NZ614493B2 - Methods of treating cancer using 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidine-2,6-dione - Google Patents

Methods of treating cancer using 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidine-2,6-dione Download PDF

Info

Publication number
NZ614493B2
NZ614493B2 NZ614493A NZ61449312A NZ614493B2 NZ 614493 B2 NZ614493 B2 NZ 614493B2 NZ 614493 A NZ614493 A NZ 614493A NZ 61449312 A NZ61449312 A NZ 61449312A NZ 614493 B2 NZ614493 B2 NZ 614493B2
Authority
NZ
New Zealand
Prior art keywords
compound
lymphoma
cell
piperidine
formula
Prior art date
Application number
NZ614493A
Other versions
NZ614493A (en
Inventor
Rajesh Chopra
Anita Gandhi
Honwah Man
George W Muller
Peter H Schafer
Linghua Zhang
Hon Wah Man
ling hua Zhang
Original Assignee
Celgene Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Celgene Corporation filed Critical Celgene Corporation
Priority claimed from PCT/US2012/028498 external-priority patent/WO2012125459A1/en
Publication of NZ614493A publication Critical patent/NZ614493A/en
Publication of NZ614493B2 publication Critical patent/NZ614493B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B18/00Surgical instruments, devices or methods for transferring non-mechanical forms of energy to or from the body
    • A61B18/18Surgical instruments, devices or methods for transferring non-mechanical forms of energy to or from the body by applying electromagnetic radiation, e.g. microwaves
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/475Quinolines; Isoquinolines having an indole ring, e.g. yohimbine, reserpine, strychnine, vinblastine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/18Antioxidants, e.g. antiradicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer

Abstract

Provided are methods of treating cancers comprising the administration of 3-(5-amino-2-methyl-4-oxo-4H-quinazolin-3- yl)-piperidine-2,6-dione, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof. In a preferred embodiment the compound is used to treat diffuse large B-cell lymphoma of the activated B-cell phenotype. n a preferred embodiment the compound is used to treat diffuse large B-cell lymphoma of the activated B-cell phenotype.

Description

METHODS OF TREATING CANCER USING MINOMETHYLOXO— 4H-QUINAZOLINYL)-PIPERIDINE-2,6-DIONE This ation claims the benefit of US. Provisional Patent Application No. 61/451,995, filed on March 11, 2011, and US. Provisional Patent Application No. 61/480,272, filed on April 28, 2011, the entireties of which are incorporated herein by reference. 1. FIELD OF THE INVENTION Provided herein are methods of treating, preventing and/or managing s, which to a t 3-(5amino-2‘-methyl—4~oxo—4H—quinazolin-3~yl)- _ comprise administering piperidine-2,6-dione, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof. 2. BACKGROUND OF THE INVENTION 2.1 IOLOGY OF CANCER Cancer is characterized primarily by an increase in the number of abnormal cells derived from a given normal tissue, invasion of adjacent s by these abnormal cells, or lymphatic or blood—borne spread of ant cells to al lymph nodes and to t sites (metastasis). Clinical data and molecular biologic studies indicate that cancer is a multistep process that begins with minor preneoplastic changes, which may under certain conditions progress to neoplasia. The neoplastic lesion may evolve clonally and develop an increasing capacity for invasion, growth, metastasis, and heterogeneity, especially under conditions in which the stic cells escape the host’s immune surveillance. Roitt, 1., Brostoff, J and Kale, D., Immzmologv, 17.1-17.12 (3rd ed., Mosby, St. Louis, Mo, 1993).
There is an enormous variety of cancers which are described in detail in the medical literature. Examples include cancer of the lung, colon, rectum, prostate, breast, brain, and intestine. The incidence of cancer continues to climb as the general population ages, as new cancers develop, and as tible populations (cg, people infected with AIDS or excessively exposed to ht) grow. A tremendous demand therefore exists for new methods and compositions that can be used to treat patients with cancer.
Many types of cancers are associated with new blood vessel formation, a process known as angiogenesis. Several of the mechanisms invoived in tumor-induced angiogenesis have been ated. The most direct of these mechanisms is the secretion by the tumor cells of cytokines with angiogenic properties. Examples of these cytokines include acidic and basic fibroblastic growth factor (a,b-FGF), angiogenin, vascular endothelial growth factor (VEGF), and TNF~a. Alternatively, tumor cells can release angiogenic peptides through the production of proteases and the subsequent breakdown the extracellular matrix Where some cytokines are stored (cg, b-FGF). Angiogenesis can also be d indirectly through the recruitment of inflammatory cells (particularly hages) and their subsequent release of enic cytokines (e. g. , TNF-u, b-FGF). ma refers to cancers that originate in the lymphatic system. Lymphoma is characterized by malignant neoplasms of lymphocytes—B lymphocytes and T cytes collections of (12a, B»cells and T-cells). Lymphoma generally starts in lymph nodes or tic tissue in organs including, but not limited to, the stomach or intestines.
Lymphoma may involve the marrow and the blood in some cases. Lymphoma may spread from one site to other parts of the body.
The treatment of various forms of lymphomas are described, for example, in US. herein by reference. Such patent no. 7,468,363, the entirety of which is incorporated lymphomas include, but are not d to, Hodgkin’s lymphoma, non—Hodgkin's B—cell lymphoma, cutaneous B-cell lymphoma, activated B-cell ma, diffuse large lymphoma (DLBCL), mantle cell lymphoma (MCL), follicular center lymphoma, transformed lymphoma, lymphocytic lymphoma of intermediate differentiation, intermediate lymphocytic lymphoma (ILL), diffuse poorly differentiated lymphocytic lymphoma (PDL), centrocytic lymphoma, diffuse small-cleaved cell ma (DSCCL), peripheral T-cell lymphomas , cutaneous T-Cell lymphoma and mantle zone lymphoma and low grade follicular ma.
Non—Hodgkin‘s lymphoma (NHL) is the fifth most common cancer for both men and and 18,660 deaths in 2007. women in the United , with an estimated 63,190 new cases Jemal A, et of, CA Cancer J Clin 2007; 57(1):43—66. The probability of developing NHL increases with age and the incidence ofNHL in the elderly has been steadily increasing the past decade, causing concern with the aging trend of the US population. Id. Clarke C A, et al., Cancer 2002; 94(7):;20l 5—2023.
Diffuse large B—cell lymphoma (DLBCL) accounts for approximately one-third of non-Hodgkin’s lymphomas. While some DLBCL patients are cured with traditional chemotherapy, the remainder die from the disease. Anticancer drugs cause rapid and persistent depletion of lymphocytes, ly by direct apoptosis ion in mature T and B cells. See K. Stahnke. a! a!., Head 2001, 98:3066-3073. Absolute lymphocyte count WO 25459 (ALC) has been shown to be a prognostic factor in follicular non—Hodgkin’s lymphoma and recent results have ted that ALC at diagnosis is an ant prognostic factor in diffuse large B-cell lymphoma. See D. Kim et al., Journal ofClinical Oncology, 2007 ASCO Annual Meeting Proceedings Part I. Vol 25, No. 188 (June 20 Supplement), 2007: U: 8082.
Leukemia refers to malignant neoplasms of the blood—forming tissues. Various forms of leukemias are described, for example, in US. patent no. 7,393,862 and US. provisional patent application no. 60/3 80,842, filed May 17, 2002, the entireties of which are orated herein by reference. Although Viruses reportedly cause several forms of leukemia in animals, causes of leukemia in humans are to a large extent unknown. The Merck Manual, 944—952 (17th ed. 1999). Transformation to malignancy typically occurs in a single cell through two or more steps with subsequent proliferation and clonal expansion.
In some leukemias, specific chromosomal ocations have been identified with consistent leukemic cell morphology and special clinical features (e.g, translocations of 9 and 22 in chronic myelocytic leukemia, and of 15 and 17 in acute locytic leukemia).
Acute leukemias are predominantly undifferentiated cell populations and chronic leukemias more mature cell forms.
Acute leukemias are divided into lymphoblastic (ALL) and non-lymphoblastic (ANLL) types. The Merck Manual, 946—949 (l 7th ed. 1999). They may be further subdivided by their morphologic and cytochemical ance according to the French- an—British (FAB) classification or according to their type and degree of differentiation. The use of specific B- and T-cell and myeloid-antigen monoclonal antibodies are most helpful for classification. ALL is predominantly a childhood disease which is established by laboratory findings and bone marrow examination. ANLL, also known as acute myelogenous leukemia or acute lastic leukemia (AML), occurs at all ages and is the more common acute leukemia among ; it is the form usually associated with irradiation as a causative agent.
Chronic leukemias are described as being lymphocytic (CLL) or myelocytic (CML).
The Merck Manual, 949-952 (17"h ed. 1999). CLL is characterized by the appearance of mature cytes in blood, bone marrow, and lymphoid . The hallmark of CLL is sustained, absolute lymphocytosis (> 5,000/uL) and an increase of lymphocytes in the bone marrow. Most CLL patients also have clonal expansion of lymphocytes with B~cell characteristics. CLL is a disease of middle or old age. In CML, the characteristic e is the predominance of granulocytic cells of all stages of differentiation in blood, bone marrow, liver, spleen, and other organs. In the symptomatic patient at diagnosis, the total White blood cell (WBC) count is usually about 200,000/uL, but may reach 000/uL.
CML is relatively easy to diagnose because of the presence of the Philadelphia chromosome.
L1: In addition to the acute and chronic categorization, neoplasms are also rized based upon the cells giving rise to such disorder into precursor or peripheral. See ag, US. patent publication no. 2008/0051379, the disclosure of which is incorporated herein by reference in its entirety. Precursor neoplasms include ALLs and lymphoblastic lymphomas and occur in lymphocytes before they have entiated into either a T- or .
Peripheral neoplasms are those that occur in lymphocytes that have differentiated into either T— or s. Such peripheral neoplasms include, but are not limited to, B-cell CLL, B-cell prolymphocytic leukemia, plasmacytic lymphoma, mantle cell lymphoma, follicular lymphoma, extranodal marginal zone B—cell lymphoma of mucosa-associated lymphoid , nodal marginal zone lymphoma, c marginal zone lymphoma, hairy cell leukemia, plasmacytoma, diffuse large B-cell lymphoma and Burkitt lymphoma. In over 95 percent of CLL cases, the clonal expansion is of a B cell e. See Cancer: Principles & Practice of Oncology (3rd Edition) (1989) (pp. 1843-1847). In less than 5 percent of CLL cases, the tumor cells have a T-cell phenotype. hstanding these classifications, however, the pathological impairment of normal hematopoiesis is the hallmark of all 2O leukemias.
Multiple myeloma (MM) is a cancer of plasma cells in the bone . Normally, plasma cells produce antibodies and play a key role in immune function. However, uncontrolled growth of these cells leads to bone pain and fractures, anemia, infections, and other complications. Multiple myeloma is the second most common hematological malignancy, although the exact causes of multiple myeloma remain unknown. Multiple myeloma causes high levels of ns in the blood, urine, and organs, including but not limited to M-protein and other immunoglobulins (antibodies), albumin, and beta microglobulin. M-protein, short for monoclonal protein, also known as paraprotein, is a particularly abnormal protein produced by the myeloma plasma cells and can be found in the blood or urine of almost all patients with multiple myeloma.
Skeletal symptoms, including bone pain, are among the most clinically significant symptoms of multiple myeloma. Malignant plasma cells release osteoclast ating factors (including ll; 1 , IL—6 and TNF) which cause calcium to be leached from bones causing lytic lesions; hypercalcemia is r m. The osteoclast stimulating WO 25459 factors, also referred to as cytokines, may t apoptosis, or death of myeloma cells.
Fifty percent of patients have radiologically detectable myeloma-related skeletal lesions at diagnosis. Other common clinical symptoms for multiple myeloma include polyneuropathy, , hyperviscosity, infections, and renal insufficiency.
Solid tumors are abnormal masses of tissue that may, but y do not contain cysts or liquid areas. Solid tumors may be benign (not cancer), or ant (cancer).
Different types of solid tumors are named for the type of cells that form them. Examples of types solid tumors include, but are not limited to malignant melanoma, adrenal carcinoma, breast carcinoma, renal cell cancer, carcinoma of the pancreas, non-small—cell lung carcinoma (NSCLC) and carcinoma ofunknown primary. Drugs commonly administered to patients with various types or stages of solid tumors e, but are not limited to, celebrex, ide, cyclophosphamide, docetaxel, abine, IFN, tamoxifen, IL~2, GM— CSF, or a combination thereof.
While ts who achieve a complete remission after initial therapy have a good chance for cure, less than 10% of those who do not respond or relapse achieve a cure or a response lasting longer than 3 years. See Cemy T, et al., Ann Oncol 2002; 13 Suppl 4:211~ 216.
Rituximab is known to deplete normal host B cells. See M. Aklilu et al., Annals of Oncology 15:1109—1114, 2004. The long—term logic effects of B cell depletion with rituximab and the characteristics of the tituting B cell pool in lymphoma patients are not well defined, despite the widespread usage of this y. See Jennifer H. Anolik at (11., Clinical Imnmnologx, vol. 122, issue 2, February 2007, pages 139—145.
The approach for patients with relapsed or refractory disease relies heavily on experimental treatments followed by stem cell transplantation, which may not be appropriate for patients with a poor performance status or ed age. Therefore, a tremendous demand exists for new methods that can be used to treat patients with NHL.
The link n cancer an altered celiular metabolism has been well established.
See Cairns, R.A., er of. Nature Rey-t, 2011, 1128395. Understanding tumor cell metabolism and the associated genetic changes thereof may lead to the identification of improved methods of cancer treatment. Id. For example, tumor cell survival and eration Via increased glucose metabolism has been linked to the PIK3 pathway, whereby mutations in tumor suppressor genes such as PTEN activate tumor cell metabolism. Id. AKTI (aka, PKB) stimulates glucose metabolism associtated with tumor cell growth by various interactions with PFKFB3, ENTPDS, mTOR and TSC2 (aka. , tuberin). 1d. 2012/028498 ription s HIFl and HIFZ are y responsible for cellular response to low oxygen conditions often associated with tumors. Id. Once activated, HlFl promotes tumor cell capacity to carry out glycolysis. Id Thus, inhibition of HIP 1 may slow or e tumor cell metabolism. tion of HIFl has been linked to P13K, tumor suppressor proteins such as VHL, succinate dehydrogenase (SDH) and fumarate hydratase.
Id. The oncogenic transcription factor MYC has also been linked to tumor cell metabolism, specifically glycolysis. Id. MYC also promotes cell proliferation by glutamine metabolic pathways. Id.
AMP-activated protein kinase (AMPK) functions as a metabolic check point which tumor cells must overcome in order to proliferate. Id. Several mutations have been ifed which suppress AMPK signaling in tumor cells. See Shackelford, D.B. & Shaw, R.J., Nature Rev. Cancer, 2009, 9: 563-575. STKll has been identified as a tumor suppressor gene related to the role of AMPK. See Cairns, R.A., er al. Nature Reta, 2011, 11:85-95.
The transcription factor p53, a tumor suppressor, also has an important role in the regulation of cellular metabolism. Id. The loss of p53 in tumor cells may be a significant contributor to s in tumor cell metabolism to the glycolytic pathway. Id. The OCTl transcription factor, another ial target for chemotherapeutics, may cooperate with p53 in regulating tumor cell metabolism. Id.
Pyruvate kinate M2 (PKMZ) promotes changes in cellular metabolism which confer metabolic advantages to cancer cells by supporting cell proliferation. Id. For example, lung cancer cells which s PKM2 over PKMl have been found to have such an advantage.
Id. In the clinic, PKM2 has been identified as being pressed in a number of cancer types. Id. Thus PKM2 may be a useful biomarker for the early detection of tumors.
Mutations in isocitrate dehydrogenases IDHl and IDH2 have been linked to tumorigenesis, specifically, in glioblastoma and acute myeloid leukemia. See Mardis, ER. at 5d,, N. EngI. J. Med, 2009, 36]: 10584066; Parsons, D.W. at 621., Science, 2008, 321: 1807-1812.
The incidence of cancer continues to climb as the general population ages, as new cancers develop, and as susceptible populations (ag, people infected with AIDS, the elderly or excessively exposed to sunlight) grow. A tremendous demand therefore exists for new methods, treatments and compositions that can be used to treat patients with cancer including but not limited to those with lymphoma, NHL, multiple myeioma, AML, leukemias, and solid tumors.
Accordingly, nds that can control and/or inhibit unwanted angiogenesis or inhibit the production of certain cytokines, including TNF—a, may be useful in the treatment and prevention of various forms of cancer. 2.2 METHODS OF TREATING CANCER Current cancer therapy may involve surgery, chemotherapy, hormonal therapy and/or radiation ent to eradicate neoplastic cells in a patient (see, for e, ale, 1998, Medicine, vol. 3, Rubenstein and Federman, eds, Chapter 12, n IV).
Recently, cancer therapy could also involve biological therapy or immunotherapy. All of these approaches may pose significant drawbacks for the patient. Surgery, for example, the patient. may be contraindicated due to the health of a patient or may be unacceptable to Additionally, surgery may not completely remove neoplastic tissue. Radiation therapy is only effective when the neoplastic tissue exhibits a higher sensitivity to radiation than normal . Radiation therapy can also often elicit serious side effects. Hormonal therapy is rarely given as a single agent. Although hormonal therapy can be ive, it is often used to prevent or delay recurrence of cancer after other ents have removed the majority of cancer cells. Certain biological and other therapies are limited in number and may produce side effects such as rashes or swellings, flu-like symptoms, including fever, chills and fatigue, ive tract problems or allergic reactions.
With respect to herapy, there are a variety of herapeutic agents 2O available for treatment of cancer. A number of cancer chemotherapeuties act by inhibiting DNA synthesis, either directly or indirectly by inhibiting the biosynthesis of ibonucleotide triphosphate precursors, to prevent DNA ation and concomitant cell division. Gilman at al. , Goodman and Gilman ’s: The Pharmacological Basis of Therapeutics, Tenth Ed. (McGraW Hill, New York).
Despite availability of a variety of chemotherapeutic agents, chemotherapy has many drawbacks. Stockdale, Medicine, vol. 3, Rubenstein and Federman, eds., ch. 12, sect. , 1998. Almost all chemotherapeutic agents are toxic, and chemotherapy causes significant and often dangerous side effects ing severe nausea, bone marrow depression, and immunosuppression. Additionally, even with administration of combinations of chemotherapeutic agents, many tumor cells are resistant or develop resistance to the chemotherapeutic agents. in fact, those cells ant to the ular chemotherapeutic agents used in the treatment protocoi often prove to be resistant to other drugs, even if those agents act by different mechanism from those of the drugs used in the WO 25459 specific treatment. This phenomenon is referred to as multidrug resistance. Because of the drug resistance, many cancers prove refractory to standard chemotherapeutic treatment protocols.
There exists a significant need for safe and effective methods of treating, preventing U: and managing cancer, particularly for cancers that are refractory to rd ents, such as surgery, radiation therapy, chemotherapy and hormonal therapy, while reducing or avoiding the toxicities and/or side s associated with the conventional therapies. 2.3 ON The protein Cereblon (CRBN) is a 442-amino acid protein conserved from plant to human. In humans, the CRBN gene has been fied as a candidate gene of an autosomal recessive nonsyndromic mental retardation R). See s, J.J. er a1, Neurology, 2004, 63:1927-1931. CRBN was initially characterized as an RGS-containing novel protein that interacted with a m-activated potassium channel protein (SLOl) in the rat brain, and was later shown to interact with a voltage-gated chloride l (C102) in the retina with AMPK7 and DDBl. See Jo, S. et (11., J. Neurochem, 2005, 94:1212-1224; Hohberger B. et al., FEBS Lett, 2009, 583:633—637; Angers S. at (11., Nature, 2006, 443:590—593.
DDBl was originally identified as a nucleotide excision repair protein that associates with damaged DNA binding protein 2 (DDB2). Its defective activity causes the repair defect in the patients with xeroderma pigmentosum complementation group B (XPE). DDB] also appears to function as a component of numerous ct DCX (DDBl—CUL4-X~box) E3 ubiquitin-protein ligase complexes which mediate the ubiquitination and subsequent proteasomal degradation of target proteins. CRBN has also been identified as a target for the development of therapeutic agents for diseases of the cerebral cortex. See WO 2010/137547 A1. is.) U) Cereblon has recentiy been identified as a key molecular target that binds to thalidomide to cause birth defects. See Ito, T. et al, Science, 2010, 327:1345-1350. DDBl was found to interact with CRBN and, thus, was indirectly ated with thalidomide.
Moreover, thalidomide was able to inhibit auto—ubiquitination of CRBN in vitro, suggesting that omide is an E3 ubiquitin-iigase inhibitor. Importantly, this activity was inhibited by thalidomide in wild-type cells, but not in cells with mutated CRBN binding sites that prevent thalidomide binding. The thalidomide binding site was mapped to a highly conserved C—terminal 104 amino acid region in CRBN. Individual point mutants in CRBN, Y384A and W386A were both defective for thalidomide binding, with the double point mutant having the lowest thaiidomide—binding activity. A link between CRBN and the - g _ teratogcnic effect of thalidomide was confirmed in animal models of zebra-fish and chick embryos. Understanding omide and other drug targets will allow the definition of the molecular mechanisms ofetlicacy and/or ty and may lead to drugs with improved efficacy and toxicity profiles. 3. SUMMARY OF THE ION Provided herein are methods oftreating and preventing cancer. including primary and metastatic cancer, as well as cancer that is refractory or resistant to conventional chemotherapy. which comprise administering to a patient in need of such treatment or prevention a therapeutically or prophylactically effective amount of 3-(5-amino-Z-methyl oxo—4H—quinazolinyl)—piperidine-2,6-dione. having the structure of Formula I: or an enantiomer or a mixture ot‘enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, elathratc, or polymorph thereof as a single agent or as a part ofa combination therapy.
In a ular embodiment, provided herein is the use of a therapeutically efi'ective amount of 3-(5-aminomethyl—4~oxo—4H-quinazolin—3-yl)-piperidine-2,6-dione, which has the following structure: N\\|/ NH2 0 II 0 N O or an enantiomer or e of omers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof in the cture of a medicament for treating or managing dgkin’s lymphoma, wherein the non-Hodgkin’s lymphoma is e large B-cell lymphoma of the activated B-cell phenotype. 9 (followed by page 9A) 9A wed by page 10) In a particular embodiment, provided herein is the use of a therapeutically effective amount of 3-(5-aminomethyloxo-4H-quinazolinyl)-piperidine-2,6-dione or a pharmaceutically acceptable salt, solvate or hydrate thereof in the preparation of a medicament for treating or managing non-Hodgkin’s lymphoma, wherein the patient is identified to have dgkin’s ma sensitive to treatment with 3-(5-aminomethyl- 4-oxo-4H-quinazolinyl)-piperidine-2,6-dione and the non-Hodgkin’s lymphoma is diffuse large B-cell lymphoma of the ted B-cell phenotype.
