METHOD OF INDIRECT IMMUNIZATION OF HUMAN OVARIAN CANCER PATIENTS THROUGH SELECTION OF XENOGENEIC IMMUNOGLOBULIN FC PORTIONS

Abstract
The present document describes a method for therapeutic indirect immunization of an optimally debulked human ovarian cancer patient having a CA125 antigen level in the blood above normal levels, comprising administering to said human patient a monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region; an immune complex formed between CA125 antigen and said monoclonal antibody having at least a xenogeneic Fc region; or a combination thereof.
Description
BACKGROUND
(a) Field

The subject matter disclosed generally relates to methods for therapeutic immunization in patients in need thereof. More specifically, the subject matter disclosed relates to methods for therapeutic indirect immunization of optimally debulked human ovarian cancer patients having a CA125 antigen level in the blood above normal levels by administering a monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region, or an immune complex formed between CA125 antigen and said monoclonal antibody, or a combination thereof.


(b) Related Prior Art

Ovarian cancer is the most common cause of gynecologic cancer deaths in the United States. Cytotoxic therapy produces high initial response rates; however, recent intergroup study involving more than 4000 patients was unable to improve progression-free (PFS) or overall survival in any of the 4 experimental combinations that added a third drug with documented single agent activity to the standard front-line treatment [Bookman M. GOG0182-ICON5: J Clin Oncol. 2006; 24(18S):256s; Braly et al. (J Immunother 2009; 32:54-65]).


It is well established that aberrant expression of membrane mucin MUC16 (also known as CA125) is associated with tumor progression and metastasis of cancers such as ovarian and pancreatic cancer. The role of MUC16 in tumor progression and metastasis occurs through interaction with oncogenic modulators. For instance, it is understood that aberrant expression of MUC16 in ovarian cancer cells facilitates peritoneal metastasis through interactions with mesothelin (tumor differentiation factor) and through immunosuppressive functions by blocking natural killer cell-mediated cytotoxicity, while overexpression of MUC16 increases breast cancer cell proliferation via stimulation of Janus kinase 2 (JAK2). It is also understood that MUC16 is upregulated in pancreatic cancers, and expression is increased in liver metastases—although expression of MUC16 was not detected in pancreatic intraepithelial neoplasia (PanIN) nor in normal pancreas, suggesting that expression of MUC16 may occur later in disease progression.


A major focus of monoclonal antibody engineering in industry is to “de-immunize” the monoclonal antibody. Multiple generations of technologies to humanize monoclonal antibodies have been developed as proprietary technologies and in fact regulatory authorities require immunogenicity studies to ascertain that the monoclonal antibody is immune silent. Therefore, the common application of monoclonal antibodies in medicine is as large molecule drugs, which are administered with pharmacologic intent. This has been reviewed by Nicodemus in Immunotherapy 2015, 7: 923-33, and Nicodemus and Schultes in Expert Opinion Biological Therapy 2004, 4: 1265-1284, for example. Pharmacologic monoclonal antibodies target cell specific receptors and can either induce antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) mechanisms of cytotoxicity or can function as inhibitors or agonists of the receptors and thus modify the biology mediated by the specific receptor. Any of these applications would be interfered with by immune responses that are generated against the monoclonal antibody, and thus the dogma for optimal monoclonal antibody design to generate immune silent monoclonal antibody.


Also, in the field of oncology, therapeutic monoclonal antibodies are dosed according to traditional pharmacologic models and the dose administered is pushed to the limits of toxicity under the assumption that biological activity is proportional to dose. Such high doses then create safety issues associated with anti-monoclonal immune responses and toxicities, such as immune complex deposition of the administered antibody, seen most commonly with prototypic pharmacologically dosed murine monoclonal antibodies such as the original anti-CD-3 antibody OKT3 (Smith, S. L. (1996). Official publication of the North American Transplant Coordinators Organization (NATCO). 6 (3): 109-119). These safety issues severely limit the clinical application of such products.


An alternative approach for the use of therapeutic monoclonal antibodies is as indirect immunizers, taking advantage of the bell-shaped curve of immune dose response in which antigen or antibody excess can dampen response. Described herein is then a novel method for use of monoclonal antibodies having xenogeneic regions, particularly xenogeneic Fc regions permitting unique antigen antibody processing profiles. The current disclosure relates specifically to the counter intuitive requirements of xenogeneic Fc constant region properties to achieve optimal therapeutic benefit using the method of indirect immunization for therapeutic intent. The inventors have discovered that xenogeneic Fc regions (for example murine IgG1 Fc) have unique properties that especially favor indirect immunization pathways, in that the immunogenicity of xenogeneic Fc regions is leveraged to improve the immunization or therapeutic potential of monoclonal antibodies.


SUMMARY

According to an embodiment, there is provided a method for therapeutic indirect immunization of an optimally debulked human ovarian cancer patient having a CA125 antigen level in the blood above normal levels, comprising administering to the human patient

    • a monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region;
    • an immune complex formed between CA125 antigen and the monoclonal antibody having at least a xenogeneic Fc region; or
    • a combination thereof.


The CA125 antigen level in the blood above normal levels may be more than 35 units/mL of CA125 antigen in the blood. The CA125 antigen level in the blood above normal levels may be at least 50 units/mL of CA125 antigen in the blood.


The xenogeneic Fc region may be from an IgA, IgD, IgG or IgM isotype.


The xenogeneic Fc region may be from an IgG isotype.


The IgG isotype may be an IgG1 isotype.


The xenogeneic Fc region may be a murine Fc region, a rat Fc region, a rabbit Fc region, a goat Fc region, a hamster Fc region.


The xenogeneic Fc region may be a murine Fc region.


The xenogeneic Fc region may be a murine Fc region from an IgG isotype.


The IgG isotype may be an IgG1 isotype.


The antibody specific to CA125 may be mAb-B43.13 (oregovomab).


The administering may be one, two, three, or a maximum of four administrations.


The method may further comprise administration of an immune adjuvant.


The immune adjuvant may be a chemotherapeutic agent, an immunostimulatory compound, an immune homeostatic checkpoint inhibitor, or a combination thereof.


The chemotherapeutic agent may be a platinum-based chemotherapy, taxol, doxorubicin, topotecan, a poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor, or combinations thereof.


The platinum-based chemotherapy comprises cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin, satraplatin, and combinations thereof.


The immunostimulatory compound may be a TLR3 agonist, a TLR4 agonist, or combinations thereof.


The TLR3 agonist may be polyiC, polyICLC (Hiltonol®).


The chemotherapeutic agent may be a combination of carboplatin and taxol.


The therapeutic monoclonal antibody specific for a tumor associated antigen may be mAb-B43.13 (oregovomab), and the chemotherapeutic agent may be a combination of carboplatin and taxol.


The immune homeostatic checkpoint inhibitor may be an anti-PDL-1 antibody, an anti-CTLA-4 antibody, and anti-PD-1 antibody, or combinations thereof.


The anti-PDL-1 antibody may be selected from the group consisting of B7-H1 antibody, BMS-936559 antibody, MPDL3280A (atezolizumab) antibody, MEDI-4736 antibody, MSB0010718C antibody or combinations thereof.


The anti-CTLA-4 antibody may be selected from the group consisting of ipilimumab or tremelimumab or combinations thereof.


The anti-PD-1 antibody may be selected from the group consisting of Nivolumab antibody, pembrolizumab antibody, pidilizumab antibody or combinations thereof, and AMP-224.


The poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor may be selected from the group consisting of olaparib, niraparib, rucaparib, talazoparib, veliparib, CEP 9722, E7016, and BGB-290, or combinations thereof.


The dose of the monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region may be from about 0.1 to about 10 mg.


The method may be for the treatment of ovarian cancer.


The therapeutic indirect immunization may be inducing a mannose receptor dependent immune response in the human patient.


According to another embodiment, there is provided a vaccine composition for use in indirect immunization of an optimally debulked human ovarian cancer patient having a CA125 antigen level in the blood above normal levels, comprising

    • a monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region;
    • an immune complex formed between CA125 antigen and the monoclonal antibody having at least a xenogeneic Fc region;
    • or a combination thereof.


The CA125 antigen level in the blood above normal levels may be more than 35 units/mL of CA125 antigen in the blood. The CA125 antigen level in the blood above normal levels may be at least 50 units/mL of CA125 antigen in the blood.


The xenogeneic Fc region may be from an IgA, IgD, IgG or IgM isotype.


The xenogeneic Fc region may be from an IgG isotype.


The IgG isotype may be an IgG1 isotype.


The xenogeneic Fc region may be a murine Fc region, a rat Fc region, a rabbit Fc region, a goat Fc region, a hamster Fc region.


The xenogeneic Fc region may be a murine Fc region.


The xenogeneic Fc region may be a murine Fc region from an IgG isotype.


The IgG isotype may be an IgG1 isotype.


The antibody specific to CA125 may be mAb-B43.13 (oregovomab).


The vaccine composition may be for administration one, two, three, or four administrations.


The vaccine composition may be administered five, six, or more administrations.


The vaccine composition may further comprise an immune adjuvant.


The immune adjuvant may be a chemotherapeutic agent, an immunostimulatory compound, an immune homeostatic checkpoint inhibitor, or a combination thereof.


The chemotherapeutic agent may be a platinum-based chemotherapy, taxol, doxorubicin, topotecan, a poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor, or combinations thereof.


The platinum-based chemotherapy comprises cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin, satraplatin, and combinations thereof.


The immunostimulatory compound may be a TLR3 agonist, a TLR4 agonist, or combinations thereof.


The TLR3 agonist may be polyiC, polyICLC (Hiltonol®).


The chemotherapeutic agent may be a combination of carboplatin and taxol.


The therapeutic monoclonal antibody specific for a tumor associated antigen may be mAb-B43.13 (oregovomab), and the chemotherapeutic agent may be a combination of carboplatin and taxol.


The immune homeostatic checkpoint inhibitor may be an anti-PDL-1 antibody, an anti-CTLA-4 antibody, and anti-PD-1 antibody, or combinations thereof.


The anti-PDL-1 antibody may be selected from the group consisting of B7-H1 antibody, BMS-936559 antibody, MPDL3280A (atezolizumab) antibody, MEDI-4736 (Durvalumab) antibody, MSB0010718C (Avelumab) antibody or combinations thereof.


The anti-CTLA-4 antibody may be selected from the group consisting of ipilimumab or tremelimumab or combinations thereof.


The anti-PD-1 antibody may be selected from the group consisting of Nivolumab antibody, pembrolizumab antibody, pidilizumab antibody or combinations thereof, and AMP-224.


The poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor may be selected from the group consisting of olaparib, niraparib, rucaparib, talazoparib, veliparib, CEP 9722, E7016, and BGB-290, or combinations thereof.


The dose of the monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region may be from about 0.1 to about 10 mg.


The vaccine composition may be for the treatment of ovarian cancer.


The indirect immunization may be inducing a mannose receptor dependent immune response in the human patient.


According to another embodiment, there is provided a monoclonal antibody specific to CA125 antigen, having at least a xenogeneic Fc region, for use in therapeutic indirect immunization of an optimally debulked human ovarian cancer patient having a CA125 antigen level in the blood above normal levels.


