pro Ile Tyr 115 Lys Asp Ser 1.00 Gly Vali Arg Arg Ser Giu Phe Leu Ser Ile Ser Giu Pro Lys Gi y Pro Gin 120 Arg Ser Giu 105 Thr Thr Giy 90 Asp Pro Vali Ser Phe G1. n Ala Gi y Ala.
Tie~ Thr Val 125 Asp Phe Tyr Tyr 110 Giy Gin Thr Cys Gly Val 135 140 Gin Leu Vai Giu Ser Giy 145 Arg His Ile Arg Met 225 Asp Trp Leu Leu Trp Ser Phe 210 Asn Gin Gly Ile Ser Val1 Tyr 195 Thr Ser Ser Lys Ser 275 Cys Arg 180 Asp Ile Leu Leu Gly 260 Giu Ala 165 Gin Gly Ser Arg Leu 245 Thr Giu 150 Aila Ala Ser Arg Thr 230 Giy Thr Asp *Gly Ser Leu Thr Asp 215 Giu Asp Vai Leu2 Gly Gly Gly Lys 200 Thr Asp T'yr rhr !Lsn ~80 Val1 Phe Lys 185 Tyr Ser Thr Asp Val 265 His Val1 Pro 170 Giv Tyr Lys Ala His 250 Ser Hi s Gin 155 Phe.
Leu Ala Asn Val1 235 Tyr Ser His Prc Arg Giu Asp Thr 220 Tyr Tyr Gly His Gly Ser Trp Ser 205 Val1 Tyr Gly Ser His 285 Arg Phe Val.
190 Val Tyr Cys Leu Glu 270 Lys Ser Ala Ala Lys Leu Ala Aso 255 Gin Leu Leu 160 Met Val Gly Lys Arg 240 Val Lys C. C
C.
SC
C.
C
C
C.
CC
CCCC
CCG C
C
S.C
CCC.
C. S C C 0* S S C
C.
INFORMATION FOR SEQ ID NO:1B: SEQUENCE CHARACTERISTICS: LENGTH: 867 base pairs TYPE: nucleic acid STRANDEDNESS: singie TOPOLOGY: linear SEQUENCE DESCRIPTION: SEQ ID NO:i8: AAGCTTTCAC TAATGGTGAT GGTGATGGTT CAGATCTTCT TCGCTGATCA GTTTTTGTTC GGATCCTGAG GAGACGGTGA CCGTGGTCCC TTTGCCCCAG ACGACCAAAC CGTAGTAGTG GTCATAGTCA CCCAACAGGC TCTGATCTCT CGCACAGTAA TAGACAGCCG TGTCCTCAGT 120 180 TCTCAGGCTG TTCATTTTTA GATACACCGT GTTCTTGGAA G7GTCTCTGG AGATGGTGAA TCGGCCCTTC ACGGAGTCTG CGTAGTATT AGTGCTlTCCA TCATATGATA TAACTGCCAC
CCACTCCAGC
GAATCCAGAG
CCCTCCGCCG
CACTGCAAAA
ACTGCCACTG
CCTGGG AG CC
ACTGGCCCTG
CTGCGTCAAC
CGCGATAGCG
CCCTTGCC-TA
GCTGCACAGG
AGCTGCACCT
AAAGTGATCT
TCTCAGGCT
AACCTGTCTG
TGGCCAGGTT
CAGGAGAGGG
ACAATATCGG
GTTTTTTTCA
GAGCCTGGCG
AGAGTCTCAG
GTCCGGATGC
GAGGTGTCTG
CCAGTCTACT
GCATGCCAGT
TCTGCTGGTA
TGGCTCTTTC
CCTGCGCAAC
TGA.ATTC
GACCCAGTGC
GGACCTCCCA
AG--CACAGTT
AGGTGAGCTA
GATGGTGAGA
GGSCCCTGGTG
CCAGGCTAAG
CCCTGGAGAC
GGTAGCGA;A
A-TAGCAAAGC
GGCTGGACCA
CGTTTGATCT
CCATACTGCT
GTGAAGTCTG
GATGCACCAT
TAGCTGCTAC
AAAGACAGGG
CCAGCCAGTG
TTCTGAAGGG
CGCCTCCCCC
CCACCTTGGT
GACAGTAATA
TCCCGGACCC
AGATGAGGAG
TAACACTCTG
TGCCTGGAGA
CAACTGCGAT
240 300 360 420 480 540 600 660 720 780 840 867 9*

Claims (144)