In some embodiments, provided herein are methods for the use of gene and protein biomarkers as a tor of clinical sensitivity to lymphoma, non~Hodgkin’s lymphoma, multiple myeloma, leukemia, AML, and/or solid tumors and patient response to ent with 3~(5-aminomethyl—4~oxo-4H-quinazolinyl)—piperidine-2,6-dione, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof.
The methods provided herein encompass methods for screening or identifying cancer patients, cg, lymphoma, non-Hodgkinis lymphoma, multiple myeloma, ia, AML, and solid tumor patients, for treatment with 3—(5—aminomethyl~4-oxo—4H— quinazolin~3—yl)—piperidine-2,6-dione, or an omer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, e, co-crystal, clathrate, or polymorph thereof. In particular, provided herein are methods for selecting patients having a higher response rate to therapy with 3-(5-aminomethyl—4—oxo-4H—quinazolin-3—yl)-piperidine~ 2,6-dione.
In one ment, provided herein is a method of ting tumor response to treatment in a lymphoma, non-Hodgkin’s lymphoma, multiple myeloma, leukemia, AML or solid tumor patient, the method comprising obtaining tumor tissue from the t, purifying protein or RNA from the tumor, and measuring the presence or absence of a biomarker by ,e.g., protein or gene sion analysis. The expression monitored may be, for example, mRNA expression or n expression.
In certain embodiments, the biomarker is a gene associated with an activated B-cell phenotype of DLBCL. The genes are selected from the group ting of IRF4/MUM1, FOXPl, SPIB, CARDll and PDRMl. In one embodiment, the biomarker is NF- In one embodiment, the mRNA or protein is purified from the tumor and the presence or absence of a biomarker is measured by gene or protein expression analysis. In certain embodiments, the presence or absence of a ker is measured by quantitative real—time PCR (QRT—PCR), microarray, flow cytometry or immunofluorescence. In other embodiments, the ce or absence of a biomarker is measured by enzyme~linl<ed immunosorbent assay-based methodologies (ELISA) or other similar methods known in the art.
In another embodiment, provided herein is a method of ting tumor response to treatment in a non-Hodgkin’s lymphoma patient, the method comprising obtaining tumor cells from the patient, culturing the cells in the presence or e of 3-(5-aminomethyl- 4~oxo~4H—quinazolin—3—yl)«piperidine-2,6-dione, purifying n or RNA from the cultured cells, and measuring the presence or absence of a ker by ,e.g., protein or gene expression analysis. The expression monitored may be, for example, mRNA expression or n expression.
In another embodiment, provided herein is a method of monitoring tumor response to 3-(5—amino—2—methyl—4—oxo-4H~quinazolin~3~yl)-piperidine—2,6~dione treatment in a lymphoma, non-Hodgkin’s lymphoma, multiple myeloma, leukemia, AML or solid tumor patient. The method comprises obtaining a biological sample from the patient, measuring the expression of a biomarker in the ical sample, administering minomethyl- 4-oxo—4H-quinazolin-3~yl)-piperidine-2,6~dione to the patient, thereafter obtaining a second biological sample from the patient, measuring biomarker expression in the second biological sample, and comparing the levels of sion, where an increased level of biomarker expression after treatment indicates the likelihood of an effective tumor response.
In one embodiment, a decreased level of ker expression after treatment indicates the likelihood of effective tumor response. The ker expression monitored can be, for example, mRNA expression or protein expression. The expression in the treated sample can increase, for example, by about 1.5X, 2.0X, 3X, 5X, or more.
In yet r embodiment, a method for monitoring patient compliance with a drug ent protocol is provided. The method comprises obtaining a biological sample from the patient, measuring the expression level of at least one biomarker in the sample, and determining if the expression level is increased or decreased in the patient sample compared to the expression level in a control untreated sample, wherein an increased or decreased expression indicates t compliance with the drug treatment protocol. In one ment, the expression of one or more biomarkers is increased. The biomarker expression monitored can be, for example, mRNA expression or protein expression. The expression in the treated sample can increase, for example, by about 1.5X, 2.0K, 3X, 5X, or more.
In another embodiment, provided herein is a method of predicting the sensitivity to treatment 3—(5~arninn-2~methyl-4~oxo~4H~quinazolinyl)~piperidine-2,6-dione in a lymphoma, non—Hodgkin’s ma, multiple myeloma, leukemia, AML or solid tumor t. In one embodiment, the patient is a nonnHodgkin’s lymphoma patient, specifically, a DLBCL patient, The method comprises obtaining a biological sample from the t, optionally ing or purifying mRNA from the biologicai sample, amplifying the mRNA transcripts by, e.g., RT-PCR, Where a higher ne level of a specific biomarker indicates a higher hood that the cancer will be sensitive to treatment with 3—(5~aminomethyl- 4—oxo—4H—quinazolinyl)—piperidine—2,6—dione. In certain embodiments, the biomarker is a gene associated with an activated B~cell phenotype. The genes are ed from the BLIMP/PDRMI. group consisting of IRF4/MUM1, FOXPI, SPIB, CARDll and Also provided herein are methods for the treatment or management of cancer with 3- (S-amino—Z-methyl—4~oxo—4H~quinazolin—3-yl)-piperidineo2,6—dione using CRBN as a predictive or prognostic factor. In certain embodiments, provided herein are methods for screening or identifying cancer patients for treatment with 3«(5-amino-2~methyl-4~oxo—4H— quinazolinyl)—piperidine-2,6~dione using CRBN levels as a predictive or prognostic IO factor. In some embodiments, provided herein are s for selecting patients having a higher se rate to therapy with 3~(S-amino—2~methyloxo—4H—quinazolin-3~yl)- piperidine—2,6-dione using CRBN levels as a predictive or prognostic factor.
In one embodiment, provided herein is a method of predicting patient response to treatment of cancer with 3~(5—amino-2—methyl—4—oxo-4H-quinazolin-3 —yl)—piperidine~2,6- dione, the method comprising obtaining ical material from the patient, and measuring the presence or absence of CRBN.
In one embodiment, the method comprises ing cancer cells from the patient, culturing the cells in the presence or absence of 3—(5-aminomethyl—4—oxo~4H-quinazolin— 3~yl)-piperidine-2,6~dione, purifying protein or RNA from the cultured cells, and measuring the presence or absence of a biomarker by ,eg, n or gene expression analysis. The expression monitored may be, for example, mRNA expression or n expression. In one embodiment, the cancer is lymphoma, leukemia, multiple myelorna, solid tumor, non- Hodgkin’s lymphoma or melanoma.
In another embodiment, provided herein is a method of monitoring tumor response to drug treatment in a cancer t. The method comprises obtaining a biological sample from the patient, measuring the expression of a biomarker in the biological sample, administering one or more drugs to the patient, fter obtaining a second ical sample from the patient, measuring biomarker expression in the second biologicai sample, and comparing the levels of sion, where an sed level of biomarker expression after treatment indicates the likelihood of an effective tumor response. In one embodiment, the cancer patient is a lymphoma, leukemia, multiple myeloma, solid tumor, non-Hodgkin’s ma or melanoma patient.
In one embodiment, a decreased level of biomarker expression after treatment indicates the likelihood of effective tumor response, The biomarker expression monitored -13.. can be, for example, mRNA expression or protein expression. The expression in the treated sample can increase, for example, by about 1.5X, 2.0x, 3X, 5X, or more. In one embodiment, the tumor is a lymphoma, leukemia, multiple myeloma, solid tumor, non- Hodgkin’s ma or melanoma.
In another embodiment, provided herein is a method of predicting the sensitivity to drug treatment in a cancer patient, specifically, a multiple myeloma or non-Hodgkin’s lymphoma patient. The method comprises obtaining a biological sample from the patient, ally isolating or purifying mRNA from the biological sample, amplifying the mRNA transcripts by, eg, RT—PCR, where a higher baseline level of a specific biomarker tes a higher likelihood that the cancer will be ive to treatment with a drug. In certain embodiments, the biomarker is a gene or protein associated with le myeloma or non- n’s lymphoma. In one embodiment, the genes are those associated with CRBN and are selected from the group ting of DDBl, DDB2, GSK3B, CUL4A, CUL4B, XBP-l, FASl, RANBP6, DU83 L, PHGDH, AMPK, IRF4 and NFKB. In another embodiment, the genes are ed from the group consisting , DDBZ, IRF4 and NFKB.
In one embodiment, identifying a patient having lymphoma, leukemia, multiple a, a solid tumor, non-Hodgkin’s ma, diffuse large B—cell lymphoma or melanoma sensitive to ent with 3-(5—aminomethyloxo-4H—quinazolin—3 —yl)— piperidine-2,6-dione; identification of a gene or protein associated with CRBN wherein the ce of the gene or protein associated with CRBN is indicative of lymphoma, leukemia, multiple myeloma, a solid tumor, non-Hodgkin’s lymphoma, diffuse large B~cell lymphoma or melanoma sensitive to treatment with 3-(5-aminomethyloxo~4H-quinazolin-3 -yl)- piperidine-2,6-dione. In one ment, the gene or protein associated with CRBN is selected from the group consisting of DDBl, DDBZ, IRF4 and NFKB.
In one embodiment, identifying a patient having lymphoma, leukemia, multiple myeloma, a solid tumor, non-Hodgkin’s lymphoma or melanoma sensitive to treatment with 3—(5—amino-2~methyloxo—4H-quinazolin-3—yl)-piperidine-2,6-dione comprises measuring the level of CRBN activity in the patient. In another embodiment, measuring the level of CRBN activity in the patient comprises measuring DDBI, DDB2, IRF4 and/or NFKB in cells obtained from the patient.
In one embodiment, provided herein is a method for treating or managing non- Hodgkin’s lymphoma, comprising: (i) identifying a patient having lymphoma, min-Hodgkin’s lymphoma, multiple myeloma, leukemia, AML or a solid tumor sensitive to treatment with 3-(5-amino—2- ‘4-oX0-4H—quinazolin-3—yl)—piperidine—2,6-dione; and (ii) administering to the patient a therapeutically effective amount of 3-(5-amino-2— methyl—4—0xo«4H—quinazoliiiyl)—piperidine—2,6-dione, or a pharmaceutically acceptable salt or solvate (e. g, hydrate) thereof.
In one embodiment, the patient has non—Hodgkin’s lymphoma. In one embodiment, the non-Hodgkin’s lymphoma is e large B-cell lymphoma. In another embodiment, the non—Hodgkin’s lymphoma is of the ted B-cell phenotype.
In one embodiment, identifying a patient having non-Hodgkin’s lymphoma sensitive to ent with 3—(S-amino—2-methyl—4-oxo-4Houinazolin-3~yl)-piperidine-2,6-dione ses identification of a gene associated With the activated B-cell phenotype. In one embodiment, the gene associated with the ted B-cell ype is selected from the BLIMP/PDRMI. group consisting of IRF4/MUM1, FOXPl, SPIB, CARDll and In one embodiment, identifying a patient having non-Hodgkin’s lymphoma sensitive to ent with 3-(5-amino-2—methyl~4-oxo-4H-quinazolinyl)—piperidine—2,6—dione comprises measuring the level ofNF-KB activity in the patient. In another embodiment, measuring the level ofNF—KB activity in the patient ses measuring the baseline NF— KB activity level in tumor cells obtained from the patient.
Also provided herein are kits useful for predicting the likelihood of an ive lymphoma, non-Hodgkin’s lymphoma, multiple myeloma, leukemia, AML or solid tumor ent or for monitoring the effectiveness of a treatment with 3—(5-aminomethyl-4— oxo—4H~quinazolin~3—yl)~piperidine~2,6—di0ne. The kit comprises a solid support, and a means for detecting the protein expression of at least one biomarker in a biological sample.
Such a kit may employ, for example, a dipstick, a membrane, a chip, a disk, a test strip, a filter, a microsphere, a slide, a multiweli plate, or an optical fiber. The solid support of the kit can be, for example, a plastic, silicon, a metal, a resin, glass, a membrane, a particle, a precipitate, a gel, a polymer, a sheet, a sphere, a poiysaccharide, a capillary, a film, a plate, or a slide. The biological sample can be, for example, a cell culture, a cell line, a tissue, an oral tissue, gastrointestinal , an organ, an organelle, a biological fluid, a blood sample, a urine sample, or a skin sample. The biological sample can be, for example, a lymph node biopsy, a bone marrow biopsy, or a sample of eral blood tumor cells.
In an additional embodiment, provided herein is a kit useful for predicting the hood of an effective treatment or for monitoring the effectiveness of a treatment with 3-(5—aminomethyl-4—oxo-4H-quinazolinyl)—piperidine-2,6~dione. The kit comprises a solid support, nucleic acids contacting the support, Where the nucleic acids are complementary to at least 20, 50, 100, 200, 350, or more bases of mRNA, and a means for detecting the expression of the mRNA in a biological sample.
In another embodiment, provided herein is a kit useful for predicting the likelihood of an effective treatment or for monitoring the effectiveness of a treatment with 3-(5-amino- 2-methyl-4—oxo-4H-quinazolin-3~yl)-piperidine-2,6—dione. The kit ses a solid support, at least one nucleic acid contacting the support, where the nucleic acid is complementary to at least 20, 50, 100, 200, 350, 500, or more bases of mRNA, and a means for detecting the expression of the mRNA in a biological sample.
In certain ments, the kits provided herein employ means for detecting the expression of a biomarker by quantitative ime PCR (QRT—PCR), microarray, flow cytometry or immunofluorescence. In other embodiments, the expression of the biomarker is measured by based methodologies or other similar methods known in the art.
Also ed herein are pharmaceutical itions comprising about 1 to 1,000 mg of 3—(S-amino~2-methyl—4-oxo-4H-quinazolin-3—yl)-piperidine-2,6-dione, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, e, co—crystal, clathrate, or polymorph thereof.
Further provided herein are pharmaceutical compositions sing about 1 to 1,000 mg of 3—(5—aminomethyl*4-oxo—4H-quinazolin—3-yl)-piperidine-2,6-dione, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutical ly acceptable salt, solvate, e, co-crystal, clathrate, or polymorph thereof; and one or more additional active ingredient. In certain embodiments, the one or more additional active ingredients are selected from oblimersen, melphalan, G—CSF, GM-CSF, EPO, a cox-2 tor, topotecan, pentoxifylline, ciprofloxacin, re, iritotecan, dexamethasone, doxorubicin, Vincristine, IL 2, lFN, dacarbazine, Ara—C, lbine and isotretinoin.
Also provided herein are kits useful for predicting the likelihood of an ive lymphoma, leukemia, muitiple myeloma, a solid tumor, non-Hodgkin’s lymphoma, diffuse large B—celi lymphoma or melanoma ent or for monitoring the effectiveness of a treatment with one or more drugs, eg, 3{5~amino—Z-methyl~4—oxo-4H-quinazoiiiiyl} piperidine-2,6-dione. The kit comprises a solid support, and a means for detecting the protein expression of at least one biomarker in a biological sample. Such a kit may employ, for example, a ck, a membrane, a chip, a disk, a test strip, a filter, a microsphere, a slide, a multiwell plate, or an optical fiber. The solid support of the kit can be, for example, a plastic, silicon, a metal, a resin, glass, a membrane, a particle, a precipitate, a gel, a polymer, a sheet, a sphere, a polysaccharide, a capillary, a film, a plate, or a slide. The biological sample can be, for example, a cell culture, a cell line, a tissue, an oral tissue, gastrointestinal tissue, an organ, an organelle, a biological fluid, a blood sample, a urine sample, or a skin sample. The biological sample can be, for e, a lymph node biopsy, a bone marrow biopsy, or a sample of peripheral blood tumor cells.
In another embodiment, the kit comprises a solid support, nucleic acids contacting the t, Where the nucleic acids are complementary to at least 20, 50, 100, 200, 350, or more bases of mRNA, and a means for detecting the expression of the mRNA in a biological sample.
In certain embodiments, the kits provided herein employ means for detecting the expression of a biomarker by quantitative real—time PCR (QRT-PCR), microarray, flow try or immunofluorescence. In other embodiments, the sion of the ker is measured by ELISA—based methodologies or other similar methods known in the art.
Also provided herein is a kit comprising (i) a pharmaceutical composition comprising 3-(5-amino—2—methyl—4—oxo-4H—quinazolin—3~yl)—piperidine—2,6—dione, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, e, hydrate, co~crystal, clathrate, or polymorph thereof; and (ii) a pharmaceutical composition comprising hematopoietic growth factor, cytokine, anticancer agent, antibiotic, a cox~2 inhibitor, immunomodulatory agent, immunosuppressive agent, osteroid, or a pharmacologically active mutant or derivative thereof, or a combination thereof.
In one embodiment, provided herein is a kit comprising (i) a phannaceutical composition comprising 3-(5-amincmethyl-4—oxo-4H—quinazoliii—3-yl)—piperidine-2,6- dione, or an enantiomer or a e of enantiomers thereof, or a ceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof; and (ii) a pharmaceutical composition comprising oblimersen, melphalan, G~CSF, GM-CSF, EPO, a cox-2 inhibitor, topotecan, pentoxifylline, taxotere, iritotecan, ciprofloxacin, thasone, doxorubicin, vincristine, IL 2, IFN, dacarbazine, Ara-C, vinorelbine, or isotretinoin.
In another embodiment, provided herein is a kit comprising (i) a ceutical ition comprising 3—(5-aminomethyloxo-4H-quinazolinyl)-piperidine—2,6~ dione, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, stal, clathrate, or poiymorph thereof; and (ii) umbilical cord blood, placental blood, peripheral blood stem cell, hematopoietic stem cell preparation or bone . 4. BRIEF DESCRIPTION OF THE FIGURES Figures 1A to 1D: tory effect of 3~(5—aminomethyl—4-oxo-4H—quinazolin-3— peridinc—2,6—dione (Comp. Formula I) on NFKB activity in DLBCL cells.
Figure 2: Antiproliferative effect of 3-(5~amincmethyl~4-oxo—4H—quinazolin-3— yl)-piperidine-2,6~dione (Compound of Formula I) in an in vitro DLBCL celLbased assay.
Figure 3: 3-(5-amino—2-methyloxo—4H~quinazolin—3~yl)-piperidine—2,6-dione (Compound of Formula I) costimulates T cells and enhances ne production.
Figure 4: Anti-angiogenic effect of 3-(5~amino-2—methyl-4—oxo~4H—quinazolin yl)~piperidine-2,6~dionein an in vitro human umbiligcal expant assay.
Figures 5A & SB: Antiproliferative effect of 3-(5-aminomethyl~4-oxo—4H— quinazolinyl)—piperidine-2,6adione in an in vitro multiple myleoma (MM) cell-based assay.
Figure 6: In vitro tumor inhibition of Antiproliferative effect of 3—(5-amino methyl-4—oxo-4H—quinazolin-3~yl)-piperidine~2,6—dione in a N929 xenograft model. s 7A—7C: Cereblon expression tes the effects of 3-(5-amino—2-methyl— 4-oxo-4H—quinazolin-3—yl)—piperidine-2,6-dione in ABC—DLBCL cell lines.
Figure 8: Knockdown ofCRBN abrogated Gl arrest induced by 3~(5—amino methyloxo—4H-quinazolinyl)—piperidine—2,6~dione.
Figure 9: CRBN knockdown abrogates effect of 3—(5—amino—2-methyl-4~oxo—4H— quinazolinyl)-piperidine—2,6~dione on phosphorylation ofpr and IRF-4 in H929 cells.
Figure 10: Antiproliferative activity of3-(5-amino-«2—methyl—4—oxo-4H-quinazolin- 3-yl)—piperidine-2,6-dione inhibits in CRBN—sensitive myeloma cells.
Ix.) U: Figure 11: Cereblon expression tes anti—invasive activity of 3-(5-amino methyloxo-4H-quinazolin~3~yl)-piperidine-2,6—dione.
Figures 12A- 1 21: 3-(5 —amino-2~methyloxo—4H—quinazolin—3-yl}‘piperidine-2,6- dione inhibits hypoxia—induced HlFl-a expression in solid tumor cell lines.
Figures lBA & 138: 3«(5-amino—24nethyl~4~oxo-4H~quinazolin-3~yi)-piperidine~ 2,6-dione inhibits breast cancer cell colony formation.
Figure 14: 3-(5-amino—2—methyl-4—oxo-4H~quinazolin~3—yl)-piperidine—2,6—dione inhibits U87 glioiblastoma tumor cell .
. DETAILED PTION OF THE INVENTION Provided herein are methods of treating, managing, or preventing cancer, which U1 comprise administering to a patient in need of such treatment, management, or prevention a therapeutically or prophylactically effective amount of 3—(5—amino—2-methyloxo—4H~ quinazolin—3 ~yl)-piperidine-2,6-dione, or an enantiomer or a mixture of omers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof as a single agent or as a part of a combination therapy.
In certain embodiments, 3—(5—amino-2—methyloxo-4H—quinazolinyl)- piperidine—2,6-dione, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceuticaliy acceptable salt, e, hydrate, co—crystal, ate, or rph thereof, is administered in combination with one or more onal drugs (or d active agents”) for use in the treatment, management, or prevention of cancer. Second active agents include small molecules and large les (eg, proteins and antibodies), some examples of which are provided herein, as well as stem cells. Methods or therapies, that can be used in combination with the administration of the compound provided herein include, but are not d to, surgery, blood transfusions, immunotherapy, biological therapy, radiation therapy, and other non-drug based therapies presently used to treat, prevent or manage cancer. In certain embodiments, the compound provided herein may be used as a vaccine adjuvant.
In some embodiments, the methods ed herein are based, in part, on the ery that the expression of certain genes or proteins associated with certain cancer cells may be utilized as biomarkers to indicate the effectiveness or progress of a e treatment. Such cancers include, but are not limited to, lymphoma. non-Hodgkin’s lymphoma, multiple a, ia, acute myeloid leukemia (AML), and solid tumors.
In certain embodiments, the cancer is of the activated B-cell phenotype in non-Hodgkin’s lymphoma. In particular, these biomarkers can be used to predict, assess and track the effectiveness of patient treatment with 3-(5-amino—2«methyl—4-oxo—4H-quinazoiin~3-yl)- piperidine~2,6~dione.