The CA125 antigen level in the blood above normal levels may be more than 35 units/mL of CA125 antigen in the blood. The CA125 antigen level in the blood above normal levels may be at least 50 units/mL of CA125 antigen in the blood.


The xenogeneic Fc region may be from an IgA, IgD, IgG or IgM isotype.


The xenogeneic Fc region may be from an IgG isotype.


The IgG isotype may be an IgG1 isotype.


The xenogeneic Fc region may be a murine Fc region, a rat Fc region, a rabbit Fc region, a goat Fc region, a hamster Fc region.


The xenogeneic Fc region may be a murine Fc region.


The xenogeneic Fc region may be a murine Fc region from an IgG isotype.


The IgG isotype may be an IgG1 isotype.


The monoclonal antibody specific to CA125 may be mAb-B43.13 (oregovomab).


The monoclonal antibody may be for administration one, two, three, or four administrations.


The monoclonal antibody may be for administration five, six or more administrations.


The use may further comprise an immune adjuvant.


The immune adjuvant may be a chemotherapeutic agent, an immunostimulatory compound, an immune homeostatic checkpoint inhibitor, or a combination thereof.


The chemotherapeutic agent may be a platinum-based chemotherapy, taxol, doxorubicin, topotecan, a poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor, or combinations thereof.


The platinum-based chemotherapy comprises cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin, satraplatin, and combinations thereof.


The immunostimulatory compound may be a TLR3 agonist, a TLR4 agonist, or combinations thereof.


The TLR3 agonist may be polyiC, polyICLC (Hiltonol®).


The chemotherapeutic agent may be a combination of carboplatin and taxol.


The therapeutic monoclonal antibody specific for a tumor associated antigen may be mAb-B43.13 (oregovomab), and the chemotherapeutic agent may be a combination of carboplatin and taxol.


The immune homeostatic checkpoint inhibitor may be an anti-PDL-1 antibody, an anti-CTLA-4 antibody, and anti-PD-1 antibody, or combinations thereof.


The anti-PDL-1 antibody may be selected from the group consisting of B7-H1 antibody, BMS-936559 antibody, MPDL3280A (atezolizumab) antibody, MEDI-4736 antibody, MSB0010718C antibody or combinations thereof.


The anti-CTLA-4 antibody may be selected from the group consisting of ipilimumab or tremelimumab or combinations thereof.


The anti-PD-1 antibody may be selected from the group consisting of Nivolumab antibody, pembrolizumab antibody, pidilizumab antibody or combinations thereof, and AMP-224.


The poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor may be selected from the group consisting of olaparib, niraparib, rucaparib, talazoparib, veliparib, CEP 9722, E7016, and BGB-290, or combinations thereof.


The dose of the monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region may be from about 0.1 to about 10 mg.


The monoclonal antibody may be for the treatment of ovarian cancer.


The indirect immunization may be inducing a mannose receptor dependent immune response in the human patient.


According to another embodiment, there is provided a use of a monoclonal antibody specific to CA125 antigen, having at least a xenogeneic Fc region, for therapeutic indirect immunization of an optimally debulked human ovarian cancer patient having a CA125 antigen level in the blood above normal levels.


The CA125 antigen level in the blood above normal levels may be more than 35 units/mL of CA125 antigen in the blood. The CA125 antigen level in the blood above normal levels may be at least 50 units/mL of CA125 antigen in the blood.


The xenogeneic Fc region may be from an IgA, IgD, IgG or IgM isotype.


The xenogeneic Fc region may be from an IgG isotype.


The IgG isotype may be an IgG1 isotype.


The xenogeneic Fc region may be a murine Fc region, a rat Fc region, a rabbit Fc region, a goat Fc region, a hamster Fc region.


The xenogeneic Fc region may be a murine Fc region.


The xenogeneic Fc region may be a murine Fc region from an IgG isotype.


The IgG isotype may be an IgG1 isotype.


The antibody specific to CA125 may be mAb-B43.13 (oregovomab).


The use may be for administration of one, two, three, or four administrations.


The use may be for administration of five, six, or more administrations.


The use may further comprise an immune adjuvant.


The immune adjuvant may be a chemotherapeutic agent, an immunostimulatory compound, an immune homeostatic checkpoint inhibitor, or a combination thereof.


The chemotherapeutic agent may be a platinum-based chemotherapy, taxol, doxorubicin, topotecan, a poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor, or combinations thereof.


The platinum-based chemotherapy comprises cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin, satraplatin, and combinations thereof.


The immunostimulatory compound may be a TLR3 agonist, a TLR4 agonist, or combinations thereof.


The TLR3 agonist may be polyiC, polyICLC (Hiltonol®).


The chemotherapeutic agent may be a combination of carboplatin and taxol.


The therapeutic monoclonal antibody specific for a tumor associated antigen may be mAb-B43.13 (oregovomab), and the chemotherapeutic agent may be a combination of carboplatin and taxol.


The immune homeostatic checkpoint inhibitor may be an anti-PDL-1 antibody, an anti-CTLA-4 antibody, and anti-PD-1 antibody, or combinations thereof.


The anti-PDL-1 antibody may be selected from the group consisting of B7-H1 antibody, BMS-936559 antibody, MPDL3280A (atezolizumab) antibody, MEDI-4736 antibody, MSB0010718C antibody or combinations thereof.


The anti-CTLA-4 antibody may be selected from the group consisting of ipilimumab or tremelimumab or combinations thereof.


The anti-PD-1 antibody may be selected from the group consisting of Nivolumab antibody, pembrolizumab antibody, pidilizumab antibody or combinations thereof, and AMP-224.


The poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor may be selected from the group consisting of olaparib, niraparib, rucaparib, talazoparib, veliparib, CEP 9722, E7016, and BGB-290, or combinations thereof.


The dose of the monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region may be from about 0.1 to about 10 mg.


The monoclonal antibody may be for the treatment of ovarian cancer.


The use may be inducing a mannose receptor dependent immune response in the human patient.


The following terms are defined below.


The terms “administration of” and/or “administering a” is intended to mean providing an antibody according to the present invention with or without additional compound(s) to a subject in need of treatment. As used herein, it also refers to any action that results in exposing or contacting a composition containing a monoclonal antibody or a therapeutic monoclonal antibody specific for CA125 alone or in combination with an immune adjuvant, for example an immunostimulatory compound, an immune homeostatic checkpoint inhibitor. As used herein, administering may be conducted in vivo, in vitro, or ex vivo. For example, a composition may be administered by injection or through an endoscope. Administering also includes the direct application to cells of a composition according to the present invention. For example, during the course of surgery, tumor cells may be exposed. In accordance with an embodiment of the invention, these exposed cells (or tumors) may be exposed directly to a composition of the present invention, e.g., by washing or irrigating the surgical site and/or the cells, or by direct intra-tumoral injection of the therapeutic monoclonal antibody specific for CA125 alone or in combination with at least one immune adjuvant, and at least one immune homeostatic checkpoint inhibitor individually or in a mixture.


The term “composition” intended to mean a product comprising the specified ingredients in the specified amounts when specified, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. Such term in relation to pharmaceutical composition, is intended to encompass a product comprising the active ingredient(s) and the inert ingredient(s) that make up the pharmaceutically acceptable carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing an antibody according to the present invention and a pharmaceutically acceptable carrier. By “pharmaceutically acceptable” it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.


The term “immune adjuvant” is intended to mean a component that potentiates the immune responses to an antigen and/or modulates it towards the desired immune responses. It is a substance that acts to accelerate, prolong, or enhance antigen-specific immune responses when used in combination with specific antigens. In the context of the present invention, this includes chemotherapeutic agents such as for example platinum-based chemotherapy, taxol, doxorubicin, topotecan, a poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor, or combinations thereof, immunostimulatory compounds such as for example TLR3 agonist and TLR4 agonist, and their combinations, immune homeostatic checkpoint inhibitor such as for example anti-PDL-1 antibody, an anti-CTLA-4 antibody, and anti-PD-1 antibody, and their combinations; or a combination thereof. In embodiments, the immune adjuvant property(ies) of the immune adjuvant may be in addition to other therapeutic properties, such as for example cytotoxicity. That is to say, without wishing to be bound by theory, the immune adjuvants as described herein may not only act as an immune adjuvant, but may have other therapeutic properties for which, for example, it may be used in therapy. Such therapy may be, for example, as standard of care therapy for a given cancer.


In some embodiments, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term “carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin. Such compositions will contain a therapeutically effective amount of the antibody or fragment thereof, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.


The terms “inhibit”, “inhibition” or “inhibiting” as used herein in the context of the invention means to slow, hinder, restrain reduce or prevent. For example, “inhibiting growth” of a tumor cell as that term is used herein means to slow, hinder, restrain, reduce or prevent the tumor cell from growing.


The term “epitope” is intended to mean the portion of an antigen capable of being recognized by and bound by an antibody at one or more of the antibody's binding regions. Epitopes generally comprise chemically active surface groupings of molecules such as amino acids or sugar side chains and have specific three-dimensional structure characteristics as well as specific charge characteristics. In one embodiment, an epitope of an antigen is a repetitive epitope. In one embodiment, an epitope of an antigen is a non-repetitive epitope.


The term “subject” as used herein, is a human patient or other animal such as another mammal with functional mast cells, basophils, neutrophils, eosinophils, monocytes, macrophages, dendritic cells, and Langerhans cells. In humans, the appropriate cells express the high affinity receptor for IgG for the administered IgG antibody of the invention, as well as IgE (FcεRI) for the administered IgE antibody of the invention.


As used herein, a reduction in growth kinetics, or complete elimination of, a cancer tumor or a metastasized cell or tumor as used herein is defined to mean that which is as understood in the art. For example, a reduction in growth kinetics means a reduction in the exponential growth, specific growth rate, or doubling time of a primary solid tumor, metastasized cell, or metastasized tumor relative to the exponential growth, specific growth rate, or doubling time normally observed in vivo or in vitro for a given tumor type. Complete elimination of a tumor is the absence of tumor presence, either by symptoms, physical exam, or radiographic imaging, in the presence of the therapeutic monoclonal antibody specific for a tumor associated antigen in combination with at least one immunostimulatory compound, and at least one immune homeostatic checkpoint inhibitor, where a tumor was previously seen to be present by these detection methodologies.


The term “tumor-associated antigen” (TAA) as used herein can be any type of cancer antigen that may be associated with a tumor as is known in the art and includes antigens found on the cell surface, including tumor cells, as well as soluble cancer antigens. More specifically, the tumor associated antigen in the context of the present invention is CA125 (MUC16). Several cell surface antigens on tumors and normal cells have soluble counterparts. Such antigens include, but are not limited to those found on cancer-associated fibroblasts (CAFs), tumor endothelial cells (TEC) and tumor-associated macrophages (TAM). Examples of cancer-associated fibroblasts (CAFs) target antigens include but are not limited to: carbonic anhydrase IX (CAIX); fibroblast activation protein alpha (FAPα); and matrix metalloproteinases (MMPs) including MMP-2 and MMP-9. Examples of Tumor endothelial cell (TECs) target antigens include, but are not limited to vascular endothelial growth factor (VEGF) including VEGFR-1, 2, and 3; CD-105 (endoglin), tumor endothelia markers (TEMs) including TEM1 and TEM8; MMP-2; Survivin; and prostate-specific membrane antigen (PMSA). Examples of tumor associated macrophage antigens include, but are not limited to: CD105; MMP-9; VEGFR-1, 2, 3 and TEM8. According to some embodiments, the tumor associated antigen may be CA125, folate binding protein (FBP), HER2/neu, MUC1 or PSA.