1. A composition comprising an antigen binding fragment which specifically recognizes C-antigen, wherein C-antigen is specifically recognized by an antibody or fragment thereof comprising the amino acid sequences of the H chain and L chain V regions of the scFv identified in SEQ ID NO:13, both said antigen binding fragment and said antibody or fragment thereof specifically recognizing, in competition with one another, an epitope on C-antigen.
2. A composition comprising an antigen binding fragment which specifically recognizes any one or more of at least glioma, melanoma, breast carcinoma, lung 10 carcinoma, ovarian carcinoma, lymphoma, gastric carcinoma, colon carcinoma or prostate carcinoma cells, but does not recognize normal, non-cancerous cells of at least brain, skin, •Too breast, lung, ovary, lymph node, large intestine and prostate tissues, and wherein said antigen binding fragment comprises amino acids which play a role in the specificity of an antibody or fragment thereof comprising the amino acid sequence of the H and L chain V regions of the scFv identified SEQ ID NO:13, said antigen binding fragment at least comprising an amino acid from one or more CDR regions of said antibody or fragment thereof, including at least one of the H and L chain CDR3s of said antibody or fragment thereof, such that said antigen binding fragment retains the specificity of said antibody or e fragment thereof. 0..
3. The composition according to claim 2, wherein said antigen binding fragment is a functionally equivalent fragment of said antibody or fragment thereof comprising a combination of one or more deletions, additions and/or substitutions relative to said antibody or fragment thereof.
4. The composition according to claim 1, 2 or 3, wherein said antigen binding fragment has an H chain CDR3 which comprises the amino acid sequence of the H chain CDR3 of said antibody or fragment thereof. The composition according to claim 1, 2 or 3, wherein said antigen binding fragment has a H chain CDR3 which consists of the amino acid sequence of the H chain CDR3 of said antibody or fragment thereof.
6. The composition according to claim 1, 2 or 3, wherein said antigen binding fragment has a H chain CDR3 which consists of the amino acid sequence of the H chain CDR3 of said antibody or fragment thereof with the exception of one or more, deletions, additions and/or substitutions, relative to said amino acid sequence.
7. The composition according to claim 4, wherein said antigen binding fragment comprises an L chain CDR3 comprising the amino acid sequence of the L chain CDR3 of said antibody or fragment thereof.
8. The composition according to claim 4, wherein said antigen binding fragment comprises an L chain CDR3 consisting of the amino acid sequence of the L chain 10 CDR3 of said antibody or fragment thereof
9.The composition according to claim 6, wherein said antigen binding fragment comprises an L chain CDR3 comprising the amino acid sequence of the L chain CDR3 of said antibody or fragment thereof. The composition according to claim 6, wherein said antigen binding fragment comprises an L chain CDR3 consisting of the amino acid sequence of the L chain CDR3 of said antibody or fragment thereof.
11. The composition according to claim 4, wherein said antigen binding fragment has an L chain CDR3 consisting of the amino acid sequence of the L chain CDR3 of said antibody or fragment thereof with the exception of one or more, deletions, additions and/or substitutions relative to said amino acid sequence.
12. The composition according to claim 6, wherein said antigen binding fragment comprises an L chain CDR3 comprising the amino acid sequence of the L chain CDR3 of said antibody or fragment thereof with the exception of one or more, deletions, additions and/or substitutions relative to said amino acid sequence.
13. The composition according to claim 6, wherein said antigen binding fragment comprises one or more conservative substitutions relative to said antibody or fragment thereof.
14. The composition according to claims 12, wherein said antigen binding fragment comprises one or more conservative substitutions relative to said antibody or fragment thereof. The composition according to claim 1, 2 or 3, wherein said antigen binding fragment specifically recognizes the tumors specifically recognized by said antibody or fragment thereof.
16. The composition according to claim 2 or 3, wherein said antigen binding fragment specifically recognizes substantially the same epitope recognized by said antibody or fragment thereof. 10 17. The composition according to claim 1, 2 or 3, wherein said antigen binding fragment specifically recognizes a heptapeptide displayed by peptide phage display, said heptapeptide selected from the group consisting of: Phe-His-Arg-Tyr-Ser-Leu-Pro; Phe-His-Arg-Tyr-Ser-Asp-Tyr; Phe-His-Arg-Tyr-Ser-Pro-Thr; o• Phe-His-Arg-Tyr-Thr-Pro-Gly; Phe-His-Arg-Tyr-Ser-Pro-Thr; and Met-His-Arg-Tyr-Thr-Pro-Leu.
18. The composition according to claim 1, 2, or 3, wherein the antigen-binding fragment specifically recognizes an N-terminus pentapeptide consensus sequence Phe-His- Arg-Tyr-Ser/Thr displayed as part of a heptapeptide by peptide phage display.
19. The composition according to claim 2, wherein the antigen-binding fragment comprises an H chain CDR3 comprising at least five consecutive amino acids of the H chain CDR3 of said antibody or fragment thereof. 101 The composition according to claim 2, wherein the antigen-binding fragment comprises a H chain CDR3 comprising at least six consecutive amino acid residues of the H chain CDR3 of said antibody or fragment thereof.
21. The composition according to claim 2, wherein the antigen-binding fragment comprises a H chain CDR3 comprising at least seven consecutive amino acid residues of the H chain CDR3 of said antibody or fragment thereof.
22. The composition according to claim 2, wherein the antigen-binding fragment comprises a H chain CDR3 comprising at least eight consecutive amino acid residues of the H chain CDR3 of said antibody or fragment thereof. 10 23. The composition according to claim 19, wherein the antigen-binding fragment comprises a L chain CDR3 comprising at least five consecutive amino acid residues of the L chain CDR3 of said antibody or fragment thereof.
24. The composition according to claim 24, wherein the antigen-binding fragment comprises a L chain CDR3 comprising at least six consecutive amino acid residues of the L chain CDR3 of said antibody or fragment thereof. "25. The composition according to claim 20, wherein the antigen-binding 0o0• fragment comprises a L chain CDR3 comprising at least seven consecutive amino acid o*o• residues of the L chain CDR3 of said antibody or fragment thereof.
26. The composition according to claim 1, 2 or 3, wherein the antigen-binding fragment comprises consecutive H chain V region amino acid residues corresponding identically to the H chain V region amino acids of said antibody -or fragment thereof.
27. The composition according to claim 26, wherein the antigen-binding fragment further comprises consecutive L chain V region amino acid residues corresponding identically to the L chain V region amino acids of said antibody or fragment thereof.
28. The composition according to any of claims 1, 2 or 3; wherein the antigen-- binding fragment comprises consecutive H chain V region amino acid residues which correspond identically to the H chain V region amino acids of said antibody or fragment 102 thereof, with the exception of one or more additions, deletions and/or substitutions relative thereto.