In some embodiments, the methods provided herein are based, in part, on the discovery that cereblon (CRBN) is associated with the anti—proiiferative activities of certain drugs, such as 3-(S~amino-Z-methyl-4~oxo-4H-quinazoiin~3~yi)-piperidine-2,6-dione. In some embodiments, CRBN may be utilized as a biomarker to indicate the effectiveness progress of a disease treatment with 3-(5~aminomethyl-4—oxo-4H~quinazolin—3-yl)- piperidine—2,6-dione. Without being bound by a particular theory, CRBN binding contribute to or even be required for anti~proliferative or other activities of certain compounds, such as 3-(5-aminc-2~methyl—4-oxo-4H-quinazolinyl)-piperidine~2,6—dione Without being d to a particular theory, 3{5-amino-2~methyl—4-oxo-4H« quinazolinyl)—piperidine-2,6-dione can mediate growth inhibition, sis and inhibition of angiogenic factors in certain types of cancer such as lymphoma, non—Hodkin’s lymphoma, multiple myeloma, leukemia, AML, and solid tumors. Upon examining the expression of several cancer-related genes in several cell types before and after the treatment with 3—(5—amino-2—methyl—4-oxo-4H—quinazolinyl)«piperidine—2,6~dione, it discovered that the expression levels of several cancer-related genes or proteins can be used as biomarkers for predicting and monitoring cancer treatments.
It was also discovered that the level ofNF-KB activity is ed in cells of the activated B-cell phenotype in non-Hodkin’s ma relative to other types of lymphoma cells, and that such cells may be sensitive to 3-(5«amino-2—methyl~4-oxo—4H-quinazolin—3- yl)-piperidine-2,6-dione treatment. This suggests that the baseline activity ofNF-KB activity in lymphoma cells may be a predictive biomarker for 3-(5-arninomethyl-4—oxo- 4H—quinazolinyl)-piperidine-2,6~dione treatment in non-Hodgkin’s lymphoma patients.
Therefore, in certain embodiments, provided herein are methods for ting tumor response to treatment in a non—Hodgkin’s lymphoma patient. In one embodiment, provided herein is a method of predicting tumor response to treatment in a non—Hodgkin’s ma patient, the method comprising obtaining tumor tissue from the patient, purifying protein or RNA from the tumor, and measuring the presence or absence of a biomarker by ,e.g., protein or gene expression analysis. The expression monitored may be, for example, mRNA expression or protein expression. In certain embodiments, the biomarker is a gene associated with an activated B~cell phenotype of DLBCL. The genes are selected from the group consisting of IRF4JMUMl, FOXPl, SPIB, CARDI I and BLIMP/PDRMl. In one embodiment, the biomarker is NF—KB.
In another embodiment, the method comprises ing tumor cells from the t, culturing the cells in the presence or absence of 3—(5~amino-2—methyl~4-oxo-4H— quinazolin—3-yI)-piperidine-2,6-dione, purifying RNA or protein from the ed cells, and ing the ce or e of a biomarker by, eg, gene or n sion analysis.
In certain ments, the presence or absence of a biomarker is measured by quantitative real-time PCR (QRT—PCR), microarray, flow cytometry or immunofluorescence. In other embodiments, the presence or absence of a biomarker is measured by based methodologies or other similar methods known in the art.
The methods ed herein encompass methods for screening or identifying cancer patients, ag, dgkin’s lymphoma patients, for treatment with 3—(5—amino-2— methyl-4—oxo—4H—quinazolin—3—yl)-piperidine~2,6-dione. In particular, provided herein are methods for selecting patients having, or who are likely to have, a higher response rate to a therapy with 3-(5-aminomethyl~4-oxoe4H—quinazolin~3—yl)-piperidine—2,6-dione.
In one embodiment, the method comprises the identification of patients likely to respond to therapy by obtaining tumor cells from the patient, culturing the cells in the presence or e of 3~(5~amino~2~methyl—4—oxo-4H—quinazolin—3-yl)—piperidine-2,6~ dione, purifying RNA or protein from the cultured cells, and measuring the presence or absence of a specific biomarker. The expression monitored can be, for example, mRNA expreSSion or protein expression. The expression in the treated sample can increase, or in some cases, decrease, for example, by about 1.5X, 2.0X, 3X, 5X, or more. In certain embodiments, the biomarker is a gene ated with an activated B~cell phenotype. The genes are selected from the group consisting of IRF4/MUM1, FOXPI, SPIB, CARDll BLIMP/PDRMI. In one embodiment, the biomarker is NF-KB. Baseline levels of expression of these genes can be tive of the sensitivity of a cancer to treatment with 3— (5—amino~2-methyl—4-oxo-4H—quinazolin-3~yl)-piperidine-2,6~dione.
In one embodiment, IRF4/MUM1 expression in cancer cells, e.g., ABC-subtype lymphoma, can be decreased with the ent of 3-(5-amino—2—methyl~4—oxo~4l—l~ quinazolin-3nyl)~piperidine-2,6-dione. In some embodiments, IRF4 downregulation by 3- is.) U1 (5-amino~2-methyl-4—oxo—4H-quinazolinyl)-piperidine-—2,6-dione can be a potential pharmacodynamic biomarker.
In another ment, provided herein is a method of monitoring tumor se to treatment with 3—(5-amino-2—methyloxo—4H-quinazolin—3—yi)-piperidine-2,6-dione in a lymphoma, non—Hodgkin’s lymphoma, multiple myeloma, leukemia, AML or a solid tumor patient. The method comprises obtaining a biological sample from the patient, measuring the sion of one or more biomarkers in, the biologicai sample, administering 3-(5- amino-2~methyl~4-oxo-4H—quinazolinyl)—piperidine-2,6—dione to the patient, thereafter obtaining a second ical sample from the patient, measuring biomarker expression in the second ical sample, and comparing the levels of biomarker expression, where an 1— increased level of biomarker sion after treatment indicates the likelihood of an effective tumor response. In one embodiment, a decreased level of biomarker expression after treatment indicates the likelihood of effective tumor se. In certain embodiments, the biomarker is a gene associated with an activated B—cell phenotype of non- Hodgkin’s lymphoma. The genes are ed from the group consisting of IRF4/MUM1, FOXPI, SPIB, CARDll and BLIMP/PDRMI. In one ment, the biomarker is NF- In n embodiments, the method ses measuring the expression of one or more biomarkers genes associated with an ted B-cell phenotype. The genes are selected from the group consisting of lRF4/MUM1, FOXPI, SPIB, CARDll and BLIMP/PDRMI . The expression monitored can be, for example, mRNA expression or protein expression. The expression in the treated sample can increase, for example, by about 15X, 2.0K, 3X, 5X, or more.
In yet another embodiment, a method for monitoring patient compliance with a drug treatment protocol is provided. The method comprises obtaining a biological sample from the patient, measuring the expression level of at least one biomarker in the , and determining if the expression level is increased or decreased in the patient sample compared to the expression level in a control untreated sample, wherein an increased or sed sion indicates patient ance with the drug treatment protocol. In one embodiment, the expression of one or more biomarker is increased. The expression monitored can be, for example, mRNA expression or protein expression. The expression in the treated sample can increase, for example, by about 1.5X, 2.0X, 3X, 5X, or more. In certain embodiments, the biomarker is a gene associated with an activated B-cell phenotype.
The genes are selected from the group consisting of IRF4/MUMI, FOXPI, SPIB, CARDll and BLIMP/PDRMl. In one embodiment, the biomarker is NF—KB.
In another embodiment, a method of predicting the sensitivity to treatment with 3- (5-amino—2«methyl~4~oxo-4H—quinazolinyl)~piperidine—2,6-dione in a ma, non« Hodgkin’s lymphoma, multiple a, leukemia, AML or a solid tumor t is provided. In one embodiment, the patient is a non-Hodgkin‘s lymphoma patient, specifically, a DLBCL patient. The method comprises obtaining a biological sample from the patient, optionally isolating or purifying mRNA from the biological sample, amplifying the mRNA transcripts by, eg, RT-PCR, Where a higher ne level of one or more specific biomarkers indicates a higher likelihood that the cancer will be sensitive to treatment with 3-(5-amino-2~methyl~4-oxo-4H-quinazolinyl)-piperidine—2,6-dione. In one embodiment, the biomarker is a gene associated with an activated B-cell phenotype selected from the group consisting of IRF4/MUM1, FOXPl , SPIB, CARDll and BLIMP/PDRMI.
In another embodiment, the method of predicting ivity to treatment with 3-(5- Ui aminomethyl~4-oxo-4H-quinazolinyl)—piperidine-2,6-dione in an NHL, eg, a DLBCL patient, comprises obtaining a tumor sample from the t, embedding the tumor sample into a paraffin—embedded, formalin-fixed block, and staining the sample with antibodies to CD20, CD10, bcla6, IRF4/MUMI, bcl-2, cyclin D2, and/or FOXPI, as described in Hans at 61]., Blood, 2004, 103: 275-282, which is hereby incorporated by reference in its entirety. In one embodiment, CD10, bcl—6, and IRF4/MUM-l staining can be used to divide DLBCL into GCB and non-GCB subgroups to predict an outcome.
In one ment, provided herein is a method for predicting tumor response to treatment in a non-Hodgkin’s lymphoma patient, sing: (i) obtaining a biological sample from the patient; (ii) measuring activity of the NF—KB pathway in the biological sample; and (iii) ing the level ofNF-KB activity in the biological sample to that of a biological sample of a non-activated B—cell lymphoma subtype; wherein an increased level of NF-KB activity relative to non-activated B-cell e lymphoma cells indicates a likelihood of an effective patient tumor se to 3-(5-amino- yloxo-4H-quinazolin-3—yl)—piperidine—2,6-dione treatment.
In one embodiment, measuring activity of the NF-KB pathway in the biological sample comprises measuring the level ofNF-KB in the biological sample.
In one embodiment, ed herein is a method of monitoring tumor response to treatment in a non-Hodgkin’s lymphoma t, comprising: (i) obtaining a biological sample from the patient; (ii) measuring the ievel ofNF-KB ty in the biologicai sample; (iii) administering a therapeuticaliy effective amount of 3{5amino-2~methyloxo—4H— quinazolin—3~yl)-piperidinc-2,6-dione, or a salt, solvate or hydrate thereof to the patient; (iv) obtaining a second icai sample from the patient; (v) measuring the level of NF~KB activity in the second bioiogicai sample; and (vi) comparing the level ofNF-KB activity in the first biological sample to that in the second biological sample; wherein a decreased levei ofNF-KB activity in the second ical sample relative to the first ical sample indicates a likeiihood of an effective patient tumor response. - 23 .. 2012/028498 In one ment, provided herein is a method for monitoring patient compliance with a drug treatment protocol in a non-Hodgkin’s lymphoma patient, comprising: (i) obtaining a biological sample from the patient; (ii) measuring the level OfNF-KB activity in the biological sample; and U! (iii) comparing the level ofNF-KB activity in the biological sample to a l ted sample; n a decreased level ofNF—KB activity in the biological sample relative to the control indicates patient compliance with the drug treatment ol.
In one embodiment, the non—Hodgkin’s lymphoma is diffuse large B-cell lymphoma.
In another embodiment, the level ofNF-KB activity is measured by an enzyme linked immunosorbent assay.
In one embodiment, provided herein is a method for predicting tumor response to treatment in a dgkin’s lymphoma patient, comprising: (i) obtaining a biological sample from the patient; (ii) culturing cells from the biological sample; (iii) ing RNA from the cultured cells; and (iv) fying increased expression of a gene associated with the activated B-cell phenotype of non—Hodgkin’s lymphoma relative to control non-activated B-cell phenotype of non-Hodgkin’s lymphoma; wherein increased sion of a gene associated with the activated B—cell phenotype of non-Hodgkin’s lymphoma indicates a likelihood of an effective patient tumor response to 3~(5~aminomethyl—4-oxo-4H—quinazolin—3-yl)~piperidine-2,6—dione treatment.
In one embodiment, increased expression is an increase of, about 15X, 2.0K, 3X, 5X, or more.
In one ment, the gene associated with the activated B-cell phenotype is selected from the group consisting of IRF4/MUM1, FOXPI, SPIB, CARDll and BLIMP/PDRMI.
In one embodiment, identifying the expression of a gene associated with the activated B-celi phenotype of non-Hodgkin’s ma is performed by quantitative real— time PCR.
Also provided herein is a method for treating or managing non-Hodgkin’s lymphoma, comprising: (i) identifying a patient having non-Hodgkin’s lymphoma sensitive to treatment with 3«(5«amino~2—methyl-4~oxo—4H—quinazolin—3-yl)-piperidine~2,6—dione; and (ii) administering to the patient a therapeutically effective amount of 3‘(5—amino methyl~4-oxo‘4H—quinazolin—3-yl)-piperidine-2,6-dione, or a ceutically acceptable salt, solvate or hydrate thereof.
In one embodiment, the non~Hodgkin’s lymphoma is diffuse large B-cell lymphoma.
In another embodiment, the dgkin’s lymphoma is of the activated B—cell phenotype.
In another ment, the diffuse large B-cell lymphoma is terized by the expression of one or more biomarkers pressed in RIVA, U2932, TMD8, OCI~Ly3 or OCLLle cell lines.
In r embodiment, the diffuse large B-cell lymphoma is characterized by the expression of one or more biomarkers overexpressed in RIVA, U2932, TMDS or OCI-Lle cell lines.
In one embodiment, identifying a patient having lymphoma sensitive to treatment with 3-(5—amino-2—methyl—4-oxo-4H—quinazolinyl)—piperidine-2,6-dione comprises characterization of the lymphoma phenotype of the patient.
In one embodiment, the lymphoma phenotype is characterized as an activated B—cell subtype.
In one embodiment} the lymphoma phenotype is characterized as an activated B—cell subtype of diffuse large B~cell lymphoma.
In certain embodiments, identification of the lymphoma phenotype comprises obtaining a ical sample from a patient having lymphoma. In one embodiment, the biological sample is a cell culture or tissue sample. In one embodiment, the biological sample is a sample of tumor cells. In another embodiment} the biological sample is a lymph node biopsy, a bone marrow biopsy, or a sample of peripheral blood tumor cells. In one embodiment, the bioiogical sample is a blood sample.
In one embodiment fying a patient having non-Hodgkin’s lymphoma sensitive to treatment with 3-(5 -amino-2~methyl-4«oxo-4H—quinazolin~3~yl)—piperidine-2.6-—dione comprises identification of a gene associated with an ted B-ceil ype. In one ment: the gene associated with the activated B—celi phenotype is selected from the group consisting of IRFéi/MI IM] FOXPI , SPIB, CARDII and BLIMPXPDRMI.
In one embodiment, identifying a patient having non—Hodgkin’s lymphoma sensitive to ent with 3~(5-amino-2—methyl-4~oxo—4H-quinazolin-3—yl)-piperidine~2,6-dione comprises measuring the level ofNF-KB activity in the patient. In another embodiment, measuring the level of NF-KB ty in a patient comprises measuring the baseline NF-KB activity level in tumor cells obtained from the patient.
In another embodiment, the diffuse large B-cell lymphoma is characterized by one or more of the following: (i) over expression of SPIB, a poietic—specific Ets family transcription factor required for survival of activated B—cell subtype cells; (ii) higher constitutive IRF4/MUM1 expression than GCB subtype cells; (iii) higher constitutive FOXPl expression up-regulated by trisomy 3; IO (iv) higher constitutive Blimpl, 229., PRDMl, expression; and (V) higher constitutive CARDll gene expression; and (vi) an increased level of NF-KB ty relative to non-activated B-cell subtype DLBCL cells.
Additional prognostic factors that may be used concurrently with those provided herein are prognostic s of disease (tumor) , absolute lymphocyte count (ALC), time since last rituximab y for lymphomas, or all of the above.
Also ed herein is a method of selecting a group of cancer patients based on the level of CRBN expression, or the levels of DDBI, DDB2, IRF4 or NFKB expression Within the cancer, for the purposes of predicting clinical response, monitoring clinical response, or 2O monitoring patient compliance to dosing by 3~(5-amino-Z—methyloxo-4H*quinazolin—3- yl)-piperidine-2,6-dione, a stereoisomer thereof, or a ceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof; wherein the cancer patients are selected from le myeloma, non-Hodgkin’s lymphoma, diffuse large B—cell lymphoma, melanoma and solid tumor patients. Baseline levels of sion of these genes can be predictive of the sensitivity ofa cancer to treatment with 3~(5-aminomethyl~ 4-oxo—4H-quinazolin»3~yl)—piperidine-2,6-dione.
In one embodiment, IRF4E’MUMI expression in cancer cells, cg, ABC-subtype lymphoma, can be decreased with the treatment of mino-Z-methyl—4-oxo-4H- quinazolin—3~yl)~piperidine~2,6-dione. In some embodiments, IRF4 downregulation by 3~ (S-amino-Z—methyloxo~4H—quinazolin—3-yl)—piperidinc-2,6-dione can be a potential pharmacodynamic biomarker.
In one ment, the cancer patients are multiple myeloma patients.
In one embodiment, the cancer patients are non~Hodgkin’s lymphoma patients.
In one embodiment, the method of selecting a group of cancer ts is based on the level of DDBI expression within the cancer.
In one embodiment, the method of selecting a group of cancer patients is based on the level of DDB2 expression within the cancer.
In one embodiment, the method of selecting a group of cancer patients is based on the level of [RF-4 expression within the cancer.
In one embodiment, the method of selecting a group of cancer patients is based on the level ofNFKB expression within the cancer.
In another embodiment, the method comprises selecting a group of cancer patients responsive to treatment with 3-(5~aminomethyl-4—oxo-4H-quinazolin—3~yl)—piperidine- 2,6-dione, a stereoisomer thereof, or a pharmaceutically able salt, solvate, hydrate, stal, clathrate, or polymorph thereof; based on the level of CRBN expression, or the levels of BBB], DDBZ, IRF4 or NFKB expression within the tis T cells, B cells, or plasma cells, for the purposes of predicting clinical response, monitoring clinical response, or monitoring patient compliance to dosing by 3~(5-amin0—2—methyl—4—oxo-4H-quinazolin- 3-yl)-piperidine—2,6—dione, a stereoisomer thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof.
In one embodiment, the cancer patients are selected from multiple myeloma, non—Hodgkin’s ma, diffuse large B-cell ma, melanoma and solid tumor patients.
Also provided herein are methods of treating cancer, cg, lymphoma, non—Hodgkin's lymphoma, multiple a, leukemia, acute myeloid leukemia (AML), and solid tumors, which result in an improvement in overall survival of the patient. In some embodiments, the improvement in l survival of the t is observed in a patient population sensitive to treatment with 3~(5-amino-2*methyl—4-oxo—4H-quinazolin—3~yl)-piperidine-2,6— k) Um dione. In some embodiments, the patient population sensitive to ent with 3—(5-amino« 2-methyIoxo-4H-quinazolinyl)—piperidine-2,6-dione is characterized by one or more biomarkers provided herein. in other embodiments, provided herein are methods of treating cancer, e. g. , Iyrnphoma, non-Hodgkin’s iymphoma, multiple myeioma, ia, acute myeloid leukemia (AML), and solid tumors, which result in disease free survival of the t. In in, a patient population some embodiments, disease free survival of the t is observed sensitive to treatment with 3-(5—aminomethyioxo—4H~quinazoii11yl)~piperidine-2,6~ dione. In some embodiments, the patient population sensitive to ent with 3‘(5~amino- 2-methyloxo-4H-quinazolin—3-yl)—piperidine-2,6~dione is characterized by one or more biomarkers provided herein.
In other embodiments, ed herein are methods of treating cancer, ag. , lymphoma, dgkin's ma, multiple a, leukemia, acute myeloid leukemia (AML), and solid tumors, which result in an improvement in the objective response rate in the patient tion. In some embodiments, the patient population sensitive to treatment with minomethyl~4-oxo-4H—quinazolinyl)—piperidine—2,6— dione. In some embodiments, the patient population sensitive to treatment with 3—(5—amino— 2«methyloxo-4H-quinazolinyl)-piperidine~2,6-dione is characterized by one or more biomarkers provided herein.
In other embodiments, ed herein are methods of treating , e.g, lymphoma, non— lymphoma, Hodgkin's lymphoma, multiple myeloma, leukemia, acute myeloid leukemia (AML), and solid tumors, which result in an improved time to progression or progression-free survival of the patient. In some embodiments, the improved time to progression or progression—free survival of the patient is observed in a patient population sensitive to treatment with 3-(5~aminomethyl-4~oxo-4H~quinazolinyl)— piperidine-2,6—dione. In some embodiments, the patient population sensitive to treatment with 3~(5-amino-2—methyloxo—4H-quinazolin-3~yl)—piperidine—2,6—dione is characterized by one or more biomarkers provided .
Also provided herein are kits useful for predicting the likelihood of an effective lymphoma, non-Hodgkin’s lymphoma, multiple myeloma, leukemia, AML or solid tumor treatment or for monitoring the effectiveness of a treatment with 3—(5-amino—2—methyl—4- oxo—4H-quinazolin—3~yl)~piperidine~2,6-dione. The kit comprises a solid support, and a means for detecting the expression of a ker in a biological sample. Such a kit may employ, for example, a dipstick, a membrane, a chip, a disk, a test strip, a filter, a microsphere, a slide, a multiwell plate, or an optical fiber. The solid support of the kit can be, for example, a plastic, silicon, a metal, a resin, glass, a membrane, a particle, a precipitate, a gel, a polymer, a sheet, a sphere, a polysaccharide, a capillary, a film, a plate, or a slide. The biological sample can be, for example, a cell culture, a cell line, a tissue, an oral tissue, gastrointestinal , an organ, an organelle, a biological fluid, a blood , a urine sample, or a skin sample. The biological sample can be, for example, a lymph node biopsy, a bone marrow biopsy, or a sample of peripheral blood tumor cells.
In one embodiment, the kit comprises a solid support, nucleic acids ting the t, where the nucleic acids are compiementary to at least 20, 50, 100, 200, 350, or in a NHL, more bases of mRNA of a gene associated with an activated B—cell ype and a means for ing the expression of the mRNA in a biological sample. In one embodiment, the gene associated with the activated B-cell phenotype is selected from the CARDII and BLIMP/PDRMI. group consisting of lRF4/MUM1, FOXPI, SPIB, In one embodiment, a kit useful for predicting the likelihood of an effective lymphoma, non-Hodgkin’s lymphoma, multiple myeloma, leukemia, AML or solid tumor treatment, or for monitoring the effectiveness of a ent with 3-(5-amino—2-methyl—4- oxo-4H—quinazolin-3~yl)—piperidine-2,6-dione is provided. The kit comprises a solid in a biological sample. In one support, and a means for detecting the sion ofNF-KB embodiment, the biological sample is a cell culture or tissue sample. In one embodiment, the biological sample is a sample of tumor cells. In another embodiment, the biological sample is a lymph node biopsy, a bone marrow biopsy, or a sample of peripheral blood tumor cells. In one embodiment, the biological sample is a blood sample. In one embodiment, the NHL is DLBCL.
In certain embodiments, the kits provided herein employ means for ing the expression of a biomarker by quantitative real-time PCR (QT—PCR), microarray, flow cytometry or immunofluorescence. In other embodiments, the expression of the biomarker is measured by ELISA-based methodologies or other similar methods known in the art.