The terms “monoclonal antibody” or “monoclonal antibodies” as used herein refer to a preparation of antibodies of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope. The monoclonal antibodies of the present invention have a xenogeneic Fc region, and may be chimeric. Monoclonal antibodies may be prepared by standard techniques including, but not limited to, recombinantly and synthetically.


The term “chimeric monoclonal antibody” refers to antibodies displaying a single binding specificity, which have one or more regions derived from one antibody and one or more regions derived from another antibody.


As used herein, “humanized” monoclonal antibodies comprise constant regions that are derived from human constant region (heavy chain) and human (light chain) constant regions. The variable regions of the antibodies preferably comprise a framework of human origin and antigen binding regions (CDRs) of non-human origin.


Fully human or human-like antibodies may be produced through vaccination of genetically engineered animals such as mouse lines produced at Amgen) and Bristol-Myers Squibb which contain the human immunoglobulin genetic repertoire and produce fully human antibodies in response to vaccination. Further, the use of phage display libraries incorporating the coding regions of human variable regions which can be identified and selected in an antigen-screening assay to produce a human immunoglobulin variable region binding to a target antigen.


The term “antigen binding region” refers to that portion of an antibody as used in the invention which contains the amino acid residues that interact with an antigen and confer on the antibody its specificity and affinity for the antigen. The antibody region includes the “framework” amino acid residues necessary to maintain the proper confirmation of the antigen binding residues.


An “antigen” is a molecule or portion of a molecule capable of being bound by an antibody, which is additionally capable of inducing an animal to produce antibody capable of binding to an epitope of that antigen. An antigen can have one or more epitopes that are the same or different. In a preferred embodiment, the antibodies of the invention are specific for a single epitope. In one embodiment, the antigen is a capable of being bound by an antibody as used in the invention to form an immune complex that in combination with at least one immunostimulatory compound, and at least one immune homeostatic checkpoint inhibitor, is capable of inhibiting cancer tumor growth. In one embodiment, the antigen, on its own, may not be capable of stimulating an immune response for any number of reasons, for example, the antigen is a “self” antigen, not normally recognized by the immune system as requiring response or the immune system has otherwise become tolerant to the antigen and does not mount an immune response. In another embodiment, the antigen is CA125.


The term “epitope” is meant to refer to that portion of an antigen capable of being recognized by and bound by an antibody at one or more of the antibody's binding regions. Epitopes generally comprise chemically active surface groupings of molecules such as amino acids or sugar side chains and have specific three-dimensional structure characteristics as well as specific charge characteristics. In one embodiment, an epitope of an antigen is a repetitive epitope. In one embodiment, an epitope of an antigen is a non-repetitive epitope.


The term “time sufficient for treatment” or “a time sufficient for treatment of the patient with the immune adjuvant” is intended to mean any period of time suitable to effect treatment with the immune adjuvant. In embodiments, that time period may be the time of a cycle used in standard to care for the immune adjuvant (e.g. chemotherapy). Examples of standard of care treatments may be found for example in Gynecologic Oncology Group Chemotherapy Procedures Manual, incorporated herein by reference. The length of chemotherapy treatment is determined by a variety of factors. These include the type of cancer, the extent of cancer, the types of drugs that are given, as well as the expected toxicities of the drugs and the amount of time necessary to recover from these toxicities. Many chemotherapy treatment schedules (often referred to as Standard of Care (SOC), including the type and length of chemotherapy treatment) have been determined through clinical trials that compared them and determined which had the most benefit and was most well tolerated. In general, chemotherapy treatment is given in cycles. This allows the cancer cells to be attacked at their most vulnerable times, and allows the body's normal cells time to recover from the damage. There are really three issues regarding the cycle time, duration of the cycle, frequency of the cycle, and how many cycles. Duration of the cycle: chemotherapy treatment may be a single drug or a combination of drugs. The drugs may all be given on a single day, several consecutive days, or continuously as an outpatient or as an inpatient. Treatment could last minutes, hours, or days, depending on the specific protocol. Frequency of the cycle: chemotherapy may repeat weekly, bi-weekly, or monthly. Usually, a cycle is defined in monthly intervals. For example, two bi-weekly chemotherapy sessions may be classified as one cycle. The number of cycles: In most cases, the number of cycles—or the length of chemotherapy from start to finish—has been determined by research and clinical trials. When cure is the treatment goal. Adjuvant chemotherapy (therapy after surgery has removed all visible cancer) may last 4-6 months. Adjuvant chemotherapy is common in cancers of the breast and colon. In cancers of the testis, Hodgkin and non-Hodgkin lymphoma, and leukemias, length of chemotherapy treatment may be up to a year. When there is visible disease, the length of chemotherapy treatment will depend upon the response of the disease to therapy. If the disease disappears completely, chemotherapy may continue for 1-2 cycles beyond this observation to maximize the chance of having attacked all microscopic disease. If the disease shrinks but does not disappear, chemotherapy will continue as long as it is tolerated and the disease does not grow. If the disease grows, the chemotherapy will be stopped. As patients experience toxicities and blood cell counts, the actual timing of the cycles is sometimes delayed according the necessities of each patient's circumstance. Depending on the health and wishes of the patient, either different drugs may be given to try to kill the cancer, or chemotherapy will be stopped and the goal changed to focus on patient comfort. In an embodiment, for example, the administration of the immune adjuvant therapy combining paclitaxel and carboplatin is often performed in cycles of about 21 days (3 weeks).


The term “optimally debulked” is intended to mean the process of surgically remove as much of the tumor as possible—in the present instance, the ovarian tumor—with the aim of leaving behind no tumor larger than 1 cm. In some embodiments, optimal debulking requires that a piece of colon be removed. In some embodiments, optimal debulking requires that a piece of the bladder be removed. In yet another embodiment, debulking may also require removing the spleen and/or the gallbladder, as well as part of the stomach, liver, and/or pancreas.


The term “CA125 level”, “CA125 antigen level in the blood above normal levels” or “units/mL of CA125” is intended to refer the amount CA125 antigen that is found in the blood of a subject or patient. The level of CA125 antigen found in the blood can be assessed by a number of clinical diagnostic tests. These tests measure the antigen level in subjects or patients, and provide results that fall either within the “normal” (i.e. values that are below, or up to a predetermined upper limit of normal (ULN) and are for patients that are not affected by disease) or “abnormal” (i.e. values that are above a predetermined upper limit of normal (ULN) and are for patients that are affected by disease) range for a given individual. As used herein, the expression “CA125 antigen level in the blood above normal levels”, or simply “above normal levels” refers to the a value measured by any given clinical diagnostic tests which is considered to be above the normal level according to that clinical diagnostic test. The units of measurement, and the values of these measurement from these clinical diagnostic tests may be different between one test and another, and absolute values may not be directly comparable, beyond the fact that they may lead to the same conclusion of a subject or patient being in the normal “non-affected” group, and the abnormal “affected” group. That being said, according to currently and commonly used diagnostic tests for measuring the levels of the CA125 antigen in the blood, the normal values for CA125 is from about 0 to about 35 units/mL. According to the present invention, the patients targeted by the present invention are ovarian cancer patients having CA125 levels more than 35 units/mL, or more than 36 units/mL, or more than 37 units/mL, or more than 38 units/mL, or more than 39 units/mL, or more than 40 units/mL, or more than 41 units/mL, or more than 42 units/mL, or more than 43 units/mL, or more than 44 units/mL, or more than 45 units/mL, or more than 46 units/mL, or more than 47 units/mL, or more than 48 units/mL, or more than 49 units/mL, or more than 50 units/mL, or more than 51 units/mL, or more than 52 units/mL, or more than 53 units/mL, or more than 54 units/mL, or more than 55 units/mL, or more than 56 units/mL, or more than 57 units/mL, or more than 58 units/mL, or more than 59 units/mL, or more than 60 units/mL, or more than 61 units/mL, or more than 62 units/mL, or more than 63 units/mL, or more than 64 units/mL, or more than 65 units/mL, or more than 66 units/mL, or more than 67 units/mL, or more than 68 units/mL, or more than 69 units/mL, or more than 70 units/mL, or in other words, above 35 units/mL, or above 36 units/mL, or above 37 units/mL, or above 38 units/mL, or above 39 units/mL, or above 40 units/mL, or above 41 units/mL, or above 42 units/mL, or above 43 units/mL, or above 44 units/mL, or above 45 units/mL, or above 46 units/mL, or above 47 units/mL, or above 48 units/mL, or above 49 units/mL, or above 50 units/mL, or above 51 units/mL, or above 52 units/mL, or above 53 units/mL, or above 54 units/mL, or above 55 units/mL, or above 56 units/mL, or above 57 units/mL, or above 58 units/mL, or above 59 units/mL, or above 60 units/mL, or above 61 units/mL, or above 62 units/mL, or above 63 units/mL, or above 64 units/mL, or above 65 units/mL, or above 66 units/mL, or above 67 units/mL, or above 68 units/mL, or above 69 units/mL, or above 70 units/mL, or of at least 36 units/mL, or of at least 37 units/mL, or of at least 38 units/mL, or of at least 39 units/mL, or of at least 40 units/mL, or of at least 41 units/mL, or of at least 42 units/mL, or of at least 43 units/mL, or of at least 44 units/mL, or of at least 45 units/mL, or of at least 46 units/mL, or of at least 47 units/mL, or of at least 48 units/mL, or of at least 49 units/mL, or of at least 50 units/mL, or of at least 51 units/mL, or of at least 52 units/mL, or of at least 53 units/mL, or of at least 54 units/mL, or of at least 55 units/mL, or of at least 56 units/mL, or of at least 57 units/mL, or of at least 58 units/mL, or of at least 59 units/mL, or of at least 60 units/mL, or of at least 61 units/mL, or of at least 62 units/mL, or of at least 63 units/mL, or of at least 64 units/mL, or of at least 65 units/mL, or of at least 66 units/mL, or of at least 67 units/mL, or of at least 68 units/mL, or of at least 69 units/mL, or of at least 70 units/mL.


Before describing the present invention in detail, a number of terms will be defined. As used herein, the singular forms “a”, “an”, and “the” include plural referents unless the context clearly dictates otherwise.


The use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one”, but it is also consistent with the meaning of “one or more”, “at least one”, and “one or more than one”. Similarly, the word “another” may mean at least a second or more.


As used in this specification and claim(s), the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “include” and “includes”) or “containing” (and any form of containing, such as “contain” and “contains”), are inclusive or open-ended and do not exclude additional, unrecited elements or process steps.


The term “about” is used to indicate that a value includes an inherent variation of error for the device or the method being employed to determine the value.


It is noted that terms like “preferably”, “commonly”, and “typically” are not utilized herein to limit the scope of the claimed invention or to imply that certain features are critical, essential, or even important to the structure or function of the claimed invention. Rather, these terms are merely intended to highlight alternative or additional features that can or cannot be utilized in a particular embodiment of the present invention.