29. A composition comprising an antigen-binding fragment which is capable of inhibiting specific binding of an antibody to a cell surface antigen present on any one or more of at least glioma, melanoma, breast carcinoma, lung carcinoma, ovarian carcinoma, lymphoma, gastric carcinoma, colon carcinoma or prostate carcinoma tumor cells, said antibody comprising the H chain and L chain V regions of the scFv identified in SEQ. ID. NO:13; said antigen binding fragment capable of specifically binding to said. antigen to decrease binding of said antibody, and wherein said antigen binding fragment does not 10 specifically recognize normal non-cancerous cells of at least brain, skin, breast, lung, ovary, lymph node, large intestine and prostate tissues. A composition according to claim 29, wherein said antigen-binding fragment comprises an H chain CDR3 which consists of the amino acid sequence of the H S"chain CDR3 of the scFv identified in SEQ. ID. NO: 13.
31. A composition according to claim 29, wherein said antigen binding fragment comprises an H chain CDR3 which comprises the amino acid sequence .of the H chain CDR3 of the scFv identified in SEQ. ID. NO:13.
32. A composition according to claim 29, wherein said antigen binding o o fragment comprises an H chain CDR3 comprising the amino acid sequence of the H chain CDR3 of the scFv identified in SEQ. ID. NO:13 with the exception of one or more additions, deletions and/or substitutions relative to said sequence.
33. A composition according to claim 30, wherein said antigen binding fragment comprises an L chain CDR3 having the amino acid sequence of the L chain CDR3 of the scFv identified in SEQ. ID. NO:13.
34. A composition according to claim 32, wherein said antigen binding fragment comprises an L chain CDR3 having the amino acid sequence of L chain CDR3 of the scFv identified in SEQ. ID. NO:13 with the exception of one or more additions, deletions and/or substitutions relative to said sequence. 103 A composition according to claim 29, wherein said antigen binding fragment specifically recognizes the tumors specifically recognized by said antibody.
36. The composition according to claims 1, 2 or 29, wherein the antigen binding fragment is selected from the group consisting of whole native antibodies, bispecific antibodies, chimeric antibodies, Fab, F(ab')2, single chain V region fragments (scFv) and fusion polypeptides.
37. The composition according to claims 1, 2 or 29, wherein the antigen-- binding fragment is a scFv comprising amino acid sequences, which are substantially the same as the amino acid sequences of H and L chain V regions of the scFv identified in 10 SEQ IDNO:13.
38. The composition according to claims 1, 2 or 29, wherein the antigen-- binding fragment is a scFv comprising amino acid sequences which are substantially the same as the amino acid sequences of H and L chain V regions and the linker region of the scFv identified in SEQ ID NO:13.
39. The composition according to claims 1, 2 or 29, wherein the antigen-- binding fragment is a scFv comprising an amino acid sequence which is substantially the same as the amino acid sequence of the scFv identified in SEQ ID NO:13. The composition according to claim 36, wherein said antigen-binding fragment is fused to a chemically functional moiety, the moiety preferably selected from the group consisting of signal peptides, agents that enhance immunologic reactivity, agents that facilitate coupling to a solid support, carriers, bioresponse modifiers, toxins, detectable labels, paramagnetic labels, and drugs.
41. The composition according to claim 40, wherein the signal peptide is prokaryotic or eukaryotic.
42. The composition according to claim 40, wherein the signal peptide is prokaryotic.
43. The composition according to claim 40, wherein the agent that enhances immunologic reactivity is a bacterial superantigen. 104
44. The composition according to claim 40, wherein the agent that facilitates coupling to a solid support is selected from the group consisting of biotin and avidin. The composition according to claim 40, wherein the carrier is selected from the group consisting of large slowly metabolized macromolecules, polysaccharides, polymeric amino acids, amino acid copolymers, inactive virus particles or attenuated bacteria, serum albumins, keyhole limpet hernacyanin (KLH), Ig molecules, thyroglobulin, ovalbumin, and tetanus toxoid.
46. The composition according to claim 40, wherein the bioresponse modifier is a cytokine. S 10 47. The composition according to claim 46, wherein the cytokine is selected from the group consisting of tumor necrosis factor, interleukin-2, interleukin-4, granulocyte macrophage colony stimulating factor and interferons.
48. The composition according to claim 40, wherein the drug is an antineoplastic agent selected from the group consisting of radioisotopes, vinca alkaloids, S 15 adriamycin, bleomycin sulfate, Carboplatin, Cisplatin, cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Duanorubicin hydrochloride, Doxorubicin hydrochloride, Etoposide, fluorouracil, lomustine, Mechlororethamine hydrochloride, melphalan, mercaptopurine, methotrexate, mitomycin, mitotane, pentostatin, pipobrornan, procarbaze hydrochloride, streptozotocin, taxol, thioguanine and Uracil mustard.
49. The composition according to claim 48, wherein the vinca alkaloid is selected from the group consisting of vinblastine sulfate, vincristine sulfate and vindesine sulfate. The composition according to claim 40, wherein the toxin is selected from the group consisting of ricin, radionuclides, pokeweed antiviral protein, Pseudornonas exotoxin A, diphtheria toxin, ricin A chain, restrictocin and phospholipase enzymes.
51. The composition according to claim 40, wherein the detectable label is selected from the group consisting of radioisotopes, fluorescent compounds, colloidal metals, chemiluminescent compounds, bioluminescent compounds, enzymes, substrates, cofactors and inhibitors. 105
52. A composition according to any of claims 1, 2, 3, 19 or 29 to 35, wherein said antigen binding fragment further comprises a heterologous immunoglobulin C region.
53. The composition according to any of claims 1, 2, 3, 19 or 29 to 35, further comprising a pharmaceutically acceptable excipient.
54. A polymeric peptide comprising a plurality of the antigen-binding fragment identified in any of claims 1, 2, 3, 19 or 29 to An immunogenic composition comprising the antigen-binding fragment identified in any of claims 1, 2, 3, 19 or 29 to 3 5 and further comprising a pharmaceutically acceptable excipient and an amount of an adjuvant effective to enhance 10 an immune response. mc 56. A composition according to claim 37 or 38, wherein the scFv comprises a c- :myc peptide portion comprising amino acid residues 285-297 in SEQ ID NO: 13.
57. A derivative of the antigen-binding fragment identified in any of claims 1, S2, 3, 19 or 29 to •00*
58. A process for making the antigen binding fragment identified in any of claims 1, 2, 3, 19 or 29 to 35 by using an expression vector encoding the antigen binding fragment to express the antigen binding fragment in a cell.