Additional mRNA and protein expression ques may be used in connection with the methods and kits provided herein, e.g, CDNA hybridization and cytometric bead array methods.
In one embodiment, provided herein is a kit for predicting tumor response to in a treatment with 3-(5~aminomethyls4-oxo-4H—quinazolin—3~yl)-piperidine-2,6-dione non-Hodgkin’s lymphoma patient, comprising: (i) a solid support; and (ii) B»cell phenotype a means for detecting the sion of a ker of an activated of non-Hodgkin’s ma in a biological sample.
In one ment, the ker is NF-KB.
In one embodiment, the biomarker is a gene associated with the activated B-eeli phenotype and is selected from the group consisting of IRF4XMUMI, FOXPI , SPIB, CARDH and BLIMP/PDRMl.
In particular s of the invention, a 3—-(S-amino-2—methyl-4—oxo~4H—quinazolin- 3-yl)-piperidine~2,6-dione is administered in combination with a therapy conventionally used to treat, prevent or manage cancer. Examples of such conventional ies include, but are not limited to, y, chemotherapy, ion therapy, hormonal therapy, biological y and immunotherapy.
Also provided herein are pharmaceutical compositions, single unit dosage forms, dosing regimens and kits which comprise 3«(5-arnino-Z-rnethyl-4—oxo-4H—quinazolin—3—yl)- piperidine~2,6—dione, or a pharmaceutically able salt, solvate, hydrate, stereoisomer, clathrate, or prodrug f, and a second, or additional, active agent. Second active agents include specific combinations, or “cocktails,” of drugs.
In some ments, the methods for treating, preventing and/or managing lymphomas provided herein may be used in patients that have not responded to standard treatment. In one embodiment, the lymphoma is relapsed, refractory or resistant to conventional y.
In other embodiments, the methods for treating, ting and/or managing lymphomas provided herein may be used in treatment naive patients, 1.6. , patients that have not yet received treatment.
In certain embodiments, 3«(5-amino—2-rnethyl-4—oxo—4H-quinazolin-3—yl)- piperidine-2,6-dione, or an enantiomer or a e of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, is administered in combination or alternation with a therapeutically effective amount of one or more additional active agents. Second active agents include small molecules and large molecules (e.g., proteins and antibodies), examples of which provided herein, as well as stem cells. Methods or therapies that can be used in combination with the administration of the compound provided herein include, but are not limited to, surgery, blood transfusions, immunotherapy, biological therapy, radiation y, and other non—drug based therapies presently used to treat, prevent or manage e and conditions associated with or terized by red angiogenesis.
In one embodiment, the additional active agent is selected from the group consisting of an alkylating agent, an adenosine analog, a glucocorticoid, a kinase inhibitor, a SYK inhibitor, a PDE3 inhibitor, a PDE7 inhibitor, doxorubicin, chlorambucii, vincristine, bendamustine, forskolin, rituximab, or a combination f.
In one embodiment, the additional active agent is rituxirnab.
In one embodiment, the giucocortieoid is hydrocortisone or thasone.
In one embodiment, 3~(5-amino—2~methyl~4-oxo-4H-quinazoiinyl)-piperidine- 2,6—dione is administered in an amount of about 5 to about 50 mg per day.
In one ment, 3-(5—amino~2-methyl-4~oxo-4H-quinazolinyl)-piperidine- one is administered in an amount of about 5 to about 25 mg per day.
In another ment, 3-(5«amino-2»methyl—4—oxo-4H—quinazolinyl)- piperidine~2,6—dione is administered in an amount of about 5, IO, 15, 25, 30 or 50 mg per day.
In another embodiment, 10 or 25 mg of minomethyl—4-oxo-4H-quinazolin— 3-yl)~piperidine~2,6-dione is administered per day.
In one embodiment, 3~(5-amino-2~methyl~4-oxo~4H-quinazolinyl)—piperidine- 2,6-dione is administered twice per day.
IO Provided herein are pharmaceutical compositions (e.g., single unit dosage forms) that can be used in methods disclosed herein. In certain embodiments, the pharmaceutical compositions se 3—(5—amino—2—methyl~4—oxo—4H—quinazolinyl)-piperidine-2,6- dione, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, stal, clathrate, or polymorph thereof, and a second active agent.
In one embodiment, 3—(5~aminomethyl~4-oxo-4H-quinazolin-3~yl)-piperidine— one is orally administered.
In one embodiment, 3—(5-amino—2—methyl—4—oxo-4H-quinazolin~3-yl)-piperidine— 2,6—dione is administered in a capsule or tablet.
In one embodiment, 3—(5-aminomethyl-4—oxo—4H—quinazolin-3—yl)—piperidine- 2,6-dione is administered for 21 days ed by seven days rest in a 28 day cycle. .1 DEFINITIONS To facilitate understanding of the disclosure set forth herein, a number of terms are defined below.
The term “subject” or “patient" refers to an animal, including, but not limited to, a mammal, including a primate (cg, human), cow, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms “subject” and “patient” are used interchangeably herein in reference, for example, to a mammalian subject, such as a human subject.
As used herein, and unless otherwise specified, the terms “treat,” “treating” and “treatment” refer to the eradication or amelioration of a disease or disorder, or of one or more symptoms associated with the disease or disorder. in certain embodiments, the terms refer to minimizing the spread or worsening of the disease or disorder ing from, the administration of one or more prophylactic or therapeutic agents to a patient with such a e or disorder. In some embodiments, the terms refer to the administration of a compound provided herein, with or without other additional active agent, after the onset of symptoms of the particular disease.
As used herein, and unless otherwise specified, the terms “prevent,“ nting” U: and “prevention” refer to the prevention of the onset, recurrence or spread of a disease or disorder, or of one or more symptoms thereof. In certain embodiments, the terms refer to the treatment with or administration of a compound provided , with or without other additional active compound, prior to the onset of symptoms, particularly to patients at risk of diseases or disorders provided herein. The terms encompass the inhibition or reduction of a symptom of the particular disease. Patients with familial history of a disease in ular are candidates for preventive regimens in certain embodiments. In addition, patients who have a history of ing symptoms are also potential candidates for the prevention. In this regard, the term “prevention” may be hangeably used with the term “prophylactic treatment.” As used herein, and unless otherwise specified, the terms “manage,” “managing” and ement” refer to preventing or slowing the progression, spread or worsening of a disease or disorder, or of one or more symptoms thereof. Often, the beneficial s that a patient derives from a prophylactic and/or therapeutic agent do not result in a cure of the disease or disorder. In this regard, the term “managing” asses treating a patient who had suffered from the particular disease in an attempt to prevent or minimize the recurrence of the disease, or lengthening the time during which the remains in remission.
As used herein, and unless otherwise specified, a “therapeutically effective amount” of a compound is an amount sufficient to provide a therapeutic benefit in the ent or management of a disease or disorder, or to delay or minimize one or more symptoms associated with the disease or disorder. A therapeutically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other ies, which provides a therapeutic benefit in the treatment or management of the e or disorder.
The term “therapeutically effective amount” can encompass an amount that improves overall therapy, reduces or avoids symptoms or causes of disease or er, or enhances the therapeutic efficacy of another therapeutic agent.
As used herein, and unless ise specified, a “prophylacticaliy effective amount” of a compound is an amount ent to prevent a disease or disorder, or t its recurrence. A prophylactically effective amount of a compound means an amount of eutic agent, alone or in combination with other agents, which provides a prophylactic 2012/028498 benefit in the prevention of the disease. The term “prophylactically effective amount” can enhances the prophylactic encompass an amount that improves overall laxis or efficacy of another prophylactic agent.
The term “pharmaceutically acceptable carrier,” “pharmaceutically acceptable Ur excipient,” “physiologically acceptable carrier,” or “physiologically acceptable excipient” refers to a pharmaceutically-acceptable material, ition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material. In one embodiment, each with the other component is “pharmaceutically acceptable” in the sense of being compatible ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, Remington: The Science and Practice rmacy, 21st Edition; Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook ofPharmaceutical Excipients, 5th n; Rowe er al., Eds, The Pharmaceutical Press and the American Pharmaceutical Association: 2005; and Handbook ofPharmaceutical Additives, 3rd Edition; Ash and Ash Eds, Gower Publishing Company: 2007; Pharmaceutical Preformulation Formulation, Gibson Ed, CRC Press LLC: Boca Raton, FL, 2004). ,” as used herein, refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre—cancerous and ous cells and tissues.
“Neoplastic,” as used herein, refers to any form of dysregulated or unregulated cell growth, whether malignant or benign, resulting in abnormal tissue growth. Thus, astic cells” include malignant and benign cells having dysregulated or unregulated cell growth.
The term sed” refers to a situation where a t or a mammal, which has had a remission of cancer after therapy has a return of cancer cells.
As used herein, an “effective t tumor response” refers to any increase in the therapeutic benefit to the patient. An “effective patient tumor se” can be, for example, a 5%, 10%, 25%, 50%, or {00% se in the rate of progress of the tumor. An tive patient tumor response” can be, for e, a 5%, 10%, 25%, 50%, or 100% decrease in the physical symptoms of a cancer. An “effective patient tumor response” can also be, for example, a 5%, 10%, 25%, 50%, 100%, 200%, or more increase in the response of the patient, as measured by any suitable means, such as gene expression, cell counts, assay results, etc.
The term “likeiihood” generally refers to an increase in the probability of an event.
The term “likelihood” when used in reference to the effectiveness of a patient tumor -33.. response generally contemplates an increased probability that the rate of tumor progress or tumor cell growth will decrease. The term “likelihood” when used in reference to the effectiveness of a patient tumor response can also generally mean the increase of indicators, such as mRNA or protein expression, that may ce an increase in the progress in U: ng the tumor.
The term “predict” generally means to determine or tell in e. When used to “predict” the effectiveness of a cancer ent, for example, the term “predict” can mean that the likelihood of the outcome of the cancer ent can be determined at the outset, before the treatment has begun, or before the treatment period has progressed substantially.
The term “monitor,” as used herein, generally refers to the overseeing, ision, regulation, watching, tracking, or surveillance of an activity. For example, the term “monitoring the effectiveness of a compound” refers to tracking the effectiveness in treating a cancer in a patient or in a tumor cell culture. Similarly, the “monitoring,” when used in connection with patient compliance, either individually, or in a clinical trial, refers to the tracking or confirming that the patient is actually taking the immunomodulatory compound being tested as prescribed. The monitoring can be performed, for example, by ing the expression ofmRNA or protein biomarkers.
An improvement in the cancer or cancer-related disease can be characterized as a te or partial response. “Complete response” refers to an absence of clinically detectable disease with ization of any previously abnormal radiographic studies, bone marrow, and cerebrospinal fluid (CSF) or abnormal monoclonal protein measurements. “Partial response” refers to at least about a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% se in all measurable tumor burden (i.e., the number of malignant cells present in the subject, or the measured bulk of tumor masses or the quantity of abnormal monoclonal protein) in the absence of new lesions. The term “treatment” plates both a complete and a partial response.
The term “refractory or resistant” refers to a circumstance where a subject or a , even after intensive treatment, has residual cancer cells in his body.
The term “drug resistance” refers to the condition when a e does not respond to the treatment of a drug or drugs. Drug resistance can be either intrinsic, which means the disease has never been responsive to the drug or drugs, or it can be acquired, which means the disease ceases responding to a drug or drugs that the disease had previously responded to. In certain embodiments, drug resistance is intrinsic. In certain embodiments, the drug resistance is acquired.
The term “sensitivity” and “sensitive” when made in reference to treatment with compound is a relative term which refers to the degree of effectiveness of the compound in ing or decreasing the progress of a tumor or the disease being treated. For e, the term ased sensitivity” when used in reference to treatment of a cell or tumor in connection with a compound refers to an increase of, at least a 5%, or more, in the effectiveness of the tumor treatment.
The term “expressed” or “expression” as used herein refers to the transcription from in part to a region of a gene to give an RNA c acid molecule at least complementary or “expression” as one of the two nucleic acid strands of the gene. The term “expressed” used herein also refers to the translation from the RNA molecule to give a protein, a polypeptide or a portion thereof.
An mRNA that is “upregulated” is generally increased upon a given treatment or condition. An mRNA that is “downregulated” generally refers to a decrease in the level of expression of the mRNA in response to a given treatment or condition. In some situations, the mRNA level can remain unchanged upon a given treatment or condition.
An mRNA from a patient sample can be “upregulated” when treated with an immunomodulatory compound, as compared to a non-treated control. This upregulation can be, for e, an increase of about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 90%, 100%, 200%, 300%, 500%, 1,000%, 5,000% or more of the comparative control mRNA level.
Alternatively, an mRNA can be “downregulated”, or expressed at a lower level, in or other . A response to administration of certain immunomodulatory compounds downregulated mRNA can be, for e, present at a level of about 99%, 95%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 1% or less of the comparative control mRNA level.
Similarly, the level of a polypeptide or protein biomarker from a patient sample can be increased when treated with an immunomod’uiatory compound, as compared to a non~ treated l. This increase can be about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 90%, 100%, 200%, 300%, 500%, 1,000%, 5,000% or more of the comparative control protein level.
Alternatively, the level of a protein biomarker can be sed in response to administration of certain immunomoduiatorv nds or other agents. This decrease can be, for example, present at a level of about 99%, 95%, 90%, 80%, 70%, 60%, 50%, 40%, %, 20%, 10%, 1% or less of the comparative control protein level.
The terms mining”, “measuring”, “evaluating”, “assessing” and “assaying” as used herein generally refer to any form of measurement, and include determining if an element is present or not. These terms include both quantitative and/or qualitative determinations. Assessing may be relative or te. “Assessing the presence of” can include determining the amount of something present, as well as determining r it is t or .
As used herein and unless otherwise ted, the term “pharmaceutically acceptable salt” encompasses non-toxic acid and base addition salts of the nd to which the term refers. Acceptable non-toxic acid on salts include those derived from organic and inorganic acids or bases know in the art, which include, for example, hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, methanesulphonic acid, acetic acid, tartaric acid, lactic acid, succinic acid, citric acid, malic acid, maleic acid, sorbic acid, aconitic acid, salicylic acid, phthalic acid, embolic acid, enanthic acid, and the like.
Compounds that are acidic in nature are capable of forming salts with various pharmaceutically acceptable bases. The bases that can be used to prepare pharmaceutically acceptable base addition salts of such acidic compounds are those that form non—toxic base addition salts, i.e., salts containing pharmacologically acceptable cations such as, but not d to, alkali metal or alkaline earth metal salts and the calcium, magnesium, sodium or potassium salts in particular. Suitable organic bases include, but are not limited to, N,N dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, nediamine, meglumaine ylglucamine), lysine, and procaine.
As used herein and unless otherwise indicated, the term “solvate” means a compound provided herein or a salt thereof, that further includes a stoichiometric or non- stoichiometric amount of solvent bound by non-covalent intermolecular forces.
Where the solvent is water, the solvate is a hydrate.
As used herein and unless otherwise indicated, the term “prodrug” means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under ical conditions (in vitro or in vivo) to provide the compound. Examples of prodrugs include, but are not limited to, derivatives of the nd of Formula I provided herein that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable s, and biohydrolyzable phosphate analogues. Other examples of prodrugs include tives of the compound of Formula I provided herein that comprise -NO, -N02, ~ONO, or ONO; moieties. Prodrugs can be prepared using such methods as described in Burger ’s Medicinal? Chemistry and Drug Discovery, 172—178, 949-982 (Manfred E. Wolff ed., 5th ed. 1995), and Design ofProdrugs (H. Bundgaard ed., Elselvier, New York 1985).
As used herein and unless otherwise indicated, the terms “biohydrolyzable amide,” “biohydrolyzable ester,” “biohydrolyzable carbamate,” “biohydrolyzable carbonate,” “biohydrolyzable ureide,” and “biohydrolyzable phosphate” mean an amide, ester, carbamate, carbonate, ureide, or phosphate, respectively, of a compound that either: 1) does not interfere with the ical activity of the compound but can confer upon that compound advantageous properties in viva, such as , duration of action, or onset of action; or 2) is biologically inactive but is converted in vivo to the biologically active compound. Examples of biohydrolyzable esters include, but are not limited to, lower alkyl esters, lower acyloxyalkyl esters (such as acetoxylmethyl, yethyl, arbonyloxymethyl, pivaloyloxymethyl, and pivaloyloxyethyl esters), lactonyl esters (such as phthalidyl and thiophthalidyl esters), lower alkoxyacyloxyalkyl esters (such as methoxycarbonyl-oxymethyl, ethoxycarbonyloxyethyl and isopropoxycarbonyloxyethyl esters), alkoxyalkyl esters, choline , and acylamino alkyl esters (such as acetamidomethyl esters). Examples of rolyzable amides include, but are not limited to, lower alkyl amides, ino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl . Examples of rolyzable carbamates include, but are not limited to, lower alkylamines, substituted ethylenediamines, amino acids, hydroxyalkylamines, heterocyclic and heteroaromatic amines, and her .
As used herein and unless otherwise indicated, the term “stereomerically pure” means a composition that comprises one stereoisomer of a compound and is substantially free of other stereoisomers of that compound. For example, a stereomerically pure composition of a compound having one chiral center will be substantially free of the opposite enantiomer of the compound. A stereomericaliy pure ition of a nd having two chiral centers will be substantially free of other diastereomers of the compound.
In certain ments, a stereomericaily pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and iess than about 20% by weight of other stereoisomers of the compound, greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, greater than about 95% by weight of one stereoisomer of the nd and less than about 5% by weight of the other stereoisomers of the compound, or greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound. As used herein and unless otherwise indicated, the term “stereomerically ed" means a composition that comprises greater than about 60% by weight of one isomer of a compound, greater than about 70% by weight, greater than about 80% by weight of one stereoisomer of a compound. As used herein and unless otherwise indicated, the term “enantiomerically pure” means a stereomerically pure composition of a compound having one chiral center. Similarly, the term “stereomerically enriched” means a stereomerically enriched composition of a compound having one chiral center.
The term “about” or ximately” means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term “about” or “approximately” means within 1, 2, 3, or 4 standard deviations. In certain ments, the term “about” or “approximately” means within 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, %, 4%, 3%, 2%, 1%, 0.5%, or 0.05% ofa given value or range. .2 AL TRIALS ENDPOINTS FOR CANCER APPROVAL “Overall survival” is defined as the time from randomization until death from cause, and is measured in the intent—to—treat population. l survival should be evaluated in randomized lled studies. Demonstration of a statistically significant improvement in overall al can be considered to be clinically significant if the ty profile is acceptable, and has often supported new drug al.
Several endpoints are based on tumor assessments. These endpoints include disease free survival (DFS), objective response rate (ORR), time to progression (TTP), ssion- free survival (FPS), and time-to-treatment failure (TTF). The collection and analysis of data on these time~dependent endpoints are based on indirect assessments, calculations, and estimates (rag, tumor measurements).
Generally, “disease free al” (DFS) is defined as the time from randomization until recurrence of tumor or death from any cause. gh overall survival is a conventional endpoint for most adjuvant settings, DFS can be an important endpoint in situations where survival may be prolonged, making a survival endpoint impractical. DFS can be a surrogate for clinical benefit or it can provide direct evidence of clinical benefit.
This determination is based on the magnitude of the , its risk-benefit relationship, the disease setting. The definition of DFS can be complicated, particuiarly when deaths noted without prior tumor progression documentation. These events can be scored either as disease recurrences or as censored events. Although ali methods for statistical analysis of deaths have some limitations, considering all deaths (deaths from all causes) as recurrences can minimize bias. DFS can be overestimated using this definition? especially in ts who die after a long period without observation. Bias can be introduced if the frequency of long-term follow~up visits is dissimilar between the study arms or if dropouts are not random because of toxicity.
“Objective response rate” (OR) is defined as the proportion of patients with tumor size ion of a predefined amount and for a minimum time period. Response duration usually is measured from the time of initial response until documented tumor progression.
Generally, the FDA has defined ORR as the sum of l responses plus complete responses. When defined in this manner, ORR is a direct measure of drug antitumor activity, which can be evaluated in a single—arm study. If available, standardized ia should be used to ascertain response. A variety of response ia have been considered appropriate (cg, RECIST criteria) (Therasse et al., (2000) J. Natl. Cancer Inst, 92: 205-16).
The significance ofOR is assessed by its magnitude and duration, and the percentage of complete responses (no able evidence of tumor).
“Time to progression” (TTP) and “progression—free survival” (PFS) have served as primary endpoints for drug approval. TTP is defined as the time from randomization until objective tumor progression; TTP does not include deaths. PFS is defined as the time from randomization until objective tumor progression or death. Compared with TTP, PFS is the preferred regulatory endpoint. PFS includes deaths and thus can be a better correlate to overall survival. PFS assumes patient deaths are randomly related to tumor progression.
However, in situations Where the ty of deaths are unrelated to , TTP can be an acceptable endpoint.
As an endpoint to support drug approval, PFS can reflect tumor growth and be assessed before the determination of a al benefit. Its determination is not confounded by subsequent therapy. For a given sample size, the ude of effect on PPS can be larger than the effect on overall survivai. However? the formal validation of PFS as a surrogate for al for the many different malignancies that exist can be difficult. Data are mes insufficient to allow a robust evaluation of the correlation between effects on survival and PFS. Cancer trials are often small. and proven survival benefits of existing drugs are generaliv modest. The role of PPS as an endpoint to support licensing approval varies in different cancer settings. r an improvement in PPS represents a direct al benefit or a surrogate for clinical benefit depends on the magnitude of the effect and the enefit of the new treatment compared to available therapies.
“Time—to—treatment failure” (TTF) is defined as a composite endpoint measuring time from randomization to discontinuation of treatment for any reason, including disease progression, treatment toxicity, and death. TTF is not recommended as a regulatory endpoint for drug approval. TTF does not adequately distinguish efficacy from these additional variables. A regulatory endpoint should clearly distinguish the efficacy of the drug from toxicity, patient or ian Withdrawal, or patient intolerance. .3 THE COMPOUND The nd suitable for use in the methods provided herein is 3—(5-amino—2- methyl-4~oxo~4H—quinazolin-3~yl)-piperidine~2,6-dione, having the structure of Formula I: ” Nil NH2 00 N O H a) or an omer or a mixture of enantiomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof.
The compound of Formula I can be ed according to the methods described in the Examples provided herein or as described in US. Pat. No. 7,635,700, the disclosure of which is incorporated herein by reference in its entirety. The compound can be also synthesized according to other methods nt to those of skill in the art based upon the teaching herein.
The compound of Formula I ly inhibits TNF-a, lL—l B, and other inflammatory cytokines in LPS-stimulated hPBMC and human Whole blood. TNF-o is an inflammatory cytokine produced by macrophages and monocytes during acute ation. ”INF-or is sible for a diverse range of signaling events Within cells.