For the purposes of describing and defining the present invention it is noted that the term “substantially” is utilized herein to represent the inherent degree of uncertainty that can be attributed to any quantitative comparison, value, measurement, or other representation. The term “substantially” is also utilized herein to represent the degree by which a quantitative representation can vary from a stated reference without resulting in a change in the basic function of the subject matter at issue.


Features and advantages of the subject matter hereof will become more apparent in light of the following detailed description of selected embodiments, as illustrated in the accompanying figures. As will be realized, the subject matter disclosed and claimed is capable of modifications in various respects, all without departing from the scope of the claims. Accordingly, the drawings and the description are to be regarded as illustrative in nature, and not as restrictive and the full scope of the subject matter is set forth in the claims.





BRIEF DESCRIPTION OF THE DRAWINGS

Further features and advantages of the present disclosure will become apparent from the following detailed description, taken in combination with the appended drawings, in which:


Further features and advantages of the present disclosure will become apparent from the following detailed description, taken in combination with the appended drawings, in which:



FIG. 1A illustrates CA125-B43.13 complex uptake. CA125 conjugated with FITC (light) pre-incubated with MAb-B43.13-Cy3 (red), then added to day 5 DC preparation for 30 min. before fixation; light indicates co-localized complex;



FIG. 1B illustrates uptake of antigen, antibody and antigen-antibody complexes in Monocytes, Immature and Mature Dendritic cells (DC). Monocytes and immature DC show enhanced uptake of CA125 in immune complex form. Only immature DC show effective binding of antibodies although monocytes express high amounts of FcγRI and II. Mature DC show low binding of CA125 and antibody and no uptake enhancement by immune complex formation.



FIG. 2 illustrates receptor expression of monocytes and dendritic cells;



FIG. 3 illustrates inhibition of antibody uptake in immature DC. Murine B43.13 is primarily inhibited by the anti-Mannose Receptor antibody; the FcR's and CR1 provide modest uptake. Chimeric B43.13 shows some uptake via FcRs, similar to murine B43.13;



FIG. 4 illustrates inhibition of CA125 and CA125-IC uptake in immature DC. CA125 alone shows no inhibition with any of the studied receptor-specific antibodies; CA125-immune complex (IC) with mIgG1 B43.13 are taken up via mannose receptor (MMR), FcγRI, and CR1; CA125-IC with hIgG3 B43.13 are taken up via FcγRII, but little via FcγRI and MMR;



FIG. 5 illustrates a schematic of the Frontline Chemoimmunotherapy Randomized Phase II trial according to an embodiment of the present invention;



FIG. 6 illustrates the time to clinical relapse in the ITT population, for the CIT treatment arm (full line) and the SOC treatment arm (dash line);



FIG. 7 illustrates relapse free survival in the ITT population, for the CIT treatment arm (full line) and the SOC treatment arm (dash line);



FIG. 8 illustrates overall survival in the ITT population, for the CIT treatment arm (full line) and the SOC treatment arm (dash line);



FIG. 9 illustrates the time to clinical relapse in the US patients part of the ITT population, for the CIT treatment arm (full line) and the SOC treatment arm (dash line);



FIG. 10 illustrates the time to clinical relapse in the Italian patients of the ITT population, for the CIT treatment arm (full line) and the SOC treatment arm (dash line);



FIG. 11 illustrates overall survival of patients with Tumor grade 3 and 4 from the patients in the study group, for the CIT treatment arm (full line) and the SOC treatment arm (dash line);



FIG. 12 illustrates time to clinical relapse of patients with FICO stage IIIC-IV from the patients in the study group, for the CIT treatment arm (full line) and the SOC treatment arm (dash line);



FIG. 13 illustrates a Forest plot of Time to clinical relapse (TTCR) hazard ratios by patient subgroups. Treatment 1=CIT; Treatment 2=SOC.



FIG. 14 illustrates a Forest plot of Relapse free survival (RFS) hazard ratios by patient subgroups. Treatment 1=CIT; Treatment 2=SOC.



FIG. 15 illustrates a Forest plot of overall survival (OS) hazard ratios by patient subgroups. Treatment 1=CIT; Treatment 2=SOC.



FIG. 16 illustrates the relapse free survival from the CIT group (left graph) and SOC group (right). The population from each group is segrated on the basis of the Neutrophil/lymphocyte ratio (NLR) at baseline and patients' clinical outcome in terms of RFS using a cut-off value of 3.612 for patients.





It will be noted that throughout the appended drawings, like features are identified by like reference numerals.


DETAILED DESCRIPTION

In embodiments there is disclosed a method for therapeutic indirect immunization of an optimally debulked human ovarian cancer patient having CA125 antigen level in the blood above normal levels, comprising administering to the human patient

    • a monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region;
    • an immune complex formed between CA125 antigen and the monoclonal antibody having at least a xenogeneic Fc region; or
    • a combination thereof.


In another embodiment, there is disclosed a method for therapeutic indirect immunization of an optimally debulked human ovarian cancer patient having CA125 antigen level in the blood above normal levels, comprising administering to the human patient

    • a monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region;
    • an immune complex formed between CA125 antigen and the monoclonal antibody having at least a xenogeneic Fc region; or
    • a fusion between the antigen and a xenogeneic Fc region and the CA125 antigen,
    • a combination thereof.


The CA125 antigen level in the blood above normal levels may be more than 35 units/mL of CA125 antigen in the blood. The CA125 antigen level in the blood above normal levels may be at least 50 units/mL of CA125 antigen in the blood.


The inventors have unexpectedly discovered that a monoclonal antibody specific for the CA125 antigen, having xenogeneic Fc region, can be used for the indirect immunization of human ovarian cancer patients to develop immunity against their individual tumors (a form of personalized therapy). Without being bound by theory, the combination of a monoclonal antibody specific for the CA125 antigen, having xenogeneic Fc region in accordance with the invention appears to be indirectly immunizing the subject, by establishing increased immunogenicity of their tumor antigen and tumor over a comparable humanized antibody used in a similar manner. The immune response to the tumor generated using the optimal schedule of the invention is capable of protecting subjects against growth of tumors. The invention is unique and unexpected in that it provides for a synergistic effect between the xenogeneic Fc region, and the Fab bound CA125 antigen, purportedly making the immune complex more antigenic and being differentially processed and thus differentially recognized by the antigen processing machinery of the patient, and therefore greatly enhance patient survival. Unexpectedly, in combination with immune adjuvants, the treatment dramatically improved clinical outcome in advanced ovarian cancer patients, especially over conventional therapy (Bookman et al. J Clin Oncol 27:1419-1425.). In particular, a direct comparison may be made between the study of Braly et al., where the present invention displays much improved progression-free survival.


In embodiments, in the method of the present invention the xenogeneic Fc region may be from an IgA, IgD, IgG or IgM isotype, preferably IgG isotype, and most preferably an IgG1 isotype. The xenogeneic Fc region may be a murine Fc region, a rat Fc region, a rabbit Fc region, a goat Fc region, a hamster Fc region, and preferably a murine Fc region. In an embodiment, the xenogeneic Fc region is a murine Fc region from an IgG isotype, preferably from the IgG1 isotype.


In a preferred embodiment, the antibody specific to CA125 (MUC16) is mAb-B43.13 (oregovomab), which is a murine IgG1 monoclonal antibody.


Methods for raising antibodies, such as murine antibodies to an antigen, and for determining if a selected antibody binds to a unique antigen epitope are well known in the art.


Screening for the desired antibody can be accomplished by techniques known in the art, e.g., radioimmunoassay, ELISA (enzyme-linked immunosorbant assay), “sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitin reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.


For preparation of monoclonal antibodies, any technique that provides for the production of antibody molecules by continuous cell lines in culture may be used (see, e.g., Antibodies—A Laboratory Manual, Harlow and Lane, eds., Cold Spring Harbor Laboratory Press: Cold Spring Harbor, N.Y., 1988). These include but are not limited to the hybridoma technique originally developed by Kohler and Milstein (1975, Nature 256:495-497), as well as the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today, 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). In an additional embodiment of the invention, monoclonal antibodies can be produced in germ-free animals utilizing recent technology (PCT/US90/02545). According to the invention, human antibodies may be used and can be obtained by using human hybridomas (Cote et al., 1983, Proc. Natl. Acad. Sci. U.S.A., 80:2026-2030) or by transforming human B cells with EBV virus in vitro (Cole et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, pp. 77-96). In fact, according to the invention, techniques developed for the production of “chimeric antibodies” (Morrison et al., 1984, J. Bacteriol. 159: 870; Neuberger et al., 1984, Nature 312:604-608; Takeda et al., 1985, Nature 314: 452-454) by splicing the genes from a mouse antibody molecule specific for a polypeptide together with genes from a human antibody molecule of appropriate biological activity can be used; such antibodies are within the scope of this invention.


In one embodiment, therapeutic monoclonal antibodies specific for a tumor associated antigen in accordance with the present invention are expressed by a positive transfectoma which is identified by enzyme-linked immunosorbent assay (ELISA) and Western Blot. The positive transfectoma will be cloned by limited dilution for highest productivity and selected for antibody production. As used herein a “transfectoma” includes recombinant eukaryotic host cells expressing the antibody, such as Chinese hamster ovary (CHO) cells and NS/O myeloma cells. Such transfectoma methodology is well known in the art (Morrison, S. (1985) Science, 229:1202). Previously published methodology used to generate mouse/human chimeric or humanized antibodies has yielded the successful production of various human chimeric antibodies or antibody fusion proteins (Helguera G, Penichet M L., Methods Mol. Med. (2005) 109:347-74).


In general, chimeric mouse-human monoclonal antibodies (i.e., chimeric antibodies) can be produced by recombinant DNA techniques known in the art. For example, a gene encoding the Fc constant region of a murine (or other species) monoclonal antibody molecule is digested with restriction enzymes to remove the region encoding the murine Fc, and the equivalent portion of a gene encoding a human Fc constant region is substituted, or vice versa. (See Robinson et al., International Patent Publication PCT/US86/02269; Akira, et al., European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496; Morrison et al., European Patent Application 173,494; Neuberger et al.; Cabilly et al. U.S. Pat. No. 4,816,567; Cabilly et al., European Patent Application 125,023; Better et al. (1988 Science, 240:1041-1043); Liu et al. (1987) PNAS, 84:3439-3443; Liu et al., 1987, J. Immunol., 139:3521-3526; Sun et al. (1987) PNAS, 84:214-218; Nishimura et al., 1987, Canc. Res., 47:999-1005; Wood et al. (1985) Nature, 314:446-449; and Shaw et al., 1988, J. Natl Cancer Inst., 80:1553-1559).


The chimeric antibody can be further humanized by replacing sequences of the Fv variable region which are not directly involved in antigen binding with equivalent sequences from human Fv variable regions. General reviews of humanized chimeric antibodies are provided by Morrison, S. L., 1985, Science, 229:1202-1207 and by Oi et al., 1986, Bio Techniques, 4:214. Those methods include isolating, manipulating, and expressing the nucleic acid sequences that encode all or part of immunoglobulin Fv variable regions from at least one of a heavy or light chain. Sources of such nucleic acid are well known to those skilled in the art and, for example, may be obtained from 7E3, an anti-GPIIbIIIa antibody producing hybridoma. The recombinant DNA encoding the chimeric antibody, or fragment thereof, can then be cloned into an appropriate expression vector. Suitable humanized antibodies can alternatively be produced by CDR substitution (U.S. Pat. No. 5,225,539; Jones et al. 1986 Nature, 321:552-525; Verhoeyan et al. 1988 Science, 239:1534; and Beidler et al. 1988 J. Immunol., 141:4053-4060).