59. A method of assessing an antigen binding fragment for its ability inhibit specific binding of an antibody comprising the H and L chain variable regions of the antibody fragment identified in SEQ.ID.NO: 13 to an antigen specifically recognized by the antibody, said method comprising the steps of: a) obtaining an antigen binding fragment having one or more deletions, additions, and/or substitutions relative to said antibody; and, b) determining whether said antigen binding fragment is capable of decreasing the binding of said antibody to said antigen A method of assessing an antigen-binding fragment for its ability to inhibit specific binding of an antibody comprising the H and L chain variable regions of the scFv 106 identified in SEQ. ID. NO:13 to a tumor cell surface antigen specifically recognized by the antibody, said method comprising the steps of: a) obtaining an antigen binding fragment which specifically recognizes any one or more of at least glioma, melanoma, breast carcinoma, lung carcinoma, ovarian carcinoma, lymphoma, gastric carcinoma, colon carcinoma or prostate carcinoma cells but does not recognize normal non-cancerous cells of at least brain, skin, breast, lung, ovary, lymph node, large intestine and prostate tissues; b) determining whether said antigen binding fragment is capable of decreasing the binding of said antibody to said cell surface antigen. 10 61. An antigen binding fragment capable of inhibiting specific binding of an antibody to a cell surface antigen of a tumor as determined by the method according to claim 59 or 60, said antigen binding fragment capable of specifically binding to said antigen to decrease binding of said antibody.
62. A composition comprising an antigen-binding fragment which specifically S 15 recognizes C-antigen, wherein C-antigen is specifically recognized by an antibody *ooo comprising the amino acid sequence of the H chain and L chain V regions of the scFv **identified in SEQ ID NO:13 or a fragment thereof having substantially the same specificity, and wherein both said antigen binding fragment and said antibody or fragment thereof specifically recognize the same tumor specific portion of C-antigen.
63. A composition according to claim 2, wherein said antigen-binding fragment is capable of inhibiting specific binding of said antibody or fragment thereof to the tumor cell surface antigen recognized by said antibody or fragment thereof.
64. A composition according to claim 2 or 29, wherein said antigen binding fragment does not specifically recognize any one of normal non-cancerous adrenal, bladder, cervix, esophagus, eye, heart, kidney liver, muscle, pancreas, parotid, pituitary, small intestine, spinal cord, spleen, thymus, thyroid, testis, cervix or uterus cells. A composition according to claim 6, wherein said antigen binding fragment specifically recognizes substantially the same epitope recognized by said antibody of fragment thereof. 107
66. A composition comprising an antigen binding fragment which specifically recognizes a heptapeptide selected from the group consisting of: Phe-His-Arg-Tyr-Ser-Leu-Pro; Phe-His-Arg-Tyr-Ser-Asp-Tyr; Phe-His-Arg-Tyr-Ser-Pro-Thr; Phe-His-Arg-Tyr-Thr-Pro-Gly; Phe-His-Arg-Tyr-Ser-Pro-Thr; and Met-His-Arg-Tyr-Thr-Pro-Leu. o.
67. A substantially isolated polynucleotide that encodes a polypeptide 10 comprising the H chain V region of an antigen binding fragment identified in any of claims 0 1 to 39.
68. A substantially isolated polynucleotide that encodes a polypeptide comprising the L chain V region of the antigen binding fragment identified in any of claims 1 to 39. 15 69. A substantially isolated polynucleotide sequence that encodes an antibody or fragment thereof comprising the H chain CDR3 of the antigen binding fragment identified in any of claims 1 to 39. A substantially isolated polynucleotide sequence that encodes an antibody or fragment thereof comprising the L chain CDR3 of an antigen binding fragment identified in any of claims 1 to 39.
71. A substantially isolated polynucleotide encoding an antibody or fragment thereof and comprising a region of at least 20 consecutive nucleotides that is capable of selectively forming a stable duplex with a polynucleotide encoding a CDR3 region of the antigen binding fragment identified in any of claims 1 to 39.
72. The polynucleotide according to any of claims 67 to 71, wherein the polynucleotide is a cloning vector. P:\Opcr\Vpa\VPA Prosecution\2363640 clean claims 2 108
73. The polynucleotide according to any of claims 67 to 71, wherein the polynucleotide is an expression vector.
74. A host cell comprising a polynucleotide according to claim 73. A pharmaceutical composition comprising a polynucleotide according to any of claims 67 to 71 and a pharmaceutically acceptable excipient, said polynucleotide encoding an antigen binding fragment having the specificity of an antibody or fragment thereof comprising the H and L chain V regions of the antibody fragment identified in SEQ. ID. NO: 13.
76. An immunogenic composition comprising the polynucleotide sequence according to any of claims 67 to 71 and a pharmaceutically acceptable excipient, said polynucleotide encoding an antigen binding fragment having the specificity of an antibody or fragment thereof comprising the H and L chain V regions of the scFv identified in SEQ. ID. NO:13. S. 77. A method of treating a patient with a neoplasia comprising administering to 15 the patient an effective amount of the antigen binding fragment as defined in any of claims 1 to 39.
78. The method according to claim 77, wherein the individual has a clinically detectable tumor.
79. The method according to claim 77, which is a method for palliating the neoplasia. The method according to claim 77, wherein a tumor that was previously detected in the individual has been treated and is clinically undetectable at the time of the administering of the antigen binding fragment.
81. The method according to claim 77, which is a method of reducing the risk of recurrence of a clinically detectable tumor.
82. The method according to claim 77, wherein administration of the antigen binding fragment is by parenteral administration selected from the group consisting of subcutaneous, intramuscular, intraperitoneal, intracavity, intrathecal, transdermal, or intravenous injection. 109
83. The method according to claim 77, wherein the administration is at a dosage of about 0.01 mg/kg/dose to about 2000 mg/kg/dose.
84. The method according to claim 77, wherein the antigen binding fragment is labeled with a therapeutic moiety.
85. The method according to claim 84, wherein the therapeutic moiety is selected from the group consisting of radioisotopes, antineoplastic agents, immunomodulators, biological response modifiers, lectins and toxins.
86. The use of an effective amount of an antigen binding fragment as defined in any of claims 1 to 39 in the manufacture of a medicament for administration to an S 10 individual for treating a neoplasia. 0**
87. A method for detecting C-antigen in a sample, comprising the steps of: a) contacting the sample with the antigen binding fragment defined in any of claims I to 39 under conditions that permit the formation of a stable antibody-antigen complex; and, 0 b) detecting any stable complex formed in step a) wherein C-antigen is the antigen specifically recognized by an antibody or fragment thereof comprising the H chain o and L chain V regions of the scFv identified in SEQ ID NO:13.
88. A process for making the antigen binding fragment identified in any of claims 1 to 39 by using an expression vector encoding the antigen binding fragment to express the antigen binding fragment in a cell.
89. A process of according to claim 88, wherein said cell is an E. coli bacterial cell and said expression vector includes a signal sequence immediately upstream of the coding sequence for said antigen binding fragment which targets the antigen binding fragment to the periplasmic space of said bacterial cell.
90. A polypeptide displayed on a phage by phage display, or a fragment thereof, having an epitope specifically recognized by an antigen binding fragment as claimed in any of claims 1 to 39. 110
91. A polypeptide according to claim 90 wherein said polypeptide is generated by a screening a peptide phage display library against said antigen binding fragment.