TNF-a may play a pathological role in cancer. t being limited by theory, one of the biological effects exerted by the immunomodulatory nd of Formula I is the reduction of synthesis of TNF-a. The immunomoduiatory compound of Formula I enhances the degradation of TNF—a mRNA. The compound of Formula I also potently inhibits lL-i B and stimulates lL-lO under these conditions.
Further, without being limited by theory, the compound of Formula I is a potent co- stimulator of T cells and increase cell eration in a dose dependent manner under appropriate conditions. 2012/028498 In certain embodiments, without being d by theory, the biological effects exerted by the immunomodulatory compound of Formula I include, but not limited to, anti- angiogenic and immune modulating effects.
In certain embodiments, the compound of Formula I is a solid. In certain U: embodiments, the compound of Formula I is hydrated. In certain embodiments, the compound of Formula I is solvated. In certain embodiments, the compound of Formula I is anhydrous. In n embodiments, the compound of Formula I is nonhygroscopic.
In certain embodiments, the solid compound of Formula I is amorphous. In certain embodiments, the solid compound of Formula I is crystalline. In certain embodiments, the solid compound of a I is in a crystalline form described in U.S. Provisional Pat. App.
No. 61/451,806, filed March I I, 2011, which is incorporated herein by reference in its The solid forms of the compound of Formula I can be prepared according to the s described in the sure of U.S. Provisional Pat. App. No. 61/451,806. The solid forms can be also prepared according to other methods apparent to those of skill in the art.
In certain embodiments, the compound of Formula I is a hydrochloride salt of 3-(5- amino~2~methyl~4-oxo-4H—quinazolinyl)—piperidine-2,6-dione, or an enantiomer or a mixture of enantiomers thereof; or a pharmaceutically acceptable solvate, hydrate, co- crystal, clathrate, or polymorph thereof. In certain embodiments, the hydrochloride salt is a solid. In n embodiments, the hydrochloride salt is anhydrous. In certain embodiments, the hydrochloride salt is nonhygroscopic. In n embodiments, the hydrochloride salt is ous. In certain embodiments, the hydrochloride salt is crystalline. In certain embodiments, the hydrochloride salt is in crystalline Form A.
The hloride salt of the compound of Formula I and solid forms thereof can be prepared according to the methods described in the disclosure of US. ional Pat. App.
No. ,806. The hydrochloride salt the solid forms thereof can be also prepared according to other methods apparent to those of skill in the art.
The compound of Formula I provided herein ns one chiral , and can exist as a mixture of enantiomers, e.g, a racemic mixture. This disclosure encompasses the use of stereomerically pure forms of such a compound, as weil as the use of mixtures of those forms. For example, mixtures comprising equal or unequal amounts of the enantiomers of the compound of Formula I provided herein may be used in methods and compositions disclosed herein. These isomers may be asymmetrically synthesized or resolved using standard techniques such as chiral s or chiral resolving agents. See, e. g.
, Jacques, I, et al, Enantz'omers, Racemai‘es and Resolutions (Wiley—lnterscience, New York, 1981); Wilen, S. H., et al, Tetrahedron 33:2725 (1977); Eliel, E. L., Stereochemz’srry ofCarbon Compounds (McGraW-Hill, NY, 1962); and Wilen, S. H., Tables ofResolving Agents and l tions p. 268 (EL. Eliel, Ed, Univ. of Notre Dame Press, Notre Dame, IN, 1972).
It should be noted that if there is a discrepancy between a depicted ure and a name given that structure, the depicted structure is to be accorded more . In addition, if the chemistry of a ure or a portion of a structure is not indicated with, for IO example, bold or dashed lines, the structure or portion of the structure is to be interpreted as encompassing all stereoisomers of the structure. .4 SECOND ACTIVE AGENTS A compound provided herein, ag. , the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co- crystal, clathrate, or polymorph thereof, can be combined with one or more other pharmacologically active compounds (“second active agents”) in methods and compositions provided herein. It is believed that n combinations work istically in the treatment of particular types of , and n diseases and ions associated with or characterized by undesired angiogenesis. The compound of Formula I provided herein can also work to alleviate adverse effects associated with certain second active agents, and some second active agents can be used to alleviate adverse s associated with the compound of Formula I provided herein.
One or more second active ingredients or agents can be used in the methods and compositions provided herein with the compound of Formula I provided herein. Second active agents can be large molecules (eg, proteins) or small les (ag, synthetic inorganic, organometallic, or organic molecules).
Examples of large molecule active agents include, but are not limited to, hematopoietic growth factors, cytokines, and monoclonal and polyclonal antibodies. In n embodiments, large molecule active agents are ical molecules, such as naturally occurring or artificially made proteins. Proteins that are particularly useful in this disclosure include proteins that stimulate the survival and/or proliferation of hematopoietic precursor cells and immunologically active poietic cells in vitro or in vivo. Others stimulate the division and differentiation of committed erythroid progenitors in cells in vitro or in vivo. Particular proteins include, but are not limited to: interleukins, such as IL-2 (including recombinant lL—ll (“rIL2”) and canarypox IL—2), IL—10, lL-12, and IL-18; interferons, such as interferon alfa-2a, interferon alfa—2b, interferon alfa-nl , interferon alfa-n3, interferon beta—l a, and interferon gamma-1 b; GM-CF and GM-CSF; and EPO.
Particular proteins that can be used in the methods and compositions of the U1 disclosure include, but are not limited to: tim, which is sold in the United States under the trade name NEUPOGEN® (Amgen, Thousand Oaks, CA); sargramostim, which is sold in the United States under the trade name LEUKINE® (lmmunex, Seattle, WA); and recombinant EPO, which is sold in the United States under the trade name EPGEN® (Amgen, Thousand Oaks, CA). inhibitors of ActRll receptors or activin—ActRlI inhibitors may be used in the methods and compositions provided herein. ActRIl receptors include ActRllA inhibitors and ActRllB inhibitors. Inhibitors of ActRlI receptors can be polypeptides comprising activin—binding domains of ActRll. In certain ments, the activin-binding domain comprising polypeptides are linked to an Fc portion of an antibody (i.e., a conjugate comprising an activin-binding domain comprising polypeptide of an ActRlI receptor and an Fc n of an antibody is generated). In certain embodiments, the activin-binding domain is linked to an Fc n of an dy Via a linker, e.g., a peptide linker.
Examples of such non-antibody proteins selected for activin or ActRllA g and methods for design and selection of the same are found in WO/2002/088171, WO/2006/055689, WO/2002/032925, 5/037989, US 2003/0133939, and US 2005/0238646, each of which is incorporated herein by reference in its entirety.
Recombinant and mutated forms of GM-CSF can be prepared as bed in US.
Patent Nos. 5,391,485; 5,393,870; and 5,229,496; the sure of each of which is incorporated herein by reference in its entirety. Recombinant and d forms of G-CSF can be prepared as described in US. Patent Nos. 4,810,643; 4,999,291; 5,528,823; and ,580,755; the disclosure of each of which is incorporated herein by reference in its entirety.
This disclosure asses the use of native, lly occurring, and recombinant proteins. The disciosure further asses mutants and tives (rag, modified forms) of lly ing proteins that exhibit, in viva, at least some of the pharmacological activity of the proteins upon which they are based. Examples of mutants include, but are not limited to, proteins that have one or more amino acid residues that differ from the corresponding residues in the naturally occurring forms of the proteins. Also encompassed by the term “mutants” are proteins that lack carbohydrate moieties normally present in their naturally occurring forms (cg, nonglycosylated forms). Examples of -43.. derivatives include, but are not limited to, pegylated tives and fusion proteins, such proteins formed by fusing IgGl or IgG3 to the protein or active portion of the protein of interest. See, e. g, Penichet, ML. and Morrison, S.L., J. Immunol. Methods 248:91—101 (2001).
Antibodies that can be used in combination with the compound of a I provided herein include monoclonal and polyclonal antibodies. es of antibodies include, but are not limited to, trastuzumab PTTN®), rituximab (RITUXAN®),bevacizumab (AVASTINTM), pertuzumab (OMNITARGTM), tositumomab (BEXXAR®), edrecolomab (PANOREX®), panitumumab and G250. The compound of Formula I provided herein can also be combined with or used in combination with anti- TNF-d antibodies.
Large molecule active agents may be administered in the form of anti—cancer vaccines. For example, vaccines that secrete, or cause the secretion of, cytokines such lL-2, SCF, CXCl4 (platelet factor 4), G—CSF, and GM-CSF can be used in the methods, pharmaceutical compositions, and kits of the disclosure. See, e. g., Emens, L.A., et al., Curr.
Opinion M01. Ther. 3(1):77-84 (2001).
Second active agents that are small molecules can also be used to alleviate adverse effects associated with the administration of the compound of Formula I provided herein. r, like some large molecules, many are believed to be capable of providing a synergistic effect when administered with (e. g. , before, after or simultaneously) the compound of Formula 1. Examples of small molecule second active agents include, but are not limited to, anti-cancer agents, antibiotics, immunosuppressive agents, and steroids.
Examples of anti-cancer agents include, but are not limited to: abraxane; ace—l l; acivicin; bicin; acodazole hydrochloride; acronine; esin; aldesleukin; altretamine; cin; ametantrone acetate; amrubicin; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; ide dimesylate; bizelesin; bleomycin sulfate; nar ; bropirimine; busulfan; ornycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefmgol; celecoxib (COX—2 inhibitor); chlorambucil; cirolemycin; cisplatin; bine; tol mesylate; cyciophosphamide; cytarabine; dacarbazine; dactinomycin; daunorubicin hydrochloride; decitabine; dexorrnaplatin; dezaguanine; dezaguanine mesylate; diaziquone; docetaxei; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hloride; trucin; enloplatin; enpromate; pidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; herceptin; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; iproplatin; irinotecan; irinotecan hydrochloride; lanreotide acetate; lapatinib; letrozoie; leuprolide e; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; rcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; paclitaxel; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; bazine hydrochloride; puromycin; purornycin hloride; pyrazofurin; riboprine; psin; safmgol; safingol hydrochloride; semustine; simtrazene; sate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; stem cell treatments such as FDA-001; streptonigrin; ozocin; sulofenur; talisomycin; tecogalan sodium; taxotere; tegafur; teloxantrone hydrochloride; temoporfln; teniposide; teroxirone; actone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone e; triciribine phosphate; trimetrexate; trimetrexate onate; triptorelin; tubulozole hydrochloride; uracil mustard; a; vapreotide; verteporfin; Vinhlastine sulfate; vincristine sulfate; ine; Vindesine sulfate; Vinepidine sulfate; cina’te sulfate; ix) U: Vinlcurosinc sulfate; Vinorelbine tartrate; Vinrosidine sulfate; Vinzolidine sulfate; vorozole; zeniplatin; zinostatin; and zorubicin hydrochloride.
Other anti-cancer drugs include, but are not limited to: 20-epi—l ,25 dihydroxyvitamin D3; S-ethynyluracil; erone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL—TK antagonists; altretamine; stine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozoie; andrographoiide; angiogenesis itors; nist D; antagonist G; antarelix; anti-dorsalizing morphogenetic n-1; antiandrogen; prostatic carcinoma; antiestrogen; antineopiaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; ic acid; ara—CDP-DL—PTBA; arginine deaminase; asulacrine; tane; atrirnustine; axinastatin l; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR/ABL antagonists; benzochlorins; benzoylstaurosporine; beta lactarn derivatives; lethine; betaclamycin B; betulinic acid; b-FGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; in B; cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; coliismycin A; collismycin B; combretastatin A4; tastatin analogue; conagenin; crambescidin 816; crisnatol; cryptophycin 8; cryptophyein A derivatives; curacin A; cyclopentanthraquinones; cycloplatam; cypemycin; cytarabine ate; cytolytic factor; atin; dacliximab; decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro-S-azacytidine; dihydrotaxol, 9—; dioxamycin; diphenyl ustine; docetaxel; docosanol; dolasetron; doxifluridine; doxorubicin; droloxifene; dronabinol; duocarmycin SA; n; ecomustine; edelfosine; edrecolomab; eflomithine; elemene; emitefur; epirubicin; epristeride; estramustine analogue; estrogen agonists; en antagonists; etanidazole; etoposide phosphate; exemestane; fadrozole; fazarabine; fenretinide; filgrastim; finasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostrieoin; fotern’ustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase tors; gemcitabine; hione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; sine; ilomastat; ib (ag, GLEEVEC@), imiquimod; immunostimulant peptides; insulin-like growth factor-1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol, 4~; iroplact; irsogladine; gazole; ohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin-N triacetate; lanreotide; ycin; lenograstirn; lentinan e; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon; lide+estrogen+progesterone; leuprorelin; sole; liarozole; linear polyarnine analogue; lipophilic disaccharide peptide; lipophilic platinum compounds; linamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; loxoribine; ecan; lutetiurn texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marirnastat; masoprocol; maspin; matrilysin inhibitors; matrix oproteinase tors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF tor; istone; miltefosine; mirimostim; azone; mitolactol; mitomycin analogues; mitonafide; mitotoxin fibroblast growth factor-saporin; mitoxantrone; motarotene; molgramostim;Erbitux, human nic gonadotrophin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; mustard anticancer agent; mycaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N—substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; oblimersen (GENASENSE‘fi’); 06—benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; paclitaxel; paclitaxel analogues; paclitaxel derivatives; palauamine; palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin; pazelliptine; pegaspargase; peldesine; an polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; yl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; rpine hydrochloride; pirarubicin; piritrexim; placetin A; placetin B; plasminogen activator inhibitor; platinum complex; platinum nds; platinum-triamine complex; r sodium; porfiromycin; sone; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras famesyl protein transferase inhibitors; ras inhibitors; rag-GAP tor; reteliiptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; Rll retinamide; rohitukine; romurtide; roquinimex; rubiginone Bl; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; sargramostim; Sdi l cs; semustine; senescence derived inhibitor 1; sense oligonucleotides; signal transduction tors; sizofiran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonennin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin l; squalamine; stipiamide; stromelysin inhibitors; sulfinosine; ctive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; taiiimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafiir; teilurapyrylium; teiomerase tors; rfin; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; raline; opoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; d stimulating hormone; tin ethyl etiopurpurin; tirapazamine; cene bichloride; topsentin; toremifene; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; eride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex; urogenital sinus—derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; sol; veramine; verdins; verteporfin; vinorelbinej, vinxaltine; vitaxin; vorozole; zanoterone; atin; zilascorb; and zinostatin stimalamer.
Specific second active agents include, but are not d to, rsen (GENASENSE®), remicade, docetaxel, xib, melphalan, dexamethasone IO (DECADRON‘EV), steroids, gemcitabine, cisplatinum, temozolomide, etoposide, cyclophosphamide, temodar, carboplatin, procarbazine, gliadel, fen, topoteean, methotrexate, ARISA®, taxol, taxotere, fluorouracil, leucovorin, irinotecan, xeloda, CPT- l l, interferon alpha, pegylated interferon alpha (ag.
, PEG INTRON-A), tabine, cisplatin, thiotepa, fludarabine, carboplatin, liposomal daunorubicin, cytarabine, doxetaxol, pacilitaxel, vinblastine, IL-2, GM~CSF, dacarbazine, vinorelbine, zoledronic acid, palmitronate, biaxin, busulphan, prednisone, bisphosphonate, arsenic trioxide, vincristine, doxorubicin (DOXIL®), paclitaxel, lovir, adriamycin, estramustine sodium phosphate (EMCYT®), sulindac, and etoposide. .5 BIOMARKERS 2O Provided herein are methods relating to the use of mRNAs or proteins as kers to ascertain the effectiveness of cancer therapy. mRNA or protein levels can be used to determine whether a particular agent is likely to be successful in the treatment of a specific type of cancer, e.g., non—Hodgkin’s ma.
A biological marker or “biomarker” is a nce whose detection tes ular biological state, such as, for example, the presence of cancer. In some embodiments, biomarkers can either be determined individually, or several biomarkers can be measured simultaneously.
In some embodiments, a “biomarker” indicates a change in the level ofmRNA expression that may correlate with the risk or progression of a disease, or with the susceptibility of the disease to a given treatment. In some embodiments, the biomarker is a nucleic acid, such as a mRNA or cDNA.
In additional embodiments, a “biomarker” indicates a change in the level of polypeptide or protein expression that may correlate with the risk, susceptibility to treatment, or progression of a disease. In some embodiments, the biomarker can be a _ 48 _ polypeptide or protein, or a fragment thereof. The relative level of specific proteins can be determined by methods known in the art. For example, antibody based methods, such as an immunoblot, enzyme-linked immunosorbent assay (ELISA), or other methods can be used. .6 METHODS OF TREATMENT AND PREVENTION in one embodiment, provided herein is a method of treating and preventing , which comprises administering to a patient a compound provided herein, e.g, the compound of Formula l, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, stal, clathrate, or polymorph it) In another embodiment, provided herein is method of managing cancer, which comprises administering to a patient a compound provided herein, 6. g, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a ceutically acceptable salt, solvate, hydrate, co—crystal, ate, or polymorph thereof.
Provided herein are methods of treating or managing lymphoma, particularly non- Hodgkin’s lymphoma. In some embodiments, provided herein are methods for the treatment or management of non-Hodgkin‘s ma (NHL), including but not limited to, diffuse large B-cell lymphoma (DLBCL), using prognostic factors.
Also provided herein are methods of ng patients who have been previously treated for cancer but are non~responsive to standard therapies, as well as those who have not previously been treated. The invention also encompasses methods of treating patients regardless of patient’s age, gh some diseases or disorders are more common in certain age groups. The invention r asses methods of ng patients who have undergone surgery in an attempt to treat the disease or condition at issue, as well as those who have not. e patients with cancer have heterogeneous clinical manifestations and g clinical outcomes, the treatment given to a patient may vary, depending on his/her prognosis. The skilled clinician will be able to readily determine without undue experimentation specific secondary agents, types of surgery, and types of non—drug based rd therapy that can be effectively used to treat an individual patient with cancer.
As used herein, the term “cancer” includes, but is not limited to, solid tumors and blood born tumors. The term r” refers to disease of skin tissues, organs, blood, and vessels, including, but not d to, cancers of the bladder, bone, blood, brain, , cervix, chest, colon, etrinm, esophagus, eye, head, kidney, liver, lymph nodes, lung, mouth, neck, ovaries, pancreas, prostate, rectum, stomach, testis, throat, and .
Specific cancers include, but are not limited to, advanced malignancy, amyloidosis, lastoma, meningioma, hemangiopericytoma, multiple brain metastase, glioblastoma multiforms, glioblastoma, brain stem , poor prognosis malignant brain tumor, malignant glioma, recurrent malignant giolma, anaplastic astrocytoma, anaplastic oligodendroglioma, neuroendocrine tumor, rectal adenocarcinoma, Dukes C & D colorectal cancer, unresectable colorectal carcinoma, atic hepatocellular carcinoma, Kaposi’s sarcoma, karotype acute myeloblastic leukemia, Hodgkin’s lymphoma, non—Hodgkin’s ma, cutaneous T-Cell lymphoma, cutaneous B-Cell lymphoma, diffuse large B-Cell lymphoma, low grade ular lymphoma, malignant melanoma, malignant mesothelioma, malignant pleural effusion mesothelioma me, neal carcinoma, papillary serous carcinoma, gynecologic sarcoma, soft tissue sarcoma, scleroderma, cutaneous yasculitis, Langerhans cell histiocytosis, leiomyosarcoma, fibrodysplasia ossificans progressive, hormone refractory prostate cancer, resected high-risk soft tissue sarcoma, unrescectable hepatocellular oma, Waldenstrom’s macroglobulinemia, smoldering myeloma, indolent a, fallopian tube cancer, androgen independent prostate , androgen dependent stage IV non—metastatic prostate cancer, hormone-insensitive prostate cancer, chemotherapy—insensitive prostate cancer, papillary thyroid carcinoma, follicular thyroid carcinoma, medullary thyroid oma, and leiomyoma In certain embodiments, the cancer is a blood borne tumor. In certain embodiments, the blood borne tumor is metastatic. In certain embodiments, the blood borne tumor is drug ant. In certain embodiments, the cancer is myeloma or lymphoma.
In certain embodiments, the cancer is a solid tumor. In certain embodiments, the solid tumor is metastatic. In certain embodiments, the solid tumor is drug-resistant. In certain embodiments, the solid tumor is hepatocellular carcinoma, prostate cancer, ovarian cancer, or astoma.
In certain embodiments, a therapeutically or lactically effective amount of the compound is from about 0.005 to about 1,000 mg per day, from about 0.01 to about 500 mg per day, from about 0.01 to about 250 mg per day, from about 0.01 to about 100 mg per day, from about 0.1 to about 100 mg per day, from about 0.5 to about 100 mg per day, from about 1 to about 100 mg per day, from about 0.01 to about 50 mg per day, from about 0.1 to about 50 mg per day, from about 0.5 to about 50 mg per day, from about 1 to about 50 mg per day, from about 0.02 to about 25 mg per day, or from about 0.05 to about 10 mg per day.
In certain ment, a therapeutically or prophylactically effective amount is from about 0.005 to about 1,000 mg per day, from about 0.01 to about 500 mg per day, from about 0.01 to about 250 mg per day, from about 0.01 to about 100 mg per day, from about 0.1 to about 100 mg per day, from about 0.5 to about 100 mg per day, from about 1 to about 100 mg per day, from about 0.01 to about 50 mg per day, from about 0.1 to about 50 mg per day, from about 0.5 to about 50 mg per day, from about 1 to about 50 mg per day, from about 0.02 to about 25 mg per day, or from about 0.05 to about 10 mg every other day.
In certain embodiments, the therapeutically or prophylactically effective amount is about 1, about 2, about 5, about 10, about 15, about 20, about 25, about 30, about 40, about 45, about 50, about 60, about 70, about 80, about 90, about 100, or about 150 mg per day.
In one embodiment, the recommended daily dose range of the nd of Formula I for the conditions described herein lie within the range of from about 0.5 mg to about 50 mg per day, preferably given as a single once-a-day dose, or in divided doses throughout a day. In some embodiments, the dosage ranges from about 1 mg to about 50 mg per day. In other embodiments, the dosage ranges from about 0.5 to about 5 mg per day. Specific doses per day include 1, 2, 3, 4, 5, 6, 7, 8, 9,10,11,12,13,14,15,l6,17,18,19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 or 50 mg per day.
In a specific embodiment, the recommended ng dosage may be 0.5, 1, 2, 3, 4, 5, , 15, 20, 25 or 50 mg per day. In another embodiment, the recommended starting dosage may be 0.5, 1, 2, 3, 4, or 5 mg per day. The dose may be escalated to 15, 20, 25, 30, 35, 40, 45 and 50 mg/day. In a specific ment, the compound can be administered in an amount of about 25 mg/day to patients with NHL (6g, DLBCL). In a particular embodiment, the compound can be stered in an amount of about 10 mg/day to patients with NHL (e.g., .