As used herein, an “effective amount” of the therapeutic monoclonal antibody specific to CA125 of the invention is the amount sufficient to recognize and bind the epitope of the CA125 (that is a cell surface antigen) and induce, elicit, or enhance the referenced indirect immune response in accordance with the invention. In according to embodiments, the administration of the therapeutic monoclonal antibody may be one, two, three, or four administrations. According to another embodiment, the administration of the therapeutic monoclonal antibody may be five, six or more administrations.


When used for therapy for the treatment of cancer, the antibodies used in the invention are administered to the patient in therapeutically effective amounts (i.e. amounts needed to treat clinically apparent tumors, or prevent the appearance of clinically apparent tumor, either at the original site or a distant site, at some time point in the future). The antibodies used in the invention and the pharmaceutical compositions containing them will normally be administered parenterally, when possible, or at the target cell site, or intravenously.


According to another embodiment, in the method of the present invention, the dose of the monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region may be from about 0.1 to about 10 mg, and in a preferred embodiment, about 2 mg. Lower dosages of the antibodies of the invention and less frequent administration may also be possible.


According to another embodiment, the method of the present invention may further comprise the administration of an immune adjuvant.


In embodiments, the immune adjuvant may be a chemotherapeutic agent, an immunostimulatory compound, an immune homeostatic checkpoint inhibitor, or a combination thereof. According to an embodiment, the chemotherapeutic agent may be a platinum-based chemotherapy, such as for example cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin, satraplatin, and combinations thereof. It may also be taxol (paclitaxel), doxorubicin, topotecan, a poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor, or combinations of each of the above chemotherapeutic agents. Examples of PARP inhibitors include, but are not limited to olaparib (AZD-2281 or Lynparza™), niraparib (MK-4827), rucaparib (AG014699 or Rubraca™), talazoparib (BMN-673), veliparib (ABT-888), CEP 9722, E7016, and BGB-290. Also contemplated as chemotherapeutic agents are cytotoxic therapeutic agents which include, but are not limited to, angiogenesis inhibitors, antiproliferative agents, kinase inhibitors, receptor tyrosine kinase inhibitors, aurora kinase inhibitors, polo-like kinase inhibitors, bcr-abl kinase inhibitors, growth factor inhibitors, COX-2 inhibitors, non-steroidal anti-inflammatory drugs (NSAIDS), antimitotic agents, alkylating agents, antimetabolites, intercalating antibiotics, platinum containing agents, growth factor inhibitors, ionizing radiation, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biologic response modifiers, immunologicals, antibodies, hormonal therapies, retinoids/deltoids plant alkaloids, proteasome inhibitors, HSP-90 inhibitors, histone deacetylase inhibitors (HDAC) inhibitors, purine analogs, pyrimidine analogs, MEK inhibitors, CDK inhibitors, ErbB2 receptor inhibitors, mTOR inhibitors and combinations thereof as well as other antitumor agents.


Angiogenesis inhibitors include, but are not limited to, EGFR inhibitors, PDGFR inhibitors, VEGFR inhibitors, TTE2 inhibitors, IGFIR inhibitors, matrix metalloproteinase 2 (MMP-2) inhibitors, matrix metalloproteinase 9 (MMP-9) inhibitors, thrombospondin analogs such as thrombospondin-1 and N-Ac-Sar-Gly-Val-D-allolle-Thr-Nva-He-Arg-Pro-NHCH2CH3 or a salt thereof and analogues of N-Ac-Sar-Gly-Val-D-allolle-Thr-Nva-Ile-Arg-PrO-NHCH2CH3 such as N-Ac-GlyVal-D-alle-Ser-Gln-Ile-Arg-ProNHCH2CH3 or a salt thereof.


Examples of EGFR inhibitors include, but are not limited to, Iressa (gefitinib), Tarceva (erlotinib or OSI-774), Icotinib, Erbitux (cetuximab), EMD-7200, ABX-EGF, HR3, IgA antibodies, TP-38 (IVAX), EGFR fusion protein, EGF-vaccine, anti-EGFr immunoliposomes and Tykerb (lapatinib).


Examples of PDGFR inhibitors include, but are not limited to, CP-673,451 and CP-868596.


Examples of VEGFR inhibitors include, but are not limited to, Avastin (bevacizumab), Sutent (sunitinib, SUI 1248), Nexavar (sorafenib, BAY43-9006), CP-547,632, axitinib (AG13736), Apatinib, cabozantinib, Zactima (vandetanib, ZD-6474), AEE788, AZD-2171, VEGF trap, Vatalanib (PTK-787, ZK-222584), Macugen, M862, Pazopanib (GW786034), ABT-869 and angiozyme.


Examples of thrombospondin analogs include, but are not limited to, TSP-I and ABT-510.


Examples of aurora kinase inhibitors include, but are not limited to, VX-680, AZD-1152 and MLN-8054. Example of polo-like kinase inhibitors include, but are not limited to, BI-2536.


Examples of bcr-abl kinase inhibitors include, but are not limited to, Gleevec (imatinib) and Dasatinib (BMS354825).


Examples of platinum containing agents includes, but are not limited to, cisplatin, Paraplatin (carboplatin), eptaplatin, lobaplatin, nedaplatin, Eloxatin (oxaliplatin) or satraplatin.


Examples of mTOR inhibitors includes, but are not limited to, CCI-779, rapamycin, temsirolimus, everolimus, RAD001, INK-128 and ridaforolimus.


Examples of HSP-90 inhibitors includes, but are not limited to, geldanamycin, radicicol, 17-AAG, KOS-953, 17-DMAG, CNF-101, CNF-1010, 17-AAG-nab, NCS-683664, Mycograb, CNF-2024, PU3, PU24FC1, VER49009, IPI-504, SNX-2112 and STA-9090.


Examples of histone deacetylase inhibitors (HDAC) includes, but are not limited to, Suberoylanilide hydroxamic acid (SAHA), MS-275, valproic acid, TSA, LAQ-824, Trapoxin, tubacin, tubastatin, ACY-1215 and Depsipeptide.


Examples of MEK inhibitors include, but are not limited to, PD325901, ARRY-142886, ARRY-438162 and PD98059.


Examples of CDK inhibitors include, but are not limited to, flavopyridol, MCS-5A, CVT-2584, seliciclib (CYC-202, R-roscovitine), ZK-304709, PHA-690509, BMI-1040, GPC-286199, BMS-387,032, PD0332991 and AZD-5438.


Examples of COX-2 inhibitors include, but are not limited to, CELEBREX™ (celecoxib), parecoxib, deracoxib, ABT-963, MK-663 (etoricoxib), COX-189 Lumiracoxib), BMS347070, RS 57067, NS-398, Bextra (valdecoxib), paracoxib, Vioxx (rofecoxib), SD-8381, 4-Methyl-2-(3,4-dimethylphenyl)-1-(4-sulfamoyl-phenyl-1H-pyrrole, T-614, JTE-522, S-2474, SVT-2016, CT-3, SC-58125 and Arcoxia (etoricoxib).


Examples of non-steroidal anti-inflammatory drugs (NSAIDs) include, but are not limited to, Salsalate (Amigesic), Diflunisal (Dolobid), Ibuprofen (Motrin), Ketoprofen (Orudis), Nabumetone (Relafen), Piroxicam (Feldene), Naproxen (Aleve, Naprosyn), Diclofenac (Voltaren), Indomethacin (Indocin), Sulindac (Clinoril), Tolmetin (Tolectin), Etodolac (Lodine), Ketorolac (Toradol) and Oxaprozin (Daypro).


Exambles of ErbB2 receptor inhibitors include, but are not limited to, CP-724-714, CI-1033, (canertinib), Herceptin (trastuzumab), Omitarg (2C4, petuzumab), TAK-165, GW-572016 (Ionafarnib), GW-282974, EKB-569, PI-166, dHER2 (HER2 Vaccine), APC8024 (HER2 Vaccine), anti-HER/2neu bispecific antibody, B7.her2IgG3, AS HER2 trifunctional bispecfic antibodies, mAB AR-209 and mAB 2B-1.


Examples of alkylating agents include, but are not limited to, nitrogen mustard N-oxide, cyclophosphamide, ifosfamide, trofosfamide, Chlorambucil, melphalan, busulfan, mitobronitol, carboquone, thiotepa, ranimustine, nimustine, temozolomide, AMD-473, altretamine, AP-5280, apaziquone, brostallicin, bendamustine, carmustine, estramustine, fotemustine, glufosfamide, KW-2170, mafosfamide, and mitolactol, carmustine (BCNU), lomustine (CCNU), Busulfan, Treosulfan, Decarbazine and Temozolomide.


Examples of antimetabolites include but are not limited to, methotrexate, 6-mercaptopurine riboside, mercaptopurine, uracil analogues such as 5-fluorouracil (5-FU) alone or in combination with leucovorin, tegafur, UFT, doxifluridine, carmofur, cytarabine, cytarabine ocfosfate, enocitabine, S-I, Alimta (premetrexed disodium, LY231514, MTA), Gemzar (gemcitabine), fludarabine, 5-azacitidine, capecitabine, cladribine, clofarabine, decitabine, eflornithine, ethnylcytidine, cytosine arabinoside, hydroxyurea, TS-I, melphalan, nelarabine, nolatrexed, ocfosate, disodium premetrexed, pentostatin, pelitrexol, raltitrexed, triapine, trimetrexate, vidarabine, vincristine, vinorelbine, mycophenolic acid, tiazofurin, Ribavirin, EICAR, hydroxyurea and deferoxamine.


Examples of antibiotics include intercalating antibiotics but are not limited to, aclarubicin, actinomycins such as actinomycin D, amrubicin, annamycin, adriamycin, bleomycin a, bleomycin b, daunorubicin, doxorubicin, elsamitrucin, epirbucin, glarbuicin, idarubicin, mitomycin C, nemorubicin, neocarzinostatin, peplomycin, pirarubicin, rebeccamycin, stimalamer, streptozocin, valrubicin, zinostatin and combinations thereof.


Examples of topoisomerase inhibiting agents include, but are not limited to, one or more agents selected from the group consisting of aclarubicin, amonafide, belotecan, camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, diflomotecan, irinotecan HCL (Camptosar), edotecarin, epirubicin (Ellence), etoposide, exatecan, gimatecan, lurtotecan, orathecin (Supergen), BN-80915, mitoxantrone, pirarbucin, pixantrone, rubitecan, sobuzoxane, SN-38, tafluposide and topotecan.


Examples of antibodies include, but are not limited to, Rituximab, Cetuximab, Bevacizumab, Trastuzimab, specific CD40 antibodies and specific IGFIR antibodies,


Examples of hormonal therapies include, but are not limited to, exemestane (Aromasin), leuprolide acetate, anastrozole (Arimidex), fosrelin (Zoladex), goserelin, doxercalciferol, fadrozole, formestane, tamoxifen citrate (tamoxifen), Casodex, Abarelix, Trelstar, finasteride, fulvestrant, toremifene, raloxifene, lasofoxifene, letrozole, flutamide, bicalutamide, megesterol, mifepristone, nilutamide, dexamethasone, predisone and other glucocorticoids.