92. A polypeptide as claimed in claim 91, wherein said epitope comprises a heptapeptide selected from a group consisting of: Phe-His-Arg-Tyr-Ser-Leu-Pro; Phe-His-Arg-Tyr-Ser-Asp-Tyr; Phe-His-Arg-Tyr-Ser-Pro-Thr; Phe-His-Arg-Tyr-Thr-Pro-Gly; ;Phe-His-Arg-Tyr-Ser-Pro-Thr; and 10 Met-His-Arg-Tyr-Thr-Pro-Leu.
93. A polypeptide as claimed in claim 90, wherein said epitope comprises a pentapeptide consensus sequence Phe-His-Arg-Tyr-Ser/Thr.
94. A method of detecting an antigen binding fragment identified in any one of claims 1 to 39, by: 15 a) reacting said antigen binding fragment with an antibody or fragment thereof Swhich specifically recognizes said antigen binding fragment but does not recognize an antigen binding fragment lacking the specificity of said antigen binding fragment, under conditions suitable to form, in virtue of such specific recognition, a stable complex; and b) detecting the formation of said stable complex.
95. A method according to claim 94, wherein said antibody or fragment thereof is reacted in a biological sample with said antigen binding fragment as identified in any one of claims 1 to 39, and wherein said biological sample is depleted of antibodies or fragments thereof which specifically recognize the constant regions of an antigen binding fragment. 111
96. A method according to claim 94, wherein said antibody or fragment thereof specifically recognizes the portion of said antigen binding fragment to which is involved in idiotype anti-idiotype contact.
97. An antibody or fragment thereof which specifically recognizes an antigen binding fragment as identified in any one of claims 1 to 39 but does not recognize an antigen binding fragment lacking the specificity of said antigen binding fragment.
98. An antibody or fragment thereof according to claim 97, which is an anti-- idiotype of the antigen binding fragment identified in any one of claims 1 to 39.
99. The use of an immunogenic composition according to claim 55 to prepare a 10 biological sample comprising antibody or fragment thereof as claimed in claim 97.
100. A method of preparing a biological sample comprising an antibody or fragment thereof according to claim 97, said method comprising immunizing an individual ag with a composition according to claim 55 and obtaining a serum sample from said individual. 15 101. A polynucleotide encoding an antibody or fragment thereof as claimed in claim 97 or 98. *55* *•0
102. A method of preparing antibody or fragment thereof as claim 97 or 98 by using an expression vector comprising a polynucleotide as claimed in claim 101 to express said antibody or fragment thereof in a cell.
103. A composition as claimed in claim 2 or 3, wherein said antigen binding fragment competitively inhibits specific binding of said antibody to C-antigen.
104. A composition comprising an antigen-binding fragment which specifically recognizes C-antigen, wherein C-antigen is specifically recognized by an antibody comprising the amino acid sequence of the H chain and L chain V regions of the scFv identified in SEQ ID NO:13 or a fragment thereof having substantially the same specificity, both said antigen binding fragment and said antibody or fragment thereof specifically recognizing the same tumor specific portion of C-antigen. 112
105. A composition according to claim 104, wherein said antigen-binding fragment is capable of inhibiting specific binding of said antibody or fragment thereof to the tumor cell surface antigen recognized by said antibody or fragment thereof.
106. A composition according to claim 1 or 104, wherein said antigen binding fragment specifically recognizes any one or more of at least glioma, melanoma, breast carcinoma, lung carcinoma, ovarian carcinoma, lymphoma, gastric carcinoma, colon carcinoma or prostate carcinoma cells, but does not recognize normal, non-cancerous cells of at least brain, skin, breast, lung, ovary, lymph node, large intestine and prostate tissues.
107. A composition according to claim 106, wherein said antigen binding S 10 fragment specifically recognizes the tumors specifically recognized by said antibody or goeS fragment thereof.
108. A composition comprising an antigen binding fragment which specifically recognizes any one or more of at least glioma, melanoma, breast carcinoma, lung carcinoma, ovarian carcinoma, lymphoma, gastric carcinoma, colon carcinoma or prostate 15 carcinoma cells, but does not recognize normal, non-cancerous cells of at least brain, skin, 55.5 breast, lung, ovary, lymph node, large intestine and prostate tissues, and wherein said S antigen binding fragment comprises amino acid sequences which play a role in the S. S.oospecificity of an antibody or fragment thereof comprising the amino acid sequence of the H and L chain V regions of the scFv identified SEQ ID NO:13, said antigen binding fragment 20 at least comprising an amino acid sequence from one or more CDR regions of said antibody or fragment thereof, including at least the H chain CDR3 of said antibody or fragment thereof, such that said antigen binding fragment retains the specificity of said antibody or fragment thereof.
109. A composition comprising an antigen-binding fragment of an antibody which specifically recognizes an epitope on C-antigen recognized by an scFv comprising SEQ ID NO.13.
110. The composition according to claim I, wherein the antigen-binding fragment specifically recognizes at least one of glioma, melanoma, breast carcinoma, lung carcinoma, ovarian carcinoma, lymphoma, gastric carcinoma, colon carcinoma or prostate 113 carcinoma cells, but does not recognize normal, non-cancerous cells of at least one of brain, skin, breast, lung, ovary, lymph node, stomach, large intestine and prostate tissues.
111. The composition according to claim 1 10, wherein the antigen-binding fragment comprises a H chain CDR3 comprising at least six consecutive amino acid residues of SEQIDNO:13.
112. The composition according to claim 110, wherein the antigen-binding fragment comprises a H chain CDR3 comprising at least seven consecutive amino acid residues of SEQ ID NO: 13. S: 113. The composition according to claim 110, wherein the antigen-binding 10 fragment comprises a H chain CDR3 comprising at least eight consecutive amino acid residues of SEQ ID NO:13.
114. The composition according to claim 109, wherein the antigen-binding fragment comprises a L chain CDR3 comprising at least five consecutive amino acid residues of the L chain CDR3 of the scFv comprising SEQ ID NO: 13. ooooo 15 115. The composition according to claim 110, wherein the antigen-binding fragment comprises a L chain CDR3 comprising at least six consecutive amino acid residues of SEQ ID NO: 13. i 116. The composition according to claim 110, wherein the antigen-binding fragment comprises a L chain CDR3 comprising at least seven consecutive amino acid residues of SEQ ID NO: 13.
117. The composition according to any of claims 109 to 116, wherein the antigen-binding fragment is selected from the group consisting of whole antibodies, bispecific antibodies, chimeric antibodies, Fac, F(ab')2, single chain V region fragments (scFv) and fusion polypeptides.
118. The composition according to any of claims 109 to 116, wherein said antigen-binding fragment is fused to a chemically functional moiety.
119. The composition according to any of claims 109 to 116, wherein said antigen-binding fragment further comprises a heterologous immunoglobulin C region. 114
120. The composition according to any of claims 109 to 116 further comprising a pharmaceutically acceptable excipient.
121. The composition according to claim 120, wherein the amount of antigen- binding fragment corresponds to a dosage of about 0.01 mg/kg/dose to about 2000 mg/kg/dose.
122. A composition according to any of claims 109 to 116, wherein said antigen- binding fragment is an antigen-binding fragment of a human antibody.