In certain embodiments, the therapeutically or prophylactical 1y effective amount is from about 0.001 to about 100 rng/kg/day, from about 0.01 to about 50 mg/kg/day, from about 0.01 to about 25 mg.’l<g/day, from about 0.01 to about 10 mg/kg/day, from about 0.01 to about 9 mgr’kg/day, 0.01 to about 8 mg/kg/day, from about 0.01 to about 7 mg/‘kgfday, from about 0.01 to about 6 mgfkg/day, from about 0.01 to about 5 mgfikg/day, from about 0.01 to about 4 mg/kg/day, from about 0.01 to about 3 mg/kg/day, from about 0.01 to about 2 mgi’kg/day, or from about 0.01 to about 1 mg/kg/day.
The stered dose can also be expressed in units other than mg’kg/day. For example, doses for parenteral administration can be expressed as mgi’mzfday. One of ordinary skit} in the art wouid readily know how to convert doses from day to mg/mZ/day to given either the height or weight of a subject or both (see, wwfda.gov/cder/cancer/animalframehtm). For example, a dose of 1 mg/kg/day for a 65 kg human is approximately equal to 38 mg/mz/day.
In n embodiments, the amount of the compound administered is sufficient to provide a plasma concentration of the compound at steady state, ranging from about 0.001 to about 500 uM, about 0.002 to about 200 uM, about 0.005 to about 100 uM, about 0.01 to about 50 MA, from about 1 to about 50 uM, about 0.02 to about 25 nM. from about 0.05 to about 20 MA, from about 0.1 to about 20 uM, from about 0.5 to about 20 uM, or from about 1 to about 20 nM.
In other embodiments, the amount of the compound administered is sufficient to e a plasma concentration of the compound at steady state, ranging from about 5 to about 100 nM, about 5 to about 50 nM, about 10 to about 100 nM, about 10 to about 50 nM or from about 50 to about 100 nM.
As used herein, the term “plasma concentration at steady state” is the concentration reached after a period of administration of a compound provided herein, 3. g, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof. Once steady state is reached, there are minor peaks and troughs on the time dependent curve of the plasma concentration of the compound.
In certain embodiments, the amount of the compound administered is sufficient to provide a maximum plasma tration (peak concentration) of the compound, ranging from about 0.001 to about 500 uM, about 0.002 to about 200 nM, about 0.005 to about 100 uM, about 0.01 to about 50 nM, from about 1 to about 50 MA, about 0.02 to about 25 uM, from about 0.05 to about 20 MM, from about 0.1 to about 20 uM, from about 0.5 to about 20 uM,or from about 1 to about 20 nM.
In certain embodiments, the amount of the nd administered is sufficient to provide a minimum plasma concentration (trough tration) of the nd, ranging from about 0.001 to about 500 nM, about 0.002 to about 200 uM, about 0.005 to about 100 MM, about 0.01 to about 50 nM, from about 1 to about 50 141%, about 0.01 to about 25 nM, from about 0.01 to about 20 uM, from about 0.02 to about 20 0M, from about 0.02 to about 20 uM, or from about 0.01 to about 20 nM. 1n certain embodiments, the amount of the compound administered is sufficient to provide an area under the curve (AUC) of the compound, ranging from about 100 to about 100,000 ng*hr:"mL, from about 1,000 to about 50,000 ng*hr/mL, from about 5,000 to about ,000 ng*hr/mL, or from about 5,000 to about 10,000 ng*hrme.
In certain embodiments, the patient to be treated with one of the methods ed herein has not been treated with anticancer therapy prior to the administration of the compound of, Formula I. In certain embodiments, the patient to be d with one of the methods provided herein has been treated with anticancer therapy prior to the administration of the compound of Formula I. In certain ments, the patient to be treated with one of the methods provided herein has developed drug resistance to the anticancer y.
The methods provided herein encompass treating a patient regardless of t’s in certain age groups. Further age, although some diseases or disorders are more common provided herein is a method for treating a patient who has undergone surgery in an attempt to treat the disease or condition at issue, as well in one who has not. Because the ts with cancer have heterogeneous clinical manifestations and varying clinical es, the treatment given to a particular subject may vary, depending on his/her prognosis. skilled clinician will be able to readily determine without undue experimentation, specific secondary agents, types of surgery, and types of non-drug based standard therapy that can be effectively used to treat an individual subject with cancer.
Depending on the e to be treated and the subj ect’s condition, the nd of Formula I, or an enantiomer or a mixture of enantiomers thereof; or a pharmaceutically acceptable salt, solvate, e, co—crystal, clathrate, or rph thereof, may be administered by oral, parenteral (cg, intramuscular, intraperitoneal, enous, CIV, intracistemal injection or infusion, subcutaneous injection, or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (cg, transdermal or local) routes of administration.
The compound of Formula I, or an enantiomer or a mixture of omers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, may be formulated, alone or together, in suitable dosage unit with pharmaceutically acceptable excipients, rs, adjuvants and vehicles, riate for each route of administration.
In one embodiment, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof; or a pharmaceutically acceptable salt, soivate, hydrate, st I, clathrate, or polymorph thereof, is administered orally. in another embodiment, the compound of Formuia i, or an enantiomer or a mixture of enantiomers f; or a pharmaceutically acceptable salt, solvate, hydrate, eo—crystal, clathrate, or polymorph thereof, is administered parenterally. In yet another embodiment, the compound of a I, or an enantiomer or a mixture of enantiomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is administered intravenously.
The compound of Formula I, or an enantiomer or a mixture of enantiomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or rph thereof, can be delivered as a single dose such as, eg, a single bolus injection, or oral tablets or pills; or over time, such as, cg, continuous infusion over time or divided bolus doses over time. The compound can be administered edly if necessary, for example, until the patient experiences stable disease or regression, or until the patient experiences disease progression or unacceptable ty. For example, stable disease for solid tumors generally means that the perpendicular diameter of measurable s has not increased by % or more from the last measurement. Response Evaluation ia in Solid Tumors (RECIST) Guidelines, Journal ofthe National Cancer Institute 92(3): 205-216 (2000).
Stable disease or lack thereof is ined by methods known in the art such as evaluation of patient symptoms, al examination, visualization of the tumor that has been imaged using X—ray, CAT, PET, or MRI scan and other commonly ed evaluation modalities.
The compound of Formula I, or an enantiomer or a mixture of enantiomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, can be administered once daily (QD), or divided into multiple daily doses such as twice daily (BID), three times daily (TID), and four times daily (QID). In addition, the administration can be continuous (i.e., daily for consecutive days or every day), intermittent, e.g., in cycles (i.e., including days, weeks, or months of rest without drug). As used herein, the term “daily” is intended to mean that a eutic compound, such as the compound of Formula I, is administered once or more than once each day, for e, for a period of time. The term “continuous” is intended to mean that a therapeutic nd, such as the compound of Formula I, is administered daily for an uninterrupted period of at least 10 days to 52 weeks. The term “intermittent” or “intermittently” as used herein is intended to mean stopping and starting at either regular or irregular intervals. For e, intermittent administration of the compound of Formula I is administration for one to six days per week, administration in cycles (ag, daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week), or administration on alternate days.
The term “cycling” as used herein is intended to mean that a eutic compound, such as the compound of Formula I, is administered daily or continuously but with a rest period.
In some embodiments, the ncy of administration is in the range of about a daily dose to about a monthly dose. In certain embodiments, administration is once a day, -54.. twice a day, three times a day, four times a day, once every other day, twice a week, once three weeks, or once every four weeks. In every week, once every two weeks, once every enantiomer or a mixture of enantiomers one embodiment, the compound of Formula I, or an thereof; or a phannaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or U1 polymorph thereof, is administered once a day. In r embodiment, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof; or a pharmaceutically administered acceptable salt, solvate, hydrate, co—crystal, cIathrate, or polymorph thereof, is twice a day. In yet another embodiment, the compound of Formula I, or an enantiomer or a mixture of omers thereof; or a pharmaceutically acceptable salt, solvate, e, co— In still another l0 l, clathrate, or polymorph thereof, is administered three times a day. of enantiomers embodiment, the compound of Formula I, or an enantiomer or a mixture thereof; or a pharmaceutically acceptable salt, solvate, hydrate, stal, clathrate, or polymorph thereof, is administered four times a day. mixture In certain ments, the compound of Formula I, or an enantiomer or a of enantiomers thereof; or a pharmaceutically acceptable salt, solyate, hydrate, co-crystal, six months, clathrate, or polymorph thereof, is administered once per day from one day to week to three from one week to three months, from one week to four weeks, from one weeks, or from one week to two weeks. In certain embodiments, the compound of a administered once per day for I, or a pharmaceutically able salt or solvate thereof, is In one embodiment, the compound of one week, two weeks, three weeks, or four weeks.
Formula I, or an enantiomer or a mixture of enantiomers thereof; or a ceutically administered acceptable salt, solvate, hydrate, co-crystal, clathrate, or rph f, is once per day for one week. In another embodiment, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is administered once per day for two weeks. In yet another embodiment, the compound of Formula I, or an enantiomer solvate, hydrate, or a e of omers thereof; or a ceutically acceptable salt, weeks. In cry-crystal, clathrate, or polymorph thereof, is administered once per day for three mixture of stili another embodiment, the nd of Formula I, or an omer or a enantiomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is administered once per day for four weeks. .6.1 COMBINATION THERAPY WITH A SECOND ACTIVE AGENT The compound of Formula I, or an enantiomer or a mixture of enantiomers thereof; or a pharmaceutically acceptable salt, e, hydrate, co—crystal, clathrate, or polymorph thereof, can also be combined or used in combination with other therapeutic agents useful in the treatment r prevention of cancer described herein.
In one embodiment, provided herein is a method of ng, preventing, or ng cancer, comprising administering to a patient 3-(5-amino~2—methyl-4~oxo-4H- quinazolinyl)-piperidine-2,6-dione, or an enantiomer or a mixture ofenantiomers thereof; or a phannaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof; in combination with one or more second active agents, and optionally in combination with ion therapy, blood transfusions, or surgery. Examples of second active agents are disclosed herein (see, e.3, section 5.3).
As used , the term “in combination” includes the use of more than one therapy (e.g, one or more prophylactic and/or therapeutic agents). However, the use of the term “in combination” does not restrict the order in which therapies (e.g., prophylactic and/or therapeutic agents) are administered to a patient with a disease or disorder. A first y (e.g., a prophylactic or therapeutic agent such as a compound provided herein, a compound provided herein, 6.g, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a ceutically acceptable salt, solvate, hydrate, co~crystal, clathrate, or polymorph f) can be administered prior to (e.g., 5 minutes, 15 minutes, minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or uent to (cg, 5 minutes, minutes, 30 minutes, 45 s, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the stration of a second therapy (cg, a prophylactic or therapeutic agent) to the subject. Triple therapy is also contemplated herein.
Administration of the compound of Formula I and one or more second active agents to a patient can occur simultaneously or sequentialiy by the same or different routes of administration. The suitability of a particular route of administration employed for a particuiar active agent will depend on the active agent itself (rag. , whether it can be administered orally without decomposing prior to ng the blood stream) and the cancer being treated. - 56 _ The route of administration of the nd of Formula I is independent of route of administration of a second therapy. In one embodiment, the compound of Formula I is administered orally. In another ment, the compound of Formula I is administered intravenously. Thus, in accordance with these embodiments, the nd of Formula I is administered orally or intravenously, and the second therapy can be administered orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermaily, sublingually, intramuscularly, ly, uccally, intranasally, liposomally, Via inhalation, vaginally, intraoccularly, via local delivery by catheter or stent, subcutaneously, intraadiposally, rticularly, intrathecally, or in a slow release dosage IO form. In one embodiment, the compound of Formula I and a second therapy are administered by the same mode of administration, orally or by IV. In another ment, the nd of Formula I is administered by one mode of administration, e.g., by IV, whereas the second agent (an anticancer agent) is administered by r mode administration, e.g., orally.
In one ment, the second active agent is administered intravenously or subcutaneously and once or twice daily in an amount of from about 1 to about 1000 mg, 50 to about from about 5 to about 500 mg, from about 10 to about 350 mg, or from about 200 mg. The specific amount of the second active agent will depend on the specific agent and the used, the type of disease being treated or managed, the severity and stage of disease, additional active amount of the compound of Formula I provided herein and any optional agents concurrently administered to the patient. In certain embodiments, the second active irinotecan, agent is oblimersen (GENASENSE‘A’i), GM—CSF, G-CSF, SCF, EPO, taxotere, dacarbazine, transretinoic acid, topotecan, pentoxifylline, ciprofloxacin, dexamethasone, vincristine, doxorubicin, COX—2 inhibitor, IL2, 1L8, ILIS, IFN, Ara-C, vinorelbine, or a combination thereof.
In certain embodiments, GM-CSF, G-CSF, SCF or EFO is administered subcutaneously during about five days in a four or six week cycle in an amount ranging from about I to about 750 mgfmzz’day, from about 25 to about 500 mgr’mZ/day, from about 50 to about 250 mgr'mZ/day, or from about 50 to about 200 mg/mzi’day. In certain ments, GM—CSF may be administered in an amount of from about 60 to about 500 meg/mt2 intravenously over 2 hours or from about 5 to about 12 mcgj’mzx’day aneously. In certain embodiments, G—CSF may be administered subcutaneously in an amount of about I meg/kgfday initially and can be adjusted depending on rise of total granulocyte counts. The maintenance dose of G-CSF may be administered in an amount of about 300 (in smaller patients) or 480 mcg subcutaneously. In certain ments, EPO maybe administered subcutaneously in an amount of 10,000 Unit 3 times per week.
In certain embodiments, a compound provided herein, ag, the compound of U: Formula I, or an enantiomer or a mixture of enantiomers f, or a pharmaceutically acceptable salt, solvate, e, co-crystal, clathrate, or polymorph thereof, is administered with lan and dexamethasone to patients with amyloidosis. In certain embodiments, a compound provided herein, ag, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a ceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, and steroids can be administered to patients with amyloidosis.
In certain embodiments, a compound provided , e. g, the nd of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, is administered with gemcitabine and cisplatinum to patients with locally advanced or metastatic transitional cell bladder cancer.
In certain ments, a compound provided herein, e.g., the compound of Formula I, or an omer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, e, hydrate, co-crystal, clathrate, or polymorph thereof, is administered in ation with a second active ingredient as follows: temozolomide to pediatric patients with ed or ssive brain tumors or recurrent neuroblastoma; celecoxib, etoposide and cyclophosphamide for relapsed or progressive CNS cancer; temodar to patients with recurrent or progressive meningioma, malignant meningioma, hemangiopericytoma, multiple brain metastases, sed brain tumors, or newly diagnosed glioblastoma multiforms; ecan to patients with recurrent glioblastoma; latin to pediatric patients with brain stem glioma‘, procarhazinc to pediatric patients with progressive maiignant gliomas; cyclophosphamide to patients with poor prognosis malignant brain tumors, newly diagnosed or ent glioblastoma multiforms; Gliadel® for high grade recurrent malignant gliomas; temozolomide and tamoxifen for anaplastic astrocytorna; or can for s, glioblastoma, anaplastic astrocytoma or anaplastic oligodendroglioma.
In certain embodiments, a compound provided herein, eg, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically is administered acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, aromatase with methotrexate, cyclophosphamide, taxane, abraxane, lapatinib, herceptin, and/or PLX3397 inhibitors, ive en modulators, estrogen receptor antagonists, (Plexxikon) to patients with metastatic breast cancer.
In certain embodiments, a compound provided herein, e.g., the compound Formula I, or an enantiomer or a mixture of omers thereof, or a pharmaceutically is administered acceptable salt, solvate, hydrate, (to-crystal, ate, or polymorph thereof, with temozolomide to patients with neuroendocrine tumors.
In certain embodiments, a compound provided herein, e.g., the nd a I, or an enantiomer or a mixture of enantiomers thereof, or a ceutically is stered acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, with gemcitabine to patients with recurrent or metastatic head or neck .
In certain embodiments, a compound provided herein, eg, the compound a I, or an enantiomer or a e of enantiomers f, or a pharmaceutically stered acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is with gemcitabine to patients with pancreatic cancer.
In n embodiments, a compound provided herein, e.g, the compound Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically is administered acceptable salt, solvate, e, co—crystal, clathrate, or polymorph thereof, and/or taxotere. to patients with colon cancer in combination with ARISA®, avastatin, taxol, In certain embodiments, a compound provided herein, ag, the compound Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically administered able salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is colorectal cancer with capecitabine andi’or PLX4032 (Plexxikon) to patients with refractory in colon or rectal or patients who fail first line therapy or have poor performance adenocarcinoma.
In certain embodiments, a compound provided herein, tag, the compound Formula I, or an enantiomer or a mixture of enantiomers thereof, or a ceutically administered acceptable salt, solvate, hydrate, co—crystal, ciathrate, or polymorph f, is with Dukes C & D in combination with fluorouracil, leucovorin, and irinotecan to patients colorectal colorectal cancer or to ts who have been previously treated for metastatic cancer.
In certain embodiments, a compound provided herein, e.g., the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is administered to patients with refractory colorectal cancer in combination with capecitabine, xeloda, and/or CPT-l I.
In certain embodiments, a compound provided herein, ag. the compound of Formula I, or an enantiomer or a mixture of enantiomers f, or a pharmaceuticaliy acceptable salt, solvate, hydrate, co~crystal, clathrate, or polymorph thereof, is administered with capecitabine and irinotecan to patients with refractory colorectal cancer or to patients with unresectable or metastatic ctal carcinoma.
In certain embodiments, a compound provided herein, rag, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceuticaliy able salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is administered alone or in combination with interferon alpha or capecitabine to patients with unresectable IS or metastatic cellular oma; or with cisplatin and thiotepa to patients with primary or metastatic liver cancer.
In certain embodiments, a compound ed herein, rag, the compound of a I, or an enantiomer or a mixture of omers thereof, or a pharmaceutically acceptable salt, solvate, e, co—crystal, clathrate, or polymorph thereof, is administered in combination with pegylated interferon alpha to ts with Kaposi’s sarcoma.
In certain embodiments, a compound provided herein, eg, the compound of Formula I, or an omer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, is administered in combination with fludarabine, carboplatin, and/or topotecan to patients with refractory or relapsed or high-risk acuted myelogenous leukemia.
In certain embodiments, a compound ed herein, ag, the compound of Formula I, or an enantiomer or a mixture of omers thereof, or a pharmaceuticaiiy acceptable salt, solvate, e, co-crystal, clathrate, or polymorph thereof, is administered in combination with liposomal daunorubicin, can and/0r cytarabine to patients with unfavorable karotype acute myeiobiastic leukemia.
In certain embodiments, a compound provided herein, 8.53., the compound of a I, or an enantiomer or a e of enantiomers thereof, or a pharmaceuticaliy acceptable salt, soivate, hydrate, stal, clathrate, or polymorph thereof, is administered —60- irinotecan to patients in combination with gemcitabine, abraxane, erlotinib, geftinib, and/or with non-small cell lung .
In certain ments, a compound provided herein, 9.g, the compound Formula I, or an omer or a mixture of enantiomers thereof, or a pharmaceutically is administered acceptable salt, solvate, hydrate, co~crystal, clathrate, or polymorph thereof, non-small cell lung cancer. in ation with carboplatin and irinotecan to patients with In n embodiments, a compound provided herein, e.g., the compound Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically is administered acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, been previously treated IO with doxetaxol to patients with non-small cell lung cancer who have with carboNF’ 16 and radiotherapy. the compound of In certain embodiments, a compound provided herein, e.g., Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically is administered acceptable salt, e, hydrate, co—crystal, clathrate, or polymorph thereof, with carboplatin, in combination with carboplatin and/or taxotere, or in combination taxel and/or thoracic radiotherapy to patients with all cell lung cancer.
In certain embodiments, a nd provided herein, 8. g., the compound Formula I, or an omer or a mixture of enantiomers f, or a pharmaceutically is administered acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, non—small cell lung cancer. in ation with taxotere to patients with stage IIIB or IV In certain embodiments, a compound provided herein, e.g, the compound Formula I, or an omer or a mixture of enantiomers thereof, or a pharmaceutically is administered acceptable salt, solvate, e, co—crystal, clathrate, or polymorph thereof, cell lung cancer. in combination with oblimersen (Genasense®) to patients with small In certain embodiments, a compound provided herein, e.g, the compound a I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically is administered acceptabie salt, soivate, hydrate, co—crystal, ciathrate, or polymorph thereof, in combination with ABT~737 (Abbott Laboratories) andfor obatoclax (GXI 5-070) patients with lymphoma and other blood cancers.
In certain embodiments, a nd provided , ag, the compound Formnia I, or an enantiomer or a mixture of enantiomers f, or a ceuticaily is administered acceptabie salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, vinblastine or fludarabine to alone or in combination with a second active ingredient such as ~61- ts with various types of lymphoma, including, but not limited to, Hodgkin’s ma, non—Hodgkin’s lymphoma, cutaneous T—Cell lymphoma, cutaneous B-Cell lymphoma, diffuse large B-Cell lymphoma or relapsed or refractory low grade follicular lymphoma.
In certain embodiments, a compound ed herein, e. g, the compound of a I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is administered in combination with taxotere, lL-2, IFN, GM-CSF, PLX4032 (Plexxikon) and/or dacarbazine to patients with various types or stages of melanoma.
IO In n embodiments, a compound provided herein, e.g., the compound of Formula I, or an enantiomer or a e of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is administered alone or in combination with vinorelbine to patients with malignant mesothelioma, or stage IIIB non-small cell lung cancer with pleural implants or ant pleural effusion mesothelioma syndrome.
In certain embodiments, a compound provided herein, e.g, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, is administered to patients with various types or stages of multiple myeloma in ation with dexamethasone, zoledronic acid, palmitronate, GM—CSF, biaxin, vinblastine, melphalan, busulphan, cyclophosphamide, IFN, palmidronate, prednisone, bisphosphonate, celecoxib, arsenic trioxide, PEG INTRON-A, vincristine, or a combination f.
In certain embodiments, a compound provided herein, e.g., the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a ceutically acceptable salt, solvate, e, co-crystal, clathrate, or polymorph thereof, is administered to ts with relapsed or refractory multiple a in combination with doxorubicin (Doxilg), Vincristine and/or dexamethasone (Decadron®).
In certain embodiments, a compound provided herein, rag, the compound of Forrnula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, ciathrate, or ponmorph thereof, is administered to patients with various types or stages of ovarian cancer such as peritoneal carcinoma, papillary serous carcinoma, refractory ovarian cancer or ent n cancer, in combination with taxol, carboplatin, doxorubicin, gemcitabine, cisplatin, xeloda, paclitaxel, dexamethasone, or a combination thereof.