Examples of retinoids/deltoids include, but are not limited to, seocalcitol (EB 1089, CB 1093), lexacalcitrol (KH 1060), fenretinide, Aliretinoin, Bexarotene and LGD-1550.


Examples of plant alkaloids include, but are not limited to, vincristine, vinblastine, vindesine and vinorelbine.


Examples of proteasome inhibitors include, but are not limited to, bortezomib (Velcade), MGI 32, NPI-0052 and PR-171.


Examples of immunologicals include, but are not limited to, interferons and numerous other immune enhancing agents. Interferons include interferon alpha, interferon alpha-2a, interferon, alpha-2b, interferon beta, interferon gamma-1a, interferon gamma-1b (Actimmune), or interferon gamma-nl and combinations thereof. Other agents include filgrastim, lentinan, sizofilan, TheraCys, ubenimex, WF-10, aldesleukin, alemtuzumab, BAM-002, decarbazine, daclizumab, denileukin, gemtuzumab ozogamicin, ibritumomab, imiquimod, lenograstim, lentinan, melanoma vaccine (Corixa), molgramostim, OncoVAC-CL, sargaramostim, tasonermin, tecleukin, thymalasin, tositumomab, Virulizin, Z-100, epratuzumab, mitumomab, oregovomab, pemtumomab (Y-muHMFGI), Provenge (Dendreon), CTLA4 (cytotoxic lymphocyte antigen 4) antibodies and agents capable of blocking CTLA4 such as MDX-010.


Examples of biological response modifiers are agents that modify defense mechanisms of living organisms or biological responses, such as survival, growth, or differentiation of tissue cells to direct them to have anti-tumor activity. Such agents include krestin, lentinan, sizofrran, picibanil and ubenimex.


Examples of pyrimidine analogs include, but are not limited to, 5-Fluorouracil, Floxuridine, Doxifluridine, Ratitrexed, cytarabine (ara C), Cytosine arabinoside, Fludarabine, and Gemcitabine.


Examples of purine analogs include but are not limited to, Mercaptopurine and thioguanine.


Examples of antimitotic agents include, but are not limited to, ABT-751, paclitaxel, docetaxel, epothilone D (KOS-862) and ZK-EPO.


In a preferred embodiment, the chemotherapeutic agent is a combination of carboplatin and taxol. In another preferred embodiment, the therapeutic monoclonal antibody specific for a tumor associated antigen is mAb-B43.13 (oregovomab), and the chemotherapeutic agent is a combination of carboplatin and taxol


In embodiments, the immune adjuvant may also be an immunostimulatory compound. According to an embodiment, the present invention includes immunostimulatory compounds. Immunostimulatory compounds are compound having the capacity to stimulate or elicit an immune response. As used herein, the term relates to exemplary immunostimulatory compounds that include toll-like receptor (TLR) agonists (e.g., TLR3, TLR4, TLR7, TLR9), N-acetylmuramyl-L-alanine-D-isoglutamine (MDP), lipopolysaccharides (LPS), genetically modified and/or degraded LPS, alum, glucan, colony stimulating factors (e.g., EPO, GM-CSF, G-CSF, M-CSF, pegylated G-CSF, SCF, IL-3, IL6, PIXY 321), interferons (e.g., gamma-interferon, alpha-interferon), interleukins (e.g., IL-2, IL-7, IL-12, IL-15, IL-18), MHC Class II binding peptides, saponins (e.g., QS21), unmethylated CpG sequences, 1-methyl tryptophan, arginase inhibitors, cyclophosphamide, antibodies that block immunosuppressive functions (e.g., anti-CTLA4 antibodies, anti-TGF-beta, etc.), and mixtures of two or more thereof.


In one preferred embodiment the immunostimulatory compound is a TLR3 agonist. In preferred embodiments, the TLR3 agonist for use according to the invention is a double stranded nucleic acid selected from the group consisting of: polyinosinic acid and polycytidylic acid, polyadenylic acid and polyuridylic acid, polyinosinic acid analogue and polycytidylic acid, polyinosinic acid and polycytidylic acid analogue, polyinosinic acid analogue and polycytidylic acid analogue, polyadenylic acid analogue and polyuridylic acid, polyadenylic acid and polyuridylic acid analogue, and polyadenylic acid analogue and polyuridylic acid analogue. Specific examples of double-stranded RNA as TLR3 agonists further include Polyadenur (Ipsen) and Ampligen (Hemispherx). Polyadenur is a polyA/U RNA molecule, i.e., contains a polyA strand and a polyU strand. Ampligen is disclosed for instance in EP 281 380 or EP 113 162. In another preferred embodiment, the TLR3 agonist may be Poly (I:C)LC or polyIC (Hiltonol®), which is a synthetic complex of carboxymethylcellulose, polyinosinic-polycytidylic acid, and poly-L-lysine double-stranded RNA. Poly (I:C)LC may stimulate the release of cytotoxic cytokines and, by inducing interferon-gamma production, may increase the tumoricidal activities of various immunohematopoietic cells.


In one embodiment the immunostimulatory compound is a TLR4 agonist. Exemplary TLR4 agonists include taxanes such as paclitaxel and docetaxal, lipopolysaccharides (LPS); E. coli LPS; and P. gingivalis LPS.


As used herein, an “effective amount” of an immunostimulatory compound of the invention is that amount sufficient to induce, elicit, or enhance the referenced immune response in accordance with the invention.


According to another embodiment, the present invention includes immune homeostatic checkpoint inhibitors. Immune homeostatic checkpoint inhibitors are monoclonal antibodies (mAb) directed to immune checkpoint molecules, which are expressed on immune cells and mediate signals to attenuate excessive immune reactions. According to an embodiment, immune homeostasis checkpoint inhibition may be performed with inhibitory monoclonal antibodies directed at the inhibitory immune receptors CTLA-4, PD-1, and PDL-1. According to some embodiments, such inhibitors have emerged as successful treatment approaches for patients with advanced melanoma. According to an embodiment, the immune homeostatic checkpoint inhibitors may be one of an anti-CTLA-4, anti-PD-1, and/or anti-PDL-1 antibody. According to an embodiment, the anti-CTLA-4 antibody may be Ipilimumab or tremelimumab or combinations thereof. According to another embodiment, the anti-PDL-1 antibody may be B7-H1 antibody, BMS-936559 antibody, MPDL3280A (atezolizumab) antibody, MEDI-4736 antibody, MSB0010718C antibody or combinations thereof. According to another embodiment, the anti-PD-1 antibody may be Nivolumab antibody, pembrolizumab antibody, pidilizumab antibody or combinations thereof. In addition, PD-1 may also be targeted with AMP-224, which is a PD-L2-IgG recombinant fusion protein. Additional antagonists of inhibitory pathways in the immune response are being advanced through clinical development. IMP321 is a soluble LAG-3 Ig fusion protein and MHC class II agonist, which is used to increase an immune response to tumors. LAG3 is an immune checkpoint molecule. Lirilumab is an antagonist to the KIR receptor and BMS 986016 is an antagonist of LAG3. A third inhibitory checkpoint pathway is the TIM-3-Galectin-9 pathway that is also a promising target for checkpoint inhibition. RX518 targets and activates the glucocorticoid-induced tumor necrosis factor receptor (GITR), a member of the TNF receptor superfamily that is expressed on the surface of multiple types of immune cells, including regulatory T cells, effector T cells, B cells, natural killer (NK) cells, and activated dendritic cells.


As used herein, an “effective amount” of an immune homeostatic checkpoint inhibitor of the invention is that amount sufficient to induce, elicit, or enhance the referenced immune response in accordance with the invention.


According to another embodiment, the method of the present invention is for the treatment of ovarian cancer. In another embodiment, the therapeutic indirect immunization may be inducing a mannose receptor dependent immune response in the human patient.


According to yet other embodiments, the present invention also encompasses vaccine compositions or monoclonal antibody specific to CA125 antigen, having at least a xenogeneic Fc region, for use in indirect immunization of an optimally debulked human ovarian cancer patient having CA125 antigen level in the blood above normal levels, comprising a monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region, an immune complex formed between CA125 antigen and said monoclonal antibody having at least a xenogeneic Fc region, or a combination thereof.


The CA125 antigen level in the blood above normal levels may be more than 35 units/mL of CA125 antigen in the blood. The CA125 antigen level in the blood above normal levels may be at least 50 units/mL of CA125 antigen in the blood.


According to yet other embodiments, the present invention also encompasses the use of a monoclonal antibody specific to CA125 antigen, having at least a xenogeneic Fc region, for therapeutic indirect immunization of an optimally debulked human ovarian cancer patient having CA125 antigen level in the blood above normal levels, comprising a monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region, an immune complex formed between CA125 antigen and said monoclonal antibody having at least a xenogeneic Fc region, or a combination thereof.


The CA125 antigen level in the blood above normal levels may be more than 35 units/mL of CA125 antigen in the blood. The CA125 antigen level in the blood above normal levels may be at least 50 units/mL of CA125 antigen in the blood.


Such vaccine or antibody compositions comprise a therapeutically effective amount of a therapeutic monoclonal antibody specific to CA125 antigen, having at least a xenogeneic Fc region, and may also include a pharmaceutically acceptable carrier.


In accordance with a method, vaccine composition for use, monoclonal antibody for use, or use of the invention compositions comprising the therapeutic monoclonal antibody specific to CA125 antigen, having at least a xenogeneic Fc region, in combination with an immune adjuvant, the immune adjuvant of the invention may be administered to the patient by any immunologically suitable route. For example, they may be introduced into the patient by an intravenous, subcutaneous, intraperitoneal, intrathecal, intravesical, intradermal, intramuscular, or intralymphatic routes, alone or as combination. The composition may be in solution, tablet, aerosol, or multi-phase formulation forms. Liposomes, long-circulating liposomes, immunoliposomes, biodegradable microspheres, micelles, or the like may also be used as a carrier, vehicle, or delivery system. Furthermore, using ex vivo procedures well known in the art, blood or serum from the patient may be removed from the patient; optionally, it may be desirable to purify the antigen in the patient's blood; the blood or serum may then be mixed with a composition that includes a binding agent according to the invention; and the treated blood or serum is returned to the patient. The invention should not be limited to any particular method of introducing the binding agent into the patient.


In a preferred embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.


The compositions of the invention can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.


The amount of the composition of the invention which will be effective in the indirect immunization, or the treatment, inhibition and prevention of tumor growth associated with the antigen to which the antibody of the invention is specific can be determined by standard clinical techniques. The presence of the antibody in the extra vascular space, can be assayed by standard skin wheal and flair responses, in response to intradermal administration of purified antigen (e.g. CA125). In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.


For the chemotherapeutic agents used in the invention, the dosage administered to a patient may be according to the ranges or concentrations that have been optimized by their respective manufacturers.


For the immunostimulatory compound used in the invention, the dosage administered to a patient may be according to the ranges or concentrations that have been optimized by their respective manufacturers.


For the immune homeostatic checkpoint inhibitor used in the invention, the dosage administered to a patient may be according to the ranges or concentrations that have been optimized by their respective manufacturers.


In embodiments, in the case of treatment of ovarian cancer, the disease is treated according to standard of care treatment(s), where dosages of therapeutics administered to a patient may be according to the ranges or concentrations that have been optimized by their respective manufacturers.