123. A composition according to any of claims 109 to 116, wherein said antigen- binding fragment comprises human framework sequences. •9 10 124. The composition of any one of claims 1 to 56 and 62 to 66 or the derivative :i of claim 57, or the method of any one of claims 58 to 60, or the antigen binding fragment •of claim 61, or. the polynucleotide of any one of claims 67 to 73, or the host cell of claim 74, or the pharmaceutical composition of claim 75, or the immunogenic composition of claim 76, or the method of any one of claims 77 to 85, or the use of claim 86, or the method of claim 87, or the process of claim 88 or claim 89, or the polypeptide of any one of claim 90 to 93, or the method of any one of claims 94 to 96, or the antibody or antibody fragment of claim 97 or claim 98, or the use of claim 99, or the method of claim 100, or the polynucleotide of claim 101, or the method of claim 102, or the composition of any one of claims 103 to 123, substantially as herein before described with reference to the figures and/or examples.
125. A composition comprising an antigen binding polypeptide fragment which preferentially binds to a C-antigen epitope recognized by an scFv comprising amino acid sequences of the H chain or L chain regions of SEQ ID NO:14.
126. A composition comprising an antigen binding polypeptide fragment which specifically recognizes a C-antigen epitope recognized by an scFv comprising amino acid sequences of the H chain or L chain regions of SEQ ID NO:14.
127. The antigen binding polypeptide fragment according to claim 125 or 126 which specifically recognizes at least one of glioma, melanoma, breast carcinoma, lung carcinoma, ovarian carcinoma, lymphoma, colon carcinoma, gastric carcinoma or prostate 115 carcinoma cells, but does not recognize normal, non-cancerous cells of at least one of brain, skin, breast, lung, ovary, lymph node, large intestine, stomach and prostate tissues.
128. The antigen binding polypeptide fragment according to claim 125 or 126, wherein said antigen binding polypeptide fragment specifically recognizes a heptapeptide displayed by peptide phage display, said heptapeptide selected from the group consisting of: Phe-His-Arg-Tyr-Ser-Leu-Pro (SEQ ID Phe-His-Arg-Tyr-Ser-Asp-Tyr (SEQ ID NO:21); Phe-His-Arg-Tyr-Ser-Pro-Thr (SEQ ID NO:23); 10 Phe-His-Arg-Tyr-Thr-Pro-Gly (SEQ ID NO:24); and Met-His-Arg-Tyr-Thr-Pro-Leu (SEQ ID NO:28).
129. The composition according to claim 125, 126 or 127, wherein the antigen binding polypeptide fragment specifically recognizes a N-terminus pentapeptide consensus sequence Phe-His-Arg-Tyr-Ser/Thr displayed as part of a heptapeptide by peptide phage 15 display.
130. The antigen binding polypeptide fragment according to claim 125 or 126, comprising a CDR region of amino acid sequences of the H chain or L chain regions of SEQ ID NO:14.
131. The composition according to claim 130, wherein the antigen binding polypeptide fragment comprises at least five consecutive amino acid residues of the H chain CDR3 of the scFv of SEQ ID NO:14.
132. The composition according to claim 130, wherein the antigen binding polypeptide fragment comprises at least six consecutive amino acid residues of the H chain CDR3 of the scFv of SEQ ID NO:14.
133. The composition according to claim 130, wherein the antigen binding polypeptide fragment comprises at least seven consecutive amino acid residues of the H chain CDR3 of the scFv of SEQ ID NO:14. 116
134. The composition according to claim 130, wherein the antigen binding polypeptide fragment comprises at least eight consecutive amino acid residues of the H chain CDR3 of the scFv of SEQ ID NO:14.
135. The composition according to claim 130, wherein the antigen binding polypeptide fragment comprises at least five consecutive amino residues of the L chain CDR3 of the scFv of SEQ ID NO:14.
136. The composition according to claim 130, wherein the antigen binding polypeptide fragment comprises at least six consecutive amino acid residues of the L chain CDR3 of the scFv of SEQ ID NO:14.
137. The composition according to claim 130, wherein the antigen binding polypeptide fragment comprises at least seven consecutive amino acid residues of the L chain CDR3 of the scFv of SEQ ID NO:14.
138. The composition according to claim 130, wherein the antigen binding polypeptide fragment comprises at least eight consecutive amino acid residues of the L chain CDR3 of the scFv of SEQ ID NO:14.
139. The composition according to claim 125 or 126, wherein said antigen binding polypeptide fragment comprises one or more deletions, additions or substitutions relative to the amino acid sequences of one or both of the H chain and L chain V regions of the scFv construct identified in SEQ ID NO: 14.
140. A composition comprising an antigen binding polypeptide fragment which under suitable conditions inhibits specific binding of an antibody comprising amino acid sequences of the H chain or L chain regions of SEQ ID NO:14 to a cell surface antigen present on glioma, melanoma, breast carcinoma, lung carcinoma, ovarian carcinoma, lymphoma, colon carcinoma, gastric carcinoma or prostate carcinoma tumor cells, but not normal non-cancerous cells of at least one of brain, skin, breast, lung, ovary, lymph node, large intestine, stomach and prostate tissues.
141. The composition of claim 140, wherein said antigen binding polypeptide fragment does not bind to normal non-cancerous cells of brain, skin, breast, lung, ovary, lymph node, large intestine, stomach and prostate tissues. 117
142. The composition according to any one of claim 125 or 126 or 140, wherein the antigen binding polypeptide fragment is selected from the group consisting of whole antibodies, bispecific antibodies, chimeric antibodies, Fab, F(ab')2, H chain or L chain regions of single chain V region fragments (scFv) and fusion polypeptides thereof.
143. The composition according to claim 142, wherein said antigen binding polypeptide fragment is fused to a chemically functional moiety, which moiety does not does not function as an scFv linker molecule.
144. The antigen binding polypeptide fragment according to claim 143, wherein the moiety is selected from the group consisting of signal peptides, agents that enhance 10 immunologic reactivity, agents that facilitate coupling to a solid support, carriers, bioresponse modifiers, toxins, detectable labels, paramagnetic labels, and drugs.
145. The composition according to claim 143, wherein said antigen binding polypeptide fragment is conjugated with a bioactive component.
146. The composition according to claim 144, wherein the signal peptide is prokaryotic or eukaryotic.
147. The composition according to claim 145, wherein the signal peptide is prokaryotic.
148. The composition according to claim 144, wherein the agent that enhances immunologic reactivity is a bacterial superantigen.
149. The composition according to claim 144, wherein the agent that facilitates coupling to a solid support is selected from the group consisting of biotin and avidin.
150. The composition according to claim 144, wherein the carrier is selected from the group consisting of large slowly metabolized macromolecules, polysaccharides, polymeric amino acids, amino acid copolymers, inactive virus particles or attenuated bacteria, serum albumins, keyhole limpet hemacyanin (KLH), Ig molecules, thyroglobulin, ovalbumin, and tetanus toxoid.
151. The composition according to claim 144, wherein the bioresponse modifier is a cytokine. 118
152. The composition according to claim 151, wherein the cytokine is selected from the group consisting of tumor necrosis factor, interleukin-2, interleukin-4, granulocyte macrophage colony stimulating factor and interferons.