In certain embodiments, a compound provided herein, 6.g, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, e, hydrate, co-crystal, clathrate, or polymorph thereof, is administered to patients with s types or stages of prostate cancer, in combination with xeloda, FU/LV, gemcitabine, ecan plus gemcitabine, hosphamide, vineristine, dexamethasone, GM-CSF, celecoxib, taxotere, ganciclovir, paclitaxel, adriamycin, docetaxel, estramustine, Emcyt, denderon or a combination thereof.
In certain embodiments, a compound ed herein, a.g, the compound of a I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, is administered to patients with various types or stages of renal cell cancer, in combination with tabine, IFN, tamoxifen, lL—Z, , Celebrex®, or a combination thereof.
In certain embodiments, a compound provided herein, e.g., the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co~crystal, clathrate, or polymorph thereof, is administered to patients with various types or stages of gynecologic, uterus or soft tissue sarcoma cancer in combination with IFN, a COX-2 inhibitor such as ex®, and/or sulindac.
In n embodiments, a compound provided herein, e.g., the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, ate, or polymorph thereof, is administered to patients with various types or stages of solid tumors in combination with celebrex, etoposide, cyclophosphamide, docetaxel, abine, IFN, tamoxifen, IL—2, GM-CSF, or a combination thereof.
In certain embodiments, a compound provided , ag, the compound of Formula I, or an omer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co~crystal, clathrate, or polymorph thereof, is administered to patients with derma or cutaneous vascuiitis in combination with celebrex, etoposide, cyclophosphamide, docetaxel, apecitabine, IFN, tamoxifen, IL~2, GM-CSF, or a ation thereof.
Aiso encompassed herein is a method of increasing the dosage of an anti—cancer drug or agent that can be safely and effectively administered to a patient, which comprises administering to the patient (6. g. , a human) or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, e, hydrate, co-crystal, clathrate, or polymorph thereof. Patients that can benefit by this method are those likely to suffer from an adverse effect associated with anti—cancer drugs for treating a specific cancer of the skin, subcutaneous tissue, lymph nodes, brain, lung, liver, bone, intestine, colon, heart, pancreas, adrenal, kidney, prostate, breast, colorectal, or combinations thereof. The administration of a compound provided herein, ag, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, crystal, clathrate, or polymorph thereof, alleviates or reduces adverse s which are of such ty that it would otherwise limit the amount of anti-cancer drug.
In one embodiment, a compound provided herein, cg, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a ceutically acceptable salt, solvate, hydrate, stal, clathrate, or polymorph thereof, is administered orally and daily in an amount ranging from about 0.1 to about 150 mg, from about I to about 50 mg, or from about 2 to about 25 mg, prior to, during, or after the occurrence of the adverse effect ated with the administration of an anti—cancer drug to a t. In certain embodiments, a compound provided , e.g, the compound of Formula I, or an enantiomer or a mixture of enantiomers f, or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, is administered in combination with c agents such as heparin, aspirin, coumadin, or G—CSF to avoid e s that are associated with anti—cancer drugs such as but not limited to neutropenia thrombocytopenia.
In one embodiment, a compound provided herein, eg, the compound of Formula I, or an omer or a mixture of enantiomers thereof, or a ceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, is stered to patients with diseases and disorders associated with or characterized by, undesired angiogenesis in combination with additional active ingredients, including, but not limited to, anti~cancer drugs, anti—inflammatories, antihistamines, antibiotics, and steroids.
In another embodiment, encompassed herein is a method of treating, preventing andfor managing cancer, which comprises administering the nd of Formula l, or an enantiomer or a mixture of omers thereof, or a ceutically acceptable salt, solvate, hydrate, co-crystal, ciathrate, or polymorph thereof, in conjunction with (eg. before, during, or after) conventional therapy including, but not limited to, surgery, immunotherapy, biological therapy, radiation therapy, or other non-drug based therapy tly used to treat, prevent or manage cancer. The combined use of the compound provided herein and conventional therapy may provide a unique treatment regimen that is unexpectedly effective in certain patients. Without being limited by theory, it is believed that the nd of Formula I may provide ve or synergistic s when given concurrently with conventional therapy.
As discussed ere herein, encompassed herein is a method of reducing, treating and/or preventing adverse or undesired effects associated with tional y including, but not limited to, surgery, herapy, radiation therapy, hormonal therapy, biological therapy and immunotherapy. A compound provided , e.g., the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, and other active ient can be administered to a patient prior to, during, or after the occurrence of the adverse effect associated with conventional therapy.
In one embodiment, the compound of Formula I can be administered in an amount g from about 0.1 to about 150 mg, from about 1 to about 25 mg, or from about 2 to about 10 mg orally and daily alone, or in combination with a second active agent sed herein (see, e. g. section 4.3), prior to, during, or after the use of conventional therapy.
In certain embodiments, a compound provided herein, e.g., the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, e, co~crystal, clathrate, or polymorph thereof, and doxetaxol are administered to patients with non-small cell lung cancer who were previously treated with carbo/VP l6 and radiotherapy. .6.2 USE WITH TRANSPLANTATION THERAPY The compound of Formula I provided herein can be used to reduce the risk of Graft Versus Host Disease . Therefore, encompassed herein is a method of treating, preventing and/or ng cancer, which comprises administering the compound of Formula I, or an enantiomer or a mixture of enantiomers f, or a pharmaceutically acceptable salt, soivate, hydrate, co-crystal, clathrate, or polymorph thereof, in ction with transplantation therapy.
As those of ordinary skiii in the art are aware, the treatment of cancer is often based on the stages and mechanism of the disease. For example, as inevitable leukemic transformation develops in certain stages of cancer, transplantation of peripheral blood stem ~65— cells, hematopoietic stem cell preparation or bone marrow may be necessary. The combined use of the compound of Formula I provided herein and transplantation therapy provides a unique and unexpected synergism. In particular, the compound of Formula I exhibits immunomodulatory activity that may provide additive or synergistic effects when given rently with transplantation therapy in patients with cancer.
The compound of Formula I can work in combination with transplantation therapy reducing cations associated with the invasive procedure of transplantation and risk of GVHD. Encompassed herein is a method of ng, preventing and/or managing cancer which comprises administering to a patient (eg, a human) the compound of Formula I, an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof, before, during, or after the transplantation of umbilical cord blood, tal blood, peripheral blood stem cell, hematopoietic stem cell preparation, or bone . Some examples of stem cells suitable for use in the methods provided herein are sed in US. patent no. 7,498,171, the disclosure of which is incorporated herein by reference in its entirety.
In one embodiment, the compound of Formula I is administered to patients with multiple myeloma before, during, or after the transplantation of autologous peripheral blood progenitor cell.
In another embodiment, the compound of Formula I is administered to patients with ing multiple myeloma after the stem cell transplantation.
In yet another embodiment, the nd of Formula I and prednisone are administered as maintenance y to patients with le myeloma following the transplantation of autologous stem cell.
In yet r embodiment, the compound of Formula I and dexamethasone stered as salvage therapy for low risk post transplantation to patients with multiple myeloma.
In yet another embodiment, the compound of Formula I and dexamethasone administered as maintenance therapy to patients with multiple myeloma following the transplantation of autologous bone marrow.
In yet another embodiment, the compound of Formula I is administered following the administration of high dose of lan and the transplantation of autologous stem cell to patients with chemotherapy responsive multiple myeloma.
In yet another embodiment, the compound of Formula I and PEG INTRO—A are administered as maintenance therapy to patients with multiple myeloma following the transplantation of autologous CD34~selected peripheral stem cell.
In yet another embodiment, the compound of Formula I is administered with post lant consolidation chemotherapy to patients with newly diagnosed multiple myeloma to evaluate anti—angiogenesis.
In still another embodiment, the compound of Formula I and dexamethasone are administered as maintenance therapy after DCEP consolidation, following the treatment with high dose of melphalan and the transplantation of peripheral blood stem cell to 65 years of age or older patients with multiple myeloma.
In one embodiment, the compound of Formula I is administered to patients with NHL (6. g., DLBCL) before, during, or after the transplantation of autologous peripheral blood progenitor cell.
In another embodiment, the compound of Formula I is administered to patients with NHL (e.g., DLBCL) after a stem cell transplantation. .6.3 G THERAPY In certain embodiments, the prophylactic or therapeutic agents provided herein are ally administered to a patient. Cycling therapy involves the administration of an active agent for a period of time, followed by a rest for a period of time, and repeating this 2O sequential administration. Cycling therapy can reduce the development of resistance to one of one of the therapies, and/or or more of the ies, avoid, or reduce the side effects improves the cy of the treatment.
Consequently, in n embodiments, the compound of Formula I provided herein is stered daily in a single or divided doses in a four to six week cycle with a rest period of about a week or two weeks. The cycling method further allows the ncy, number, and length of dosing cycies to be increased. Thus, encompassed herein in certain embodiments is the administration of a compound provided herein, 6.g, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a ceutically able salt, soivate, hydrate, cor-crystal, clathrate, or rph thereof, for more cycles than are typical when it is administered alone. in certain embodiments, a compound provided herein, eg, the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a phannaceutically acceptable salt, e, hydrate, co«crystal, clathrate, or polymorph thereof, is stered for a greater number of cycles that would - 67 _ WO 25459 typically cause dose—limiting toxicity in a patient to whom a second active ingredient is not also being administered.
In one embodiment, the compound of Formula I is administered daily and continuously for three or four weeks at a dose of from about 0.1 to about 150 mg/d followed by a break of one or two weeks.
In another embodiment, the compound of Formula I and a second active ingredient are administered , with administration of the compound of a I occurring 30 60 minutes prior to a second active ingredient, during a cycle of four to six weeks. In certain embodiments, the combination of the compound of Formula I and a second active ingredient is administered by intravenous infusion over about 90 minutes every cycle. In certain embodiments, one cycle comprises the administration from about 0.1 to about 150 mg/day of the compound of Formula I and from about 50 to about 200 mg/mz/day of a second active ingredient daily for three to four weeks and then one or two weeks of rest. In certain embodiments, the number of cycles during which the combinatorial treatment is administered to a patient is ranging from about one to about 24 cycles, from about two to about 16 cycles, or from about four to about three cycles. .7 PHARMACEUTICAL COMPOSITIONS AND DOSAGE FORMS In one ment, provided herein are pharmaceutical compositions and dosage forms, which comprise the compound of Formula I, or an omer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co—crystal, clathrate, or polymorph f. In another embodiment, pharmaceutical compositions and dosage forms further comprise one or more excipients.
In certain ments, pharmaceutical compositions and dosage forms provided herein also comprise one or more additional active ingredients. Consequently, pharmaceutical compositions and dosage forms ed herein comprise the nd of Formuia I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable sait, solvate, hydrate, co—crystai, clathrate, or polymorph thereof, and a second active agent. Examples of optional , or additional, active ingredients are disclosed herein (see, e. g, section 4.3).
Single unit dosage forms ed herein are suitable for oral, mucosal (rag, nasal, subiingual, vaginal, buccal, or rectal), parenteral (cg, subcutaneous, intravenous, bolus injection, intramuscular, or rteriai), topical (cg, eye drops or other ophthalmic preparations), ermal, or transcutaneous administration to a patient. Examples dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic aerosols gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; s; for oral or mucosal (e. g, nasal sprays or inhalers); gels; liquid dosage forms suitable administration to a t, including suspensions (cg. , aqueous or non~aqueous liquid suspensions, oil~in—water emulsions, or a water-in-oil liquid emulsions), solutions, elixirs; liquid dosage forms suitable for eral administration to a patient; eye drops or solids (eg, other ophthalmic preparations suitable for l administration; and sterile forms lline or amorphous solids) that can be reconstituted to provide liquid dosage suitable for parenteral administration to a patient.
The composition, shape, and type of dosage forms provided herein may vary ing on their use. For example, a dosage form used in the acute treatment of a disease of the active ingredients than a dosage form may contain larger amounts of one or more used in the chronic treatment of the same disease. Similarly, a parenteral dosage form may form contain smaller s of one or more of the active ingredients than an oral dosage used to treat the same disease. See, e. g, Remington ’5 Pharmaceutical es, 18th ed., Mack Publishing, Easton PA (1990).
Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form provided herein depends on a y of factors, including, not limited to, the route of stration. For example, oral dosage forms such as tablets for use in parenteral dosage forms. The suitability of a may contain excipients not suited form. ular excipient may also depend on the specific active ingredients in the dosage For example, the decomposition of some active ingredients may be accelerated by some excipients such as lactose, or when exposed to water. Active ingredients that comprise ha U‘i primary or secondary amines are particularly tible to such accelerated decomposition.
Consequently, encompassed herein are phannaceutical compositions and dosage forms that contain little, if any, lactose. As used herein, the term “lactose—free” means that the amount of an of lactose present, if any, is insufficient to substantially increase the degradation rate active ingredient.
Lactose—free compositions provided herein can comprise excipients that are listed, for example, in the US. Pharmacopeia (USP) 25—NF20 . in certain embodiments, lactoseatiee compositions comprise active ingredients, a binderffiller, and a lubricant ceutically compatible and phannaceutically acceptable amounts. in certain embodiments, lactose—free dosage forms comprise active ingredients, microcrystalline cellulose, pre-gelatinized starch, and magnesium stearate.
Further encompassed herein are anhydrous ceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds. For example, the addition of water (eg. , 5%) is Widely accepted in the pharmaceutical arts as a means of simulating long-term e in order to ine characteristics such as life or the stability of formulations over time. See, eg, Jens T.
Carstensen, Drug Stability: Principles & Practice, 2d. Ed, Marcel Dekker, NY, NY, 1995, pp. 379-80. In effect, water and heat accelerate the decomposition of some compounds.
Thus, the effect of water on a formulation can be of great significance since re and/or humidity are commonly encountered during manufacture, handling, packaging, nt, and use of formulations.
Anhydrous pharmaceutical compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. Pharmaceutical compositions and dosage forms that se lactose and at least one active ingredient that comprises a primary or secondary amine are preferably anhydrous if substantial t with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, in certain embodiments, provided herein are ous itions packaged using materials to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose ners (eg, vials), blister packs, and strip packs.
Encompassed herein are pharmaceutical, compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose. Such compounds, which are referred to herein as lizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
Like the amounts and types of excipients, the amounts and specific types of active ingredients in a dosage form may differ depending on factors such as, but not limited to, the route by which it is to be administered to patients. In certain embodiments, the dosage forms provided herein comprise the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceuticaliy acceptable salt, solvate, hydrate, co‘crystal, clathrate, or polymorph thereof, in an amount ranging from about 0.10 to about 1000 mg, from about 0.10 to about 500 mg, from about 0.10 to about 200 mg, from about 0.10 to about 150 mg, from about 0.10 to about 100 mg, or from about 0.10 to about 50 mg. In certain embodiments, the dosage forms provided herein comprise the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a ceutically able salt, solvate, hydrate, co—crystal, clathrate, or polymorph thereof, in an amount of about 0.1, about 1, about 2, about 5, about 75, about 10, about 12.5, about 15, about 17.5, about 20, about 25, about 50, about 100, about 150, or about 200 mg. .7.1 ORAL DOSAGE FORMS In certain embodiments, pharmaceutical compositions provided herein that are suitable for oral administration are ated as discrete dosage forms, examples ofwhich e, but are not limited to, tablets (ag. , chewable s), caplets, es, and liquids (cg, flavored ). Such dosage forms contain predetermined amounts of active ingredients and may be prepared by some known methods of cy. See generally, Remington ’3 Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton PA (1990).
In certain embodiments, the oral dosage forms provided herein are prepared by combining the active ients in an intimate admixture with at least one excipient according to conventional ceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. For example, excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring , preservatives, and coloring agents. Examples ofexcipients suitable for use in solid oral dosage forms (cg, powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro‘ lline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms may be prepared by some known methods of pharmacy. In certain embodiments, pharmaceutical compositions and dosage forms are prepared by uniformly and tely admixing the active ingredients with liquid carriers, finely d solid carriers, or both, and then shaping the product into the desired presentation if necessary.
In certain embodiments, a tablet is prepared by compression or molding. In certain embodiments, compressed tablets are be prepared by compressing in a suitable machine the active ingredients in a free-flowing form, e.g., powder or granules, optionally mixed with an ent. In certain embodiments, molded tablets are made by g in a suitable machine a e of a powdered compound ned with an inert liquid diluent.
Examples of ents that can be used in oral dosage forms provided herein include, but are not limited to, s, fillers, egrants, and ants. Binders suitable for use in pharmaceutical compositions and dosage forms provided herein include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as , sodium alginate, alginic acid, other tes, powdered tragacanth, guar gum, cellulose and its derivatives (2g, ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl idone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g, Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
Suitable forms of microcrystalline cellulose include, but are not d to, AVICEL—PH—IOI, AVICEL-PH-103 AVICEL RC—581, AVICEL-PH—IOS (FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, PA), and es f. An specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose (cg, AVICEL RC—S81). Suitable anhydrous or low moisture excipients or additives include AVICEL—PH—103TM and Starch 1500 LM.
Examples of fillers suitable for use in the pharmaceutical compositions and dosage forms provided herein include, but are not limited to, talc, calcium carbonate (tag, granules or powder), microcrystalline cellulose, powdered cellulose, tes, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof. In certain embodiments, the binder or filler in pharmaceutical compositions provided herein is present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
Disintegrants are used in the compositions provided herein to provide tablets the ability to disintegrate when exposed to an aqueous environment. s that contain too much disintegrant may disintegrate in e, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms provided herein. The amount of disintegrant used varies based upon the type of formulation. In certain ments, the pharmaceutical compositions provided herein comprise from about 0.5 to about 15 weight t or from about 1 to about 5 weight percent of disintegrant.
Disintegrants that are suitable for use in pharmaceutical compositions and dosage forms provided herein include, but are not d to, agar-agar, alginic acid, calcium ate, microcrystalline ose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other es, clays, other algins, other celluloses, gums, and mixtures thereof.
Lubricants that are suitable for use in pharmaceutical itions and dosage forms provided herein include, but are not limited to, m stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g, peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl , ethyl laureate, agar, and mixtures thereof. Additional lubricants include, but are not limited to, a syloid silica gel (AEROSILZOO, W.R. Grace Co., Baltimore, MD), coagulated aerosol of synthetic silica (Degussa Co. of Plano, TX), CAB-O—SIL (a pyrogenic silicon dioxide, Cabot Co. of Boston, MA), and mixtures thereof. In certain embodiments, if used at all, ants are used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
In certain embodiments, provided herein is a solid oral dosage form, comprising the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof; and one or more excipients selected from anhydrous lactose, microcrystalline cellulose, nylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin.
In certain embodiments, provided herein is a solid oral dosage form, comprising the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically acceptable salt, e, hydrate, co-crystal, clathrate, or polymorph thereof; and anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin.
In certain embodiments, provided herein is a solid oral dosage form, comprising hydrochloride sale of the compound of a I, or an enantiomer or a mixture of omers thereof, or a pharmaceutically e, e, co-crystal, clathrate, or polymorph thereof; and one or more ents selected from anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, dal anhydrous silica, and gelatin.
In n embodiments, ed herein is a solid oral dosage form, comprising a hydrochloride sale of the compound of Formula I, or an enantiomer or a mixture of enantiomers thereof, or a pharmaceutically solvate, hydrate, co-crystal, clathrate, or polymorph thereof; and anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin. .7.2 DELAYED RELEASE DOSAGE FORMS In n embodiments, the active ingredients provided herein are administered by controlled release means or by ry devices. Examples e, but are not limited to, those described in US. Patent Nos: 3,845,770; 3,916,899; 809; 3,598,123; 4,008,719, 5,674,533, 5,059,595, 767, 5,120,548, 5,073,543, 5,639,476, 5,354,556, and 5,733,566, each of which is incorporated herein by reference in its entirety. In certain embodiments, such dosage forms are be used to provide slow or controlled—release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, c systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Encompassed herein are single unit dosage forms suitable for oral administration, ing, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled~release.
All controlled~release pharmaceutical products have a common goal of improving drug y over that achieved by their non-controlled rparts. Ideally, the use of an optimally designed controlled—release preparation in medical treatment is characterized by a minimum of drug substance being ed to cure or control the condition in a minimum amount of time. Advantages of controlled—release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance. in addition, controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood ievels of the drug, and can thus affect the occurrence of side (cg. , e) effects.
Most controlled—release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release of other s of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level that will of drug in the body, the drug must be released from the dosage form at a rate Controlled— replace the amount of drug being metabolized and excreted from the body. but not release of an active ingredient can be stimulated by various ions including, d to, pH, temperature, enzymes, water, or other physiological conditions or compounds. .7.3 ERAL DOSAGE FORMS Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, intraarterial. Because their administration typically bypasses patients’ natural es of being IO against contaminants, parenteral dosage forms are preferably sterile or capable sterilized prior to administration to a t. Examples of parenteral dosage forms include, be ved or but are not limited to, solutions ready for injection, dry products ready to suspended in a pharmaceutically able vehicle for injection, sions ready injection, and emulsions.
Some suitable vehicles that can be used to provide parenteral dosage forms provided herein include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, ’s Injection, Dextrose Injection, water-miscible Dextrose and Sodium Chloride Injection, and Lactated Ringer’s Injection; vehicles such as, but not limited to, ethyl alcohol, polyethylene , and opylene 2O glycol; and non-aqueous vehicles such as, but not d to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
Compounds that se the solubility of one or more of the active ingredients herein. disclosed herein can also be incorporated into the parenteral dosage forms provided the solubility of a For example, cyclodextrin and its derivatives can be used to increase compound provided herein, 6.g, the compound of Formula I, or an enantiomer or a of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, eo—crystal, clathrate, or poiymorph thereof. See, the disclosure of e.g., US. Patent No. 5,134,127, which is incorporated herein by reference in its entirety. .7.4 L AND MUCOSAL DOSAGE FORMS Topical and mucosal dosage forms provided herein inciude, but are not limited to, or other ophthaimic sprays, aerosols, solutions, emuisions, suspensions, eye drops preparations, or other forms known to one of skill in the art. See, ag, Remington ’5 WO 25459 Pharmaceutical Sciences, 16th and 18th eds, Mack hing, Easton PA (1980 & 1990); and Introduction to Pharmaceutical Dosage F arms, 4th ed, Lea & Febiger, Philadelphia (1985). Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels.
Suitable excipients (cg, carriers and diluents) and other materials that can be used to provide topical and mucosal dosage forms encompassed herein depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied. With that fact in mind, in certain embodiments, the excipients e, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene , butane-1,3-diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form solutions, emulsions or gels, which are non—toxic and pharmaceutically acceptable. Moisturizers or humectants can also be added to pharmaceutical itions and dosage forms if desired.