The pharmaceutical compositions of the present invention have in vitro and in vivo diagnostic and therapeutic utilities. For example, these molecules can be administered to cells in culture, e.g., in vitro or ex vivo, or in a subject, e.g., in vivo, to treat cancer. As used herein, the term “subject” is intended to include human and non-human animals. A preferred subject is a human patient with cancer. As used herein the terms “treat” “treating” and “treatment” of cancer includes: preventing the appearance of tumor metastasis in a patient, inhibiting the onset of cancer in a patient; eliminating or reducing a preexisting tumor burden in a patient either with metastatic cancer or cancer localized to the organ of origin; prolonging survival in a cancer patient; prolonging the remission period in a cancer patient following initial treatment with chemotherapy and/or surgery; and/or prolonging any period between cancer remission and cancer relapse in a patient.


According to yet another embodiment, the present invention also encompasses kits for use for the indirect immunization of an optimally debulked human ovarian cancer patient, or inhibiting cancer tumor growth in said patient in need thereof. The kits may comprise a therapeutic monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region, with or without at least one immunostimulatory compound, at least one immune homeostatic checkpoint inhibitor, and instructions on how to use the kit.


The present invention will be more readily understood by referring to the following examples which are given to illustrate the invention rather than to limit its scope.


EXAMPLE 1
Receptor Identification Studies

Materials: Leukaphoresis samples from healthy normal donors were obtained from Biological Speciality Corporation (Colmar, Pa.). Peripheral Blood Leukocytes (PBL) were purified on Ficoll (Histopaque 1.077, Sigma). MAb-B43.13 is a murine monoclonal IgG1 antibody to CA125 with high affinity (1.1×1010 M−1). MAb-AR47.47 is an IgG1 specific for the PSA epitope EEPEFLTPKKL (AltaRex Corp.). CA125 was purified from tissue culture supernatant of NIH:OVCAR-3 cells (AltaRex Corp.). PSA was purified from human seminal plasma (Scripps).


Dendritic Cell Preparation: Human DC were prepared from normal human PBL by Ficoll-Hypaque and negative selection with anti-CD3, CD16 and CD19, followed by anti-mouse-Ig-magnetic beads (Dynal) to obtain approximately 70% pure monocytes. Cells were cultured in GM-CSF and IL-4 (1000 U/ml each, R&D Systems) for 4 days to generate immature DC. DC were matured with TNF-α (10 ng/ml) and IFN-α (50 U/ml) for 3 days.


Antibody Binding Studies to Monocytes and DC: Monocytes (day 0), immature DC (day 4) and mature DC (day 7) were incubated with FITC- or Cy3-labeled antibody or antigen for 1 h at 37° C. In some cases, unlabeled antigen or antibody was added simultaneously to study the effect of complex formation on the uptake by the two antigen-presenting cells. Binding was quantified by flow cytometry.


Fluorescently labeled antigens/antibodies were incubated in the presence of 10× excess of blocking antibodies (anti-CD16, anti-CD32 and anti-CD64 for FcγRIII, II and I receptors, anti CD11b/CD18 (Mac-1), anti-CD11c/CD18 (p150.95), anti-CD35 and anti-CD21 for complement receptors CR3, CR4, CR1 and CR2, anti-mannose receptor for mannose receptor) or inhibitor of macropinocytosis (wortmannin) and endocytosis (sucrose, 4° C.).


Now referring to FIG. 1A, the figure shows that dendritic cells uptake of CA125-B43.13 complexes. The light dots illustrate the co-localization of CA125 with B43.13. In FIG. 1B, it is shown that monocytes and immature DC show enhanced uptake of CA125 in immune complex form, and that immature DC show effective binding of antibodies although monocytes express high amounts of FcγRI and II. Lastly, mature DC show low binding of CA125 and antibody and no uptake enhancement by immune complex formation.


Now referring to FIG. 2, it is shown that DC express varying amounts of FcγRI and II, complement receptors 1, 3 and 4 and mannose receptor depending on their maturation stage.


Now referring to FIG. 3, it is shown that murine B43.13 binding to immature DC is primarily inhibited by the anti-Mannose Receptor antibody; the FcR's and CR1 also contribute to binding and uptake but less so than the mannose receptor uptake. Chimeric B43.13 having a hIgG3 Fc region shows some uptake via FcRs, similar to murine B43.13 but with a different relative profile and minimal use of the mannose receptor pathway. As shown in FIG. 4, CA125 alone shows no inhibition with any of the studied receptor-specific antibodies. CA125-IC with mIgG1 B43.13 are taken up via MMR, FcγRI, and CR1; CA125-IC with hIgG3 B43.13 are taken up via FcγRII, but little via FcγRI and MMR.


EXAMPLE 2
Combining Front Line Chemotherapy and Immunotherapy

Now referring to FIG. 5, which is a schematic of the Frontline Chemoimmunotherapy Randomized Phase II trial according to an embodiment of the present invention.


Design: Phase II Randomized trial (centers in US and Italy).


Patients: Initial treatment of newly diagnosed optimally debulked stage III/IV Ovarian cancer expressing CA125 (MUC16) at least 2× normal at baseline (70 units/mL, although a few patients with lower values were also included in the study).


Treatment: Standard of care (SOC) chemotherapy (6 cycles IV carboplatin-paclitaxel) (Control) vs SOC chemotherapy plus oregovomab immunotherapy (IT) (CIT)


Schedule: In CIT group oregovomab is administered at cycles 1, 3, 5 in combination with the SOC chemotherapy, as well as at cycle 5 plus 12 weeks as a single oregovomab immunotherapy immunization (without SOC chemotherapy). Initial Analysis post completion of treatment phase. Final Analysis post 3 year follow up.


Endpoints: Safety, Immune Response and Clinical Outcomes (TTCR, PFS and OS).


A total of 97 patients were enrolled in the intent to treat (ITT) population.


Oregovomab MAb-B43.13

All patients in the CIT group enrolled in this study were to receive single IV doses of MAb-B43.13 containing 2 mg of the monoclonal antibody concurrent with conventional chemotherapy Cycles 1, 3 and 5. Subsequently all patients in the CIT group were to receive single IV doses of MAb-B43.13 at Cycle +12 weeks in the post-chemotherapy phase (without concurrent chemotherapy).


The lyophilized contents of a vial of MAb-B43.13 were to be dissolved in 2 mL of 0.9% Sodium Chloride Injection USP (or equivalent). The vial contents were to be mixed by gentle swirling to avoid the formation of foam and then examined to ensure that the solution was free of foreign or particulate matter. The resulting solution was to be withdrawn from the vial with a suitable needle and syringe and added to 50 mL of 0.9% Sodium Chloride Injection USP (or equivalent) in a small (50 mL) infusion bag.


Each dose of MAb-B43.13 (containing 2 mg of MAb-B43.13 in 50 mL of Sodium Chloride Injection USP) was to be administered to the patient by slow (20 minutes) IV infusion in an appropriate treatment area. The dose of MAb-B43.13 was to be administered after paclitaxel but prior to carboplatin.


Chemotherapy

Maximum body surface area (BSA) used for chemotherapy dose calculations was to be determined per acceptable standard (e.g., Gynecologic Oncology Group Chemotherapy Procedures Manual). Maximum creatinine clearance was to be 120 mL/min for the purpose of this study.


Paclitaxel

Paclitaxel is supplied as a sterile solution concentrate, 6 mg/mL, in 5 mL vials (30 mg/vial) or 17 mL vials (100 mg/vial) in polyoxyethylated castor oil (Cremophor EL) 50% and dehydrated alcohol, USP, 50%, was to be used for this trial. The appropriate dose of paclitaxel was to be diluted in 500-1000 mL of 9% Sodium Chloride injection, USP or 5% Dextrose injection, USP (D5W). Paclitaxel was to be prepared in glass or polyolefin containers due to leaching of diethylhexylphthalate plasticizer from polyvinyl chloride (PVC) bags and intravenous tubing by the Cremophor vehicle in which paclitaxel is solubilized.


Paclitaxel, at a dose of 175 mg/m2, was to be administered via an infusion control device (pump) using non-PVC tubing and connectors, as a 3-hour continuous IV infusion. In-line filtration was necessary for administration of paclitaxel solutions. Due to the risk of immediate hypersensitivity reaction, paclitaxel should always be the first drug to be infused during any combination.


Carboplatin

Carboplatin is supplied as a sterile lyophilized powder available in single-dose vials containing 50 mg, 150 mg and 450 mg of carboplatin for administration by IV infusion. Each vial contains equal parts by weight of carboplatin and mannitol.


Immediately prior to use, the contents of each vial was to be reconstituted with either sterile water for injection, USP, 5% dextrose in water, or 9% sodium chloride injection, USP, according to the following schedule: 50 mg vial with 5 mL, 150 mg vial with 15 mL and 450 mg vial with 45 mL, all producing a concentration of 10 mg/m L.


The dose of carboplatin was to be calculated to reach a target area under the curve (AUC) of concentration x time of 6 according to the Calvert formula using an estimated glomerular filtration rate (GFR) from the Jelliffe formula for creatinine clearance (CrCl).





Calvert Formula: Carboplatin dose (mg)=target AUC×(GFR+25)


For the purposes of this protocol, the GFR was considered to be equivalent to the CrCl. The creatinine clearance was to be estimated by the method of Jelliffe using the following formula:






CrCl
=

0.9
×


[

98
-

(

0.8






(

Age
-
20

)


)


]


Serum





Creatinine







Where: CrCl=estimated creatinine clearance in mL/min; Age=patient's age in years; serum creatinine in mg/dL.


The initial dose of carboplatin was to be calculated using GFR. In the absence of new renal obstruction or other renal toxicity (i.e., serum creatinine>1.5×ULN), the dose of carboplatin was not to be recalculated for subsequent cycles, but was to be subject to dose modification for hematologic criteria and other events.


In patients with an abnormally low serum creatinine, due to reduced protein intake and/or low muscle mass, the creatinine clearance (CrCl) was to be determined from a 24 hour urine collection, rather than a Jelliffe formula.


Carboplatin was to be administered as a 30 minute IV infusion. When administered in conjunction with other medications, carboplatin was to be infused after the other agents.