153. The composition according to claim 144, wherein the drug is an antineoplastic agent selected from the group consisting of radioisotopes, vinca alkaloids, adriamycin, bleomycin sulfate, Carboplatin, Cisplatin, cyclophosphamide, Cytarabine, Dacarbazine, Dactinomycin, Duanorubicin hydrochloride, Doxorubicin hydrochloride, Etoposide, fluorouracil, lomustine, Mechlororethamine hydrochloride, melphalan, mercaptopurine, methotrexate, mitomycin, mitotane, pentostatin, pipobroman procarbaze 10 hydrochloride, streptozotocin, taxol, thioguanine and Uracil mustard.
154. The composition according to claim 153, wherein the vinca alkaloid is selected from the group consisting of vinblastine sulfate, vincristine sulfate and vindesine sulfate.
155. The composition according to claim 144, wherein the toxin is selected from 15 the group consisting of ricin, radionuclides, pokeweed antiviral protein, Pseudomonas exotoxin A, diphtheria toxin, ricin A chain, restrictocin and phospholipase enzymes.
156. The composition according to claim 144, wherein the detectable label is selected from the group consisting of radioisotopes, fluorescent compounds, colloidal metals, chemiluminescent compounds, bioluminescent compounds, enzymes, substrates, cofactors and inhibitors.
157. A composition according to claim 125, 126, 127 or 140, wherein said antigen binding polypeptide fragment further comprises a heterologous immunoglobulin constant region.
158. The composition according to claim 125 or 126, 127 or 140 further comprising a pharmaceutically acceptable excipient.
159. The composition according to claim 158, wherein the antigen binding polypeptide fragment is present in a diagnostically effective amount. 119
160. A polymeric peptide comprising a plurality of the antigen binding polypeptide fragment according to claim 125 or 126, 127 or 140.
161. A method of inhibiting binding of a first antigen binding polypeptide fragment which preferentially binds to an epitope on a tumor cell recognized by an scFv comprising the heavy and light chain variable regions of SEQ ID NO:14 to an antigen which preferentially binds to the first antigen binding polypeptide fragment, said method comprising the steps of: a) obtaining a second antigen binding polypeptide fragment which preferentially binds to a tumor cell type recognized by the first antigen binding 10 polypeptide; and b) determining whether said second antigen binding polypeptide fragment decreases the binding of said first antigen binding polypeptide fragment to said epitope.
162. A method of inhibiting specific binding of a first antigen binding polypeptide fragment which specifically recognizes an epitope on a tumor cell recognized by an scFv comprising the heavy and light chain variable regions of SEQ ID NO:14 to an S' antigen specifically recognized by the first antigen binding polypeptide fragment, said method comprising the steps of: S. a) obtaining a second antigen binding polypeptide fragment which specifically binds to a tumor cell type recognized by the first antigen binding polypeptide; and b) determining whether said second antigen binding polypeptide fragment decreases the binding of said first antigen binding polypeptide fragment to said epitope.
163. An antigen binding polypeptide fragment of an antibody obtained by the method of claim 161 or 162.
164. The composition according to claim 125, 126, 140 or 163 wherein the antigen binding polypeptide fragment is an antigen binding polypeptide fragment of a human antibody.
165. The composition according to claim 125, 126, 140 or 163 wherein the antigen binding polypeptide fragment comprises human immunoglobulin sequences. 120
166. The composition according to claim 125, 126, 140 or 163 wherein the antigen binding polypeptide fragment is a humanized antigen binding polypeptide fragment.
167. A composition according to claim 125 or 126 wherein said antigen binding polypeptide fragment comprises at least one amino acid sequence within the VH region thereof which is encoded by a first DNA sequence that forms a stable duplex with a second DNA sequence encoding the VH region of said scFv.
168. A composition according to claim 125 or 126, wherein said antigen binding polypeptide fragment comprises at least one amino acid sequence within the VL region 10 thereof which is which is encoded by a first DNA sequence that forms a stable duplex •:with a second DNA sequence encoding the VL region of said scFv. :169. A composition according to claim 167 or 168, wherein said antigen binding fragment comprises at one amino acid which plays a role in the specificity of said scFv.
170. A composition according to claim 167 or 168, wherein said at least one amino acid sequence is a VH or VL CDR sequence which comprises 5 or more amino acids of the corresponding CDR sequence of the VH or VL region of the scFv.
171. A composition according to claim 170, wherein said CDR sequence is a CDR3 sequence.
172. A composition according to any of claims 167 to 171, wherein said antigen binding polypeptide fragment comprises one or more additions, deletions and/or substitutions relative to said at least one amino acid sequence.
173. The composition according to any one of claims 1-53, or the polymeric peptide according to claim 54, or the composition according to claim 55 or claim 56, or the derivative according to claim 57, or the process according to claim 58, or the method according to claim 59 or claim 60, or the antigen-binding fragment according to claim 61, or the composition according to any one of claims 62-66, or the polynucleotide according to any one of claims 67-73, or the host cell according to claim 74, or the composition according to claim 75 or claim 76, or the method according to any one of claims 77-85, or the use according to claim 86, or the method according to claim 87, or the process 121 according to claim 88 or claim 89, or the polypeptide according to any one of claims 93, or the method according to any one of claims 94-96, or the antibody or fragment according to claim 97 or claim 98, or the use according to claim 99 or the method according to claim 100, or the polynucleotide according to claim 101, or the method according to claim i02, or the composition according to any one of claims 103-126, or th antigen-binding polypeptide fragment according to claim 127 or claim 128, or the composition according to claim 129, or the antigen-binding polypeptide fragment according to claim 130, or the composition according to any one of claims 131-143, or the antigen-binding polypeptide fragment according to claim 144, or the composition according to any one of claims 145-159, or the polymeric peptide according to claim 160, or the method according to claim 161 or claim 162, or the antigen-binding polypeptide fragment according to claim 163, or the composition according to any one of claims 164- 172, substantially as herein before described with reference to the figures and/or examples. o DATED this 10th day of August 2001 Viventia Biotech Inc. DAVIES COLLISON CAVE Patent Attorneys for the applicant a *OO *O I
AU72432/00A 1996-05-22 2000-12-20 Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers Ceased AU775448B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU72432/00A AU775448B2 (en) 1996-05-22 2000-12-20 Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/657449 1996-05-22
AU33696/97A AU725238B2 (en) 1996-05-22 1997-05-22 Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers
AU72432/00A AU775448B2 (en) 1996-05-22 2000-12-20 Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU33696/97A Division AU725238B2 (en) 1996-05-22 1997-05-22 Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers

Publications (2)

Publication Number Publication Date
AU7243200A AU7243200A (en) 2001-03-22
AU775448B2 true AU775448B2 (en) 2004-07-29

Family

ID=32739128

Family Applications (1)

Application Number Title Priority Date Filing Date
AU72432/00A Ceased AU775448B2 (en) 1996-05-22 2000-12-20 Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers

Country Status (1)

Country Link
AU (1) AU775448B2 (en)

Also Published As

Publication number Publication date
AU7243200A (en) 2001-03-22

Similar Documents

Publication Publication Date Title
AU725238B2 (en) Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers
JP2000511421A5 (en)
JP3683270B2 (en) Monoclonal antibodies that recognize receptor Tie and uses thereof
EP0733072B1 (en) Antibody against carcinoembryonic antigen (cea)
CA2020247C (en) Antibodies reactive with human carcinomas
KR101143035B1 (en) Monoclonal antibody hPAM4
US7183384B2 (en) Monoclonal antibody 7H11 reactive with human cancer
JP3492373B2 (en) Monoclonal antibody
CA2295375A1 (en) Antigen binding fragments, designated 4b5, that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers
US20020122807A1 (en) Antigen binding fragments, designated 4B5, that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers
US20070025911A1 (en) Cancer specific monoclonal antibodies
AU775448B2 (en) Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers
CA2255540C (en) Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers
US7115722B1 (en) Antigen binding fragments that specifically detect cancer cells, nucleotides encoding the fragments, and use thereof for the prophylaxis and detection of cancers
NZ505305A (en) Antigen binding fragments that inhibit an antibody comprising amino acid sequences of H and L chain V regions of a scFv from binding to the surface of a cancer cell
WO2001040292A1 (en) Antigen-binding fragments specific for tumor associated antigens
FI111267B (en) Antibody against a Tie-receptor tyrosine kinase - used to diagnose and treat neoplastic diseases and for radiological imaging
Sikora Monoclonal antibodies and drug targeting in cancer
Thorpe University of London Royal Postgraduate Medical School Symposium on Advances in the Applications of Monoclonal Antibodies in Clinical
PL168700B1 (en) Method of obtaining a novel chimeric monoclonal antibody of human and mouse's origin

Legal Events

Date Code Title Description
TC Change of applicant's name (sec. 104)

Owner name: VIVENTIA BIOTECH INC.

Free format text: FORMER NAME: NOVOPHARM BIOTECH INC.