TABLE 1







Demographics analysis of the ITT population









ITT Population











Treatment
Treatment




Arm 1 CIT
Arm 2 SOC
All



(N = 47)
(N = 50)
(N = 97)















Age (years)
n
47
50
97















Mean (SD)
57.9
(11.4)
58.1
(10.4)
58.0
(10.8)












Median
58.3
57.6
57.8












Mann-Whitney-
0.9971





Wilcoxon p-value













Race
African
0
1
(2.0%)
1
(1.0%)















White
47
(100%)
49
(98.0%)
96
(99.0%)












Other
0
0
0












Fisher p-value
1.0000
















ECOG
0
39
(83.0)
46
(92.0%)
85
(87.6%)


performance


status


ECOG
1
8
(17.0%)
4
(8.0%)
12
(12.4%)


performance


status











Chi-square p-value
0.1775

















TABLE 2







Ovarian Cancer Characteristics in the ITT









ITT Population











Treatment
Treatment




Arm 1
Arm 2
All



(N = 47)
(N = 50)
(N = 97)















Time from
n
47
49
96


diagnosis to


randomization


(years)



n. missing
0
1
1















Median
0.10
(0.01, 0.16)
0.11
(0.04, 0.36)
0.10
(0.01, 0.36)












(Min, Max)















Mann-Whitney-
0.4813





Wilcoxon p-value














Tumor grade
0
0
(0.0%)
0
(0.0%)
0
(0.0%)



1
0
(0.0%)
0
(0.0%)
0
(0.0%)



2
6
(12.8%)
4
(8.0%)
10
(10.3%)



3
40
(85.1%)
44
(88.0%)
84
(86.6%)



4
1
(2.1%)
0
(0.0%)
1
(1.0%)



Unknown
0
(0.0%)
2
(4.0%)
2
(2.1%)












Fisher p-value
0.3234
















Histology
Mucinous
1
(2.13%)
1
(2.0%)
2
(2.1%)



Serous
42
(89.4%)
44
(88.0%)
86
(88.7%)



Undifferentiated
0
(0.0%)
0
(0.0%)
0
(0.0%)



Endometroid
4
(8.5%)
2
(4.0%)
6
(6.2%)



Clear Cell
0
(0.0%)
2
(4.0%)
2
(2.1%)



Other
0
(0.0%)
1
(2.%)
1
(1.0%)












Fisher p-value
0.6041
















Organ of origin
Ovary
42
(89.4%)
43
(86.0%)
85
(87.6%)



Fallopian Tube
3
(6.4%)
2
(4.0%)
5
(5.2%)



Peritoneum
0
(0.0%)
4
(8.0%)
4
(4.1%)



Ovary +
1
(2.1%)
1
(2.0%)
2
(2.1%)



Fallopian Tube



Missing
1
(2.1%)
0
(0.0%)
1
(1.0%)












Fisher p-value
0.2488
















FIGO stage
III
1
(2.1%)
2
(4.0%)
3
(3.1%)



IIIA
5
(10.6%)
3
(6.0%)
8
(8.3%)



IV
5
(10.6%)
3
(6.0%)
8
(8.3%)



IIIB
9
(19.2%)
3
(6.0%)
12
(12.4%)



IIIC
27
(57.5%)
39
(78.0%)
66
(68.0%)



Missing
0
(0.0%)
0
(0.0%)
0
(0.00%)












Fisher p-value
0.1508

















TABLE 3







Summary of adverse events









Safety Population











Treatment
Treatment




Arm 1 CIT
Arm 2 SOC
All



(N = 46)
(N = 49)
(N = 95)

















Patients with any adverse event
38
(82.6%)
40
(81.6%)
78
(82.1%)


Patients with any related adverse event
8
(17.4%)
9
(18.4%)
17
(17.9%)


Patients with any serious adverse event
10
(21.7%)
8
(16.3%)
18
(19.0%)


Patients with any serious related adverse event
0
(0.0%)
0
(0.0%)
0
(0.0%)


Patients with any grade 3-4 adverse event
24
(52.2%)
28
(57.1%)
52
(54.7%)


Patients with any grade 3-4 related adverse event
2
(4.4%)
4
(8.2%)
6
(6.3%)


Patients with any adverse event leading to study
3
(6.5%)
1
(2.0%)
4
(4.2%)


drug discontinuation (*)


Patients with any adverse event leading death
1
(2.2%)
1
(2.0%)
2
(2.1%)





(*) Patients with permanent discontinuation













TABLE 4







Preliminary Clinical Data









All patients











Treatment
Treatment




Arm 1 CIT
Arm 2 SOC
All










Relapse Data
(N = 47)
(N = 50)
(N = 97)














Patients with
Yes
14 (29.8%)
29 (58.0%)
43 (44.3%)


clinical relapse
No
33 (70.2%)
21 (42.0%)
54 (55.7%)



Chi-square
0.0052



p-value
















TABLE 5







Progression free survival (PFS) data maturation









All patients










Treatment Arm 1 CIT
Treatment Arm 2 SOC



(N = 47)
(N = 50)














Censor summary
Total:
47  
49













Censors:
30
(63.8%)
17
(34.7%)



Events:
17
(36.2%)
32
(65.3%)











Quartiles
75th percentile
[—]
33.95
[18.74-]


estimation
50th percentile
[21.30-]
15.39
[10.93-19.33]













25th percentile
19.18
[9.98-28.59]
9.35
[7.12-11.11]


Time to event
Mean (SD)
24.76
(1.38)
18.67
(1.73)


estimation
 0 month
100.00
[100.00-100.00]
100.00
[100.00-100.00]


Survival time
 6 months
97.78
[85.25-99.68]
91.66
[79.28-96.79]



12 months
84.13
[69.57-92.10]
57.55
[42.25-70.17]



18 months
75.04
[59.47-85.33]
42.53
[28.30-56.04]



24 months
65.30
[49.09-77.47]
33.25
[20.17-46.90]



30 months
61.46
[44.43-74.67]
33.25
[20.17-46.90]



36 months
56.34
[37.89-71.23]
16.63
[1.80-44.87]












42 months
56.34
[37.89-71.23]
[—]











48 months
[—]
[—]


Log-rank test
Pr > Chi-Square
0.0009
















TABLE 6







Early Survival Data









All patients












Treatment Arm 1 CIT
Treatment Arm 2 SOC


Overall Survival ITT

(N = 47)
(N = 50)





Censor summary
Total:
46(*)  
49













Censors:
42
(91.3%)
33
(67.3%)



Events:
4
(8.7%)
16
(32.7%)










Quartiles
75th percentile
[—]
[38.34-]











estimation
50th percentile
[—]
38.34
[26.45-]



25th percentile
[30.91-]
21.18
[11.33-38.34]












Time to event
Mean (SD)
29.32
(1.04)
29.89
(1.88)


estimation
 0 month
100.00
[100.00-100.00]
100.00
[100.00-100.00]


Survival time
 6 months
97.73
[84.94-99.68]
95.87
[84.49-98.95]



12 months
93.07
[80.04-97.71]
87.30
[73.88-94.09]



18 months
93.07
[80.04-97.71]
78.39
[63.51-87.76]



24 months
93.07
[80.04-97.71]
73.37
[57.73-83.98]



30 months
93.07
[80.04-97.71]
66.67
[49.62-79.09]



36 months
87.60
[67.52-95.63]
57.14
[33.49-75.11]



42 months
87.60
[67.52-95.63]
42.86
[15.14-68.37]












48 months
87.60
[67.52-95.63]
[—]










Log-rank test
Pr > Chi-Square
0.0025









While preferred embodiments have been described above and illustrated in the accompanying drawings, it will be evident to those skilled in the art that modifications may be made without departing from this disclosure. Such modifications are considered as possible variants comprised in the scope of the disclosure.

Claims
  • 1. A method for therapeutic indirect immunization of an optimally debulked human ovarian cancer patient having a CA125 antigen level in the blood above normal levels, comprising administering to the human patient: (a) a monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region;(b) an immune complex formed between CA125 antigen and the monoclonal antibody having at least a xenogeneic Fc region; or(c) a combination thereof.
  • 2. The method of claim 1, wherein the CA125 antigen level in the blood above normal levels is more than 35 units/mL of CA125 antigen in the blood.
  • 3. The method of claim 1, wherein the CA125 antigen level in the blood above normal levels is at least 50 units/mL of CA125 antigen in the blood.
  • 4. The method of claim 1, wherein the xenogeneic Fc region is from an IgA, IgD, IgG, or IgM isotype.
  • 5. The method of claim 1, wherein the xenogeneic Fc region is from an IgG isotype.
  • 6. The method of claim 5, wherein the IgG isotype is an IgG1 isotype.
  • 7. The method of claim 1, wherein the xenogeneic Fc region is a murine Fc region, a rat Fc region, a rabbit Fc region, a goat Fc region, or a hamster Fc region.
  • 8-10. (canceled)
  • 11. The method of claim 1, wherein the antibody specific to CA125 is mAb-B43.13 (oregovomab).
  • 12. The method of claim 1, wherein administering is one, two, three, or a maximum of four administrations.
  • 13. The method of claim 1, further comprising administration of an immune adjuvant.
  • 14. The method of claim 13, wherein the immune adjuvant is a chemotherapeutic agent, an immunostimulatory compound, an immune homeostatic checkpoint inhibitor, or a combination thereof.
  • 15. The method of claim 14, wherein the chemotherapeutic agent is a platinum-based chemotherapy, taxol, doxorubicin, topotecan, a poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor, or combinations thereof, wherein the immunostimulatory compound is a TLR3 agonist, a TLR4 agonist, or combinations thereof, andwherein the immune homeostatic checkpoint inhibitor is an anti-PD-L1 antibody, an anti-CTLA-4 antibody, and anti-PD-1 antibody, or combinations thereof.
  • 16. The method of claim 15, wherein the platinum-based chemotherapy comprises cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin, satraplatin, and combinations thereof.
  • 17. (canceled)
  • 18. The method of claim 15, wherein the TLR3 agonist is polyIC, polylCLC (Hiltonol®).
  • 19. The method of claim 14, wherein the chemotherapeutic agent is a combination of carboplatin and taxol.
  • 20. The method of claim 14, wherein the therapeutic monoclonal antibody specific for a tumor associated antigen is mAb-B43.13 (oregovomab), and the chemotherapeutic agent is a combination of carboplatin and taxol.
  • 21. (canceled)
  • 22. The method of claim 15, wherein the anti-PD-L1 antibody is selected from the group consisting of B7-H1 antibody, BMS-936559 antibody, MPDL3280A (atezolizumab) antibody, MEDI-4736 antibody, MSB0010718C antibody or combinations thereof, wherein the anti-CTLA-4 antibody is selected from the group consisting of ipilimumab or tremelimumab or combinations thereof,wherein the anti-PD-1 antibody is selected from the group consisting of Nivolumab antibody, pembrolizumab antibody, pidilizumab antibody or combinations thereof, and AMP-224, andwherein the poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor is selected from the group consisting of olaparib, niraparib, rucaparib, talazoparib, veliparib, CEP 9722, E7016, and BGB-290, or combinations thereof.
  • 23-25. (canceled)
  • 26. The method of claim 1, wherein a dose of the monoclonal antibody specific to CA125 antigen having at least a xenogeneic Fc region is from about 0.1 to about 10 mg.
  • 27. The method of claim 1, wherein the method is for the treatment of ovarian cancer.
  • 28. The method of claim 1, wherein the therapeutic indirect immunization is inducing a mannose receptor dependent immune response in the human patient.
  • 29-112. (canceled)
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority of US provisional patent application No. 62/455,114 filed on Feb. 6, 2017, U.S. patent application Ser. No. 15/470,733, filed Mar.27, 2017, and U.S. patent application Ser. No. 15/654,415, filed Jul. 19, 2017 the specifications of which are hereby incorporated by reference in their entireties.

PCT Information
Filing Document Filing Date Country Kind
PCT/CA2017/050901 7/27/2017 WO 00
Related Publications (1)
Number Date Country
20200093925 A1 Mar 2020 US
Provisional Applications (1)
Number Date Country
62455114 Feb 2017 US
Continuations (2)
Number Date Country
Parent 15654415 Jul 2017 US
Child 16482163 US
Parent 15470733 Mar 2017 US
Child 15654415 US