Additional examples of such ingredients can be found, eg, in Remington ’5 Pharmaceutical Sciences, 16th and 18‘h eds, Mack Publishing, Easton PA (1980 & 1990).
The pH of a pharmaceutical composition or dosage form may also be adjusted to improve delivery of one or more active ingredients. rly, the polarity of a solvent carrier, its ionic th, or tonicity can be adjusted to improve delivery.
Compounds such as tes can also be added to pharmaceutical compositions or dosage forms to ageously alter the hydrophilicity or lipophilicity of one or more active ingredients as to improve delivery. In this regard, stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery—enhancing ation—enhancing agent. Different salts, hydrates or solvates of the active ients can be used to further adjust the properties of the resulting composition. .7.5 KITS In certain embodiments, active ingredients provided herein are not administered to a patient at the same time or by the same route of administration. ore, encompassed herein are kits which, when used by the medical practitioner, can simplify the administration of riate amounts of active ingredients to a patient.
In certain embodiments, a kit ed herein comprises a dosage form of a compound provided herein, e. g.,. the compound of Formula 1, or an enantiomer or a mixture of enantiomers thereof, or a phannaceutically acceptable salt, solvate, hydrate, co-crystal, clatlnate, or polymorph thereof. In certain embodiments, the kit provided herein further comprises additional active ingredients, such as oblimersen (GENASESEg), melphalan, G- - 76 _ 2012/028498 CSF, GM-CSF, EPO, topotecan, azine, irinotecan, taxotere, IFN, COX—2 inhibitor, pentoxifylline, oxacin, dexamethasone, 1L2, 1L8, lLl 8, Ara-C, vinorelbine, isotretinoin, 13 cis—retinoic acid, or a phannacologically active mutant or derivative f, or a combination thereof. Examples of the onal active ingredients include, but are not limited to, those disclosed herein (see, e. g, section 4.3). that is In n embodiments, the kit provided herein further comprises a device used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes, drip bags, patches, and inhalers. blood for In certain embodiments, the kit provided herein further ses cells or transplantation as well as pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided kit can comprise in a solid form that must be reconstituted for parenteral administration, the the active ingredient can be dissolved to a sealed container of a suitable vehicle in which form a particulated’ree sterile solution that is suitable for parenteral administration.
Water for Examples of pharmaceutically acceptable vehicles include, but are not limited to: Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer’s Injection, se Injection, Dextrose and Sodium Chloride Injection, ed Ringer’s ion; water-miscible vehicles such as, but not limited to, ethyl such as, alcohol, polyethylene glycol, and polypropylene glycol; and non—aqueous vehicles but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate. 6. EXAMPLES n embodiments of the invention are illustrated by the following non-limiting examples. 6.1 PREPARATION OF 3-(5—AMINO-2—METHYL0XO-4H— QUINAZOLIN-S—YL)-PIPERIDINE-2,6-DIONE till“NHgoUNJ\/ Step l: To a solution of potassium hydroxide (16.1 g, 286 mmol) in water (500 The suspension mL), was added 3-nitrophthalimide (25.0 g, 130 mmol) in portion at 0 OC.
°C for 3 hrs. To the solution, was added was stirred at 0 CC for 3 hrs, and then heated to 30 HCI (100 mL, 6N). The resulting suspension was cooled to 0 °C for 1 hr. The suspension was filtered and washed with cold water (2 x 10 mL) to give 3-nitro—phthalamic acid as a white solid (24.6 g, 90% yield): 1H NMR (DMSO-dg) (3 7.69 (hrs, 1H, NHH), 7.74 (t, J = 8 Hz, 1H, Ar), 7.92 (dd, J: 1, 8 Hz, 1H, Ar), 8.13 (dd,J= 1, 8 Hz, 1H, Ar), 8.15 (brs, 1H, U! NHH), 13.59 (s, 1H, OH); 13C NMR (DMSO~d6) 6 125.33, 129.15, 130.25, 132.54, 136.72, 147.03, 165.90, 167.31.
Step 2: To a mixture of 3—nitro-phthalamic acid (24.6 g, 117 mmol) and potassium ide (6.56 g, 117 mmol) in water (118 mL), was added a mixture of bromine (6 mL), potassium hydroxide (13.2 g, 234 mmol) in water (240 mL) at 0 OC, followed by addition of a solution of potassium hydroxide (19.8 g, 351 mmol) in water (350 mL). After 5 minutes at 0 0C, the mixture was heated in a 100 OC oil bath for 1 hr. The reaction solution was cooled to room temperature, and then, in an ice-water bath for 30 minutes. To the e, a solution of HCl (240 mL, 2N) was added dropwise at 0 OC, and the ing mixture was kept for 1 hr. The sion was filtered and washed with water (5 mL) to give 2—amino- 6-nitro—benzoic acid as yellow solid (15.6 g, 73% yield): HPLC: Waters Symmetry C13, Sum, 3.9 x 150 mm, 1 mL/min, 240 nm, CH3CN/0.1% 4, 5% grad to 95% over 5 min, .83 min (85%); 11‘1 NMR (DMSO-d6) a" 6.90 (dd, J: 1, 8 Hz, 1H, Ar), 7.01 (dd, J: 1, 9 Hz, 1H, Ar), 7.31 (t, J: 8 Hz, 1H, Ar), 8.5-9.5 (brs, 3H, OH, NH;); 13C NMR (DMSO-dé) a 105.58, 110.14, 120.07, 131.74, 149.80,151.36, ; LCMS: MH = 183.
Step 3: A mixture of 2—amino-6—nitro—benzoic acid (1.5 g, 8.2 mmol) in acetic anhydride (15 mL) was heated at 200 0C for 30 minutes in a microwave oven. The mixture was filtered and washed with ethyl acetate (20 mL). The filtrate was concentrated in vacuo.
The solid was stirred in ether (20 mL) for 2 hrs. The suspension was filtered and washed with ether (20 mL) to give 2-methylnitro-benzo[d][1,3]oxazin—4-one as a light brown solid (1.4 g, 85% yield): HPLC: Waters Symmetry C13, 5 um, 3.9 x 150 mm, 1 mL/min, 240 nm, CH3CN!0.1% 1131304, 5% grad 95% in 5 min, 5.36 min (92%); 1H NMR (DMSO—d(,) 6 2.42 (s, 3H, CH3), 7.79 (dd, J: 1, 8 Hz, 1H, Ar), 7.93 (dd, J: 1, 8 Hz, 1H, Ar), 8.06 (t, J: 8 Hz, 1H, Ar); 13C NMR (DMSCLdg) 5 20.87, 107.79, 121.54, , 137.19, 147.12, 148.46, 155.18, 161.78; LCMS: MH 2 207.
Step 4: Two Vials each with a suspension of 5-nitromethyl-benzo[d][1,3]oxazin- 4—one (0.60 g, 2.91 mmol) and 3~amino-piperidine~2,6udione hydrogen Chloride (0.48 g, 2.91 mmoi) in pyridine (15 mL) were heated at 170 0C for 10 minutes in a microwave oven.
The suspension was filtered and washed with ne (5 mL). The filtrate was concentrated in vacuo. The resulting mixture was stirred in HCl (30 mL, 1N), ethyl acetate (15 mL) and ether (15 mL) for 2 hrs. The suspension was filtered and washed with water (30 mL) and ethyl acetate (30 mL) to give a dark brown solid, which was stirred with methanol (50 mL) at room temperature overnight. The suspension was filtered and washed with methanol to give ethyl—5—nitro—4—oxo~4H-quinazolin~3—yl)-piperidine~2,6-dione as a black solid (490 mg, 27% yield). The solid was used in the next step without further purification.
Step 5: A mixture of ethyl—5-nitro-4—oxo~4H-quinazolin~3vyl)-piperidine—2,6~ dione (250 mg) and Pd(OH)2 on carbon (1 10 mg) in DMF (40 mL) was shaken under hydrogen (50 psi) for 12 hrs. The suspension was filtered through a pad of Celite and washed with DMF (10 mL). The filtrate was concentrated in vacuo and the resulting oil was purified by flash column chromatography (silica gel, methanol/methylene chloride) to give 3—(5-amino-2—methyl-4—oxo~4H-quinazolin-3~yl)~piperidine—2,6~dione as a white solid (156 mg, 69% yield): HPLC: Waters Symmetry C18, Sum, 3.9 x 150 mm, 1 , 240 nm, 10/90 CH3CN/0.1% 1131304, 3.52 min ); mp: 293-295 0C; IH NMR (DMSO‘dfi) 85 2.10—2.17 (m, 1H, CHH), 2.53 (s, 3H, CH3), 2.59—2.69 (m, 2H, CH2), 2.76-2.89 (m, 1H, CHH), 5.14 (dd, .12 6,11 Hz, 1H, NCH), 6.56 (d, J: 8 Hz, 1H, Ar), 6.59 (d, J: 8 Hz, 1H, Ar), 7.02 (s, 2H, NH;), 7.36 (t, J: 8 Hz, 1H, At), 10.98 (s, 1H, NH); ”C NMR (DMSO~d6) 620.98, 23.14, 30.52, 55.92, 104.15, 110.48, 111.37, 134.92, 148.17, 150.55, 153.62, 162.59, 169.65, 172.57; LCMS: MH = 287; Anal. Calcd. for CMHMN403 + 0.3 H20: C, 57.65; H, 5.05; N, 19.21. Found: C, 5750,11, 4.73; N, 19.00. 6.2 ASSAYS 6.2.1 TNFu INHIBITION ASSAY IN PMBC Peripheral blood mononuclear cells (PBMC) from normal donors are obtained by Ficoll Hypaque (Pharmacia, Piscataway, NJ, USA) density centrifugation. Cells are cuitured in RPMI 1640 (Life Technologies, Grand Island, NY, USA) supplemented with 10% an serum (Gemini Bio-products, nd, CA, USA), 2 mM L—glutamine, 100 1131111. peniciilin, and 100 ug/mL streptomycin (Life Technologies».
The PBMCs (2 X 105 cells) were plated in 96-we11 flat—bottom Costar tissue culture piates (Corning, NY, USA) in triplicate. The cells were ated with LPS (from Salmoneila abortus equi, Sigma cat. no. L-1887, St. Louis, MO, USA) at 1, ngimL final in the absence or presence of compounds to be tested. The compounds were dissolved in DMSO (Sigma) and further diiutions were done in e medium immediately before use.
The final DMSO concentration in all assays was about 0.25%. The compounds were added to cells 1 hour before LPS stimulation. The cells were then incubated for 18—20 hours at 37°C in 5% C03, and supernatants were then collected, diluted with culture medium and assayed for TNFa. levels by ELISA (Endogen, Boston, MA, USA). Ing values were calculated using non—linear regression, sigmoidal dose-response, constraining the top to 100% and bottom to 0%, allowing variable slope (GraphPad Prism V3.02). 6.2.2 INHIBITION OF MM CELL PROLIFERATION The ability of compounds to effect the proliferation ofMM cell lines was investigated in an in vitro study. Uptake of [3H]-thymidine by H929 MM cells and 7—AAD uptake in several MM cell lines (H929, U266Bl, Anbl—6, KMS-34, OPM-Z, DF-IS, DFl S/R, CAG, MM1.S and LP~1) was ed as an indicator of cell proliferation. Cells were incubated in the presence of compounds for 72 hours ([3H]-thymidine was included for the last 6 hours of the tion period) or 5 days ed by 7-AAD uptake to measure and count Viable cells. 6.2.3 CYTOKINE PRODUCTION BY T CELLS T cells were isolated from buffy coat by negative selection using the RosetteSep® T Cell Enrichment Cocktail. The cturer’s procedures were followed accordingly. All l plates were ated with 3 rig/ml anti—human CD3 antibody in 100 ul 1X PBS for 4 hours at 37°C. The plates were washed three times with RPMI-1640 Complete Media prior to the T cell assay. T cells were then plated in CD3 ated plates at a density of 2.5 X 105 cells/well in 180 pl RPMI-1640 Complete Media. The cells were treated with 20 ul 10X titrated compounds at 10, 1, 0.1, 0.01, 0.001, 0.0001 and 1 uM. Final DMSO concentrations were 0.25%. The plates were incubated for 48 hours at 370C, 5% C02. After 48 hours, the supernatants were harvested and tested by a multi-plex cytomteric bead array [“0 {J} (CBA) assay for the following cytokines/chemokines: lL-2, IL—3, IL-5, IL—10, 1L~l3, IL-IS, IL-17a, GM«CSF, G~SCF, lFN-y, TNF-ot and RANTES. The CBA plates were analyzed on the Luminex 18100 instrument.
Cytokine levels were normalized to the amount ed in the presence of the amount of a compound tested, and ECSO values were ated using non-linear regression, sigmoidal dose—response, constraining the top to 100 % and bottom to 0 %, allowing variable slope (GraphPad Prism $53.02). -30..
Anti-CD3-stimulated human T cell assay All 96—well plates were pre-coated with 3 ug/mL uman CD3 antibody in 100 uL 1X PBS for 4 hours at 37°C. The plates were washed 3 times with RPMl-1640 Complete Media prior to the T cell assay. The T cells were then plated in anti-CD3 —pre-coated plates at a density of 2.5 X 105 cells/well in 180 uL 640 Complete Media. The cells were treated with 20 uL 10X titrated Celgene compounds at 10, 1, 0.1, 0.01, 0.001, 0.0001, and 0.00001 uM in duplicate. The final DMSO concentrations were 0.25%. The plates were incubated for 48 hours at 37°C, 5% CO2. After 48 hours, the supernatants were harvested and tested by a multiplex cytometric bead array (CBA) assay for the following cytokines/chemokines: lL-Z, lL—3, lL-5, IL—10, IL—13, 1L-15, lL—17A, GM-CSF, G-CSF, lFN-y, TNF—u, and . The CBA plates were ed on the Luminex 18100 instrument. 6.2.4 CYTOTOXICITY ASSAY Cells line Farage, DOHH2 and Rec-1 were obtained from an Type Culture tion (Manassas, VA, USA) xicity assays were measured in 3 day ATP production assays as follows: The cells were plated in black/c1ear—bottom 96-well TC plates (BD Falcon, Cat # 353948) at 3000 cells/75 pL (for DoHH-Z and Farage cells) or 6000 cells/75 uL (Rec—1 cells) media per well. Stock solutions (40X) of nds were prepared in DMSO and 4X solutions were prepared by diluting the 40X stock solutions 1:10 with 1% DMSO in culture medium In each assay plate well, 25 uL of the compound of, a I in 1% DMSO were added to the cells in triplicate so that the final volume was 100 uL and [DMSO] final was 0.25%. Plates were then sealed with breathable sealing films (ISC BioExpress, Cat # T— 2421-50) and placed in a 37°C, 5% C02 humidified incubator for 72 hours. In addition, cells were seeded in a separate plate in the same manner as above, 25 uL medium in 1% DMSO was added to each well. This plate was immediately tested in the CellTiter—Glo Luminescent Cell Viability Assay (Promega, Cat a 67572) as 0 time point and the results were used to ate Gngo in the Farage and DOHH-ZZ cell experiments After 72 hours of incubation, 100 at. of CellTiter-Glo reagent were added to each well and incubated at room temperature with gentle shaking for 30 minutes. The plates were then analyzed for luminescence in a TopCount NXT Reader (Packard). Each well was counted for one second. Values for duplicate wells were averaged and then compared to the zero time point DMSO control (0% inhibition) to calculate the percentage inhibition of cell growth. Mean DOHH-2 Gleo values and Farage GIC50 values were calculated from three experiments. Rec-l IC50 values were calculated from two ments. 6.2.5 CELL CYCLE ANALYSIS Cells were treated with DMSO or an amount of a compound provided herein for 48 VI hours. Propidium iodide staining for cell cycle was performed using CycleTEST PLUS (Becton Dickinson) according to manufacturer’s protocol. Following staining, cells were analyzed by a FACSCalibur flow cytometer using ModFit LT software (Becton Dickinson). 6.2.6 APOPTOSIS IS Cells were treated with DMSO or an amount of a compound provided herein at various time points, then washed with annexin-V wash buffer (BD Biosciences). Cells were incubated with annexin-V binding protein and propidium iodide (BD Biosciences) for 10 minutes. Samples were analyzed using flow cytometry. 6.2.7 NK CELL ANALYSIS Ninety—siX-well flat—bottom plates were coated with 100 ug/mL of human lgG (Sigma) ght at 4°C. The next day, unbound lgG was washed away with cold 1X PBS. NK cells were then plated in the lgG-coated 96-well plates at 2 x 105 cells per well in 180 uL RPMI~1640 Media and 10 ng/mL of rhIL-2 (R & D Systems, MN) was added. Test nds were added in a volume of 20 uL DMSO. Final concentrations of test nds were 0.0001, 0.001, 0.01, 0.1, 1, or 10 uM. Final DMSO concentrations were 0.25%. After 48 hours, the supernatants were harvested and analyzed by ELISA for lFN—y production. 6.2.8 RESULTS The ical activities of the nd of Formula I are summarized in Tables 1 to 5. in the anti—CD3-stimulated human T cell assay described abovea the compound of Formula 1 enhanced production of IL-2, lL—3, lL-S, lL-lO, lL-lS, GM-CSFg lNF~v, RANTES, and TNF-d at concentrations of 0.01 to 10 MM. Enhancement of lL-Z, lL—3, IL— 13, GM-CSFS TNF~d, and RANTES by the nd was concentration-dependent. At a concentration of 0.1 uM of the compound of Formula 1, production of lL—Z and lL-13 was enhanced to levels 14x and 7X those in control cells, respectively. At a concentration of 1 nM of the compound of Formula 1, production of lL-2 and 1L~l3 was enhanced to levels 17X and 8x those in control ceiis, respectively. The compound ed lL-lO production 2-foid at low concentrations (fé 0.01 uM) but inhibited lL-10 production at 1 and 10 MA. and 0.1 uM, respectively, The nd increased IL-5 production 3— and 4—fold at 0.01 showing less enhancement at both lower and higher concentrations.
Additionally, it was observed that, in a human umbilical artery assay. the compound of 9.4 nM; and the compound of Formula I was a potent anti-angiogenic agent with an Ing of a I did not inhibit HUVEC proliferation.
In a mouse Matrigelum angiogenesis assay, it was observed that the compound of and exhibited a dose Formula I significantly inhibited blood vessel growth at 30 mpk dependent inhibition of angiogenesis. in DoHHZ and It was observed that the nd of Formula I induced Gl arrest of Formula WSU-DLCL2. It was also observed that, in proliferation assays, the compound method.
I acted istically with Rituxan, as calculated using the Chou-Talalay of Formula I In a DoHH2 xenograft model, it was observed that the compound of Formula I with inhibited tumor growth and that the combination of the compound mg/kg dose. Tumor Rituxan significantly delayed time to tumor endpoint (63%) at of Formula I in IS growth inhibition was observed at 3 and 30 mpk of the compound It was also combination with Rituxan (1 , at 45% and 55% on day 12, respectively. s counts in observed that the compound of Formula I significantly inhibited blood tumor. of Formula I In a CLZ xenograft model, the combination of the compound volume < with Rituxan (2 rug/kg iv qw) yielded 60% and 90% te sions (tumor mm3) at 3 and 30 mg, respectively.
I inhibited H929 In a NCl-H929 MM xenograft model, the compound of Formula the compound showed 93% tumor tumor growth in a dose-dependent manner. On day 19, and 59% tumor growth inhibition at 30 mg/kg, 73% tumor growth inhibition at 3 mg/kg, growth inhibition at 0.3 mg/kg. volume was In a U87 GB xenograft model, dose dependent inhibition of tumor at 3 and observed. The compound of Formula I significantly inhibited U87 tumor growth mg/kg qd.
TABLE 1. In Vitro Activities Assay ICso or EC50 (11M) PBMC TNFO. 0.063a WB TNFOL 0.164a LPS~induced TNFa 0.0173 T cell lL-2 0.012 ~ 0.014C RECI (MCL) 0.47a DoHH2 (FL) 0.61b Farage (GCB-DLBCL) 0.70b Human angiogenesis 0.00943 NK cell IFNy 0.0015c B cell proliferation 0.015C B cell lgG 0.061a Immature MK colonies >10"11 Intermediate MK es >10a a ""3 IC50, b 3 GleO, C ""3 EC50 TABLE 2. In Vitro Activities (5 Day 3H Thymidine Incorporation Assay) [€50 (HM) OCI~Ly10 0.0085 U2932 0.11 m 0.12 ABC Subtype f mos 0.44 RIVA 4.3 m 7 PMBL Karpas~1106P 0.58 —. 0.71 g 7 CLZ 0.79 _ 2.1 l ~§ GCB Subtype 1 SUDHL4 j >10 % l ; OCl—Lyl9 >10 § ~84- TABLE 3. Activity of the Compound of Formula 1 against Lenalidomide Resistant Cell Lines (ICSQ (01%)) H929 D1 1 1051 1052 1053 1054 Lenolidomide 12.64 No IC50 No Ing No [€50 No ICSO No ICSO (11:3) .1 1 Cmpd. of 0.1539 0.3092 2.974 4.238 2.099 6.593 Form. I <n=3> TABLE 4. Effect of the Compound of Formula I in HIF-lu protein expression in solid tumor cell % Inhibition at Cancer cell lines. (111M) ctal Cancer HCTl 16 74.60% HCTI 5 69.26% Ovarian Cancer 100.00% Renal Cancer 7360 41 70% Brain Cancer 73 . 8 1% TABLE 5. Anti-Proliferative Activity of the Compound of Formula I in DLBCL cell lines Correlation with Compound of Formula I Anti-proliferative Statistics fl activity: £100 nM) OneomineTM ABC scores Correlated P < 0.05 r2 = 0.48 neTM NFKB scores Not Correlated P > 0.05 r2 = 0.35 Baseline activity ofNFKB subunit Conelated P < 0.005 r2 2 0.60 baseline activity ofNFKB subunit Correlated P < 0.01 r2 = 0.65 ne IRF4 gene expression Correlated P < 0.05 r2 = 0.47 Baseline SPIB gene expression Not Correlated P > 0.05 r2 = 0.027 Baseline cyclin D1 gene Not Correlated P > 0.05 r2 x 0.21 expression Baseline A20 gene sion Not Correlated P > 0.05 r2 == 0.044 Baseline CARDll gene expression Correlated P < 0.05 r2 = 0.54 Baseline CRBN gene expression Correlated P < 0.05 r2 = 0.45 6.3 PHARMACOKINETICS It was observed that the compound of Formula 1 had a fig of 230 min in human plasma. The oral pharmacokinetic ters in mouse, rat, and monkey are summarized in Tables 5 to 7. res (AUC(0-0) of the compound of Formula I increased in a dose proportional manner up to 30 mg/kg in SCID mouse, CD-IDS rat! and male monkey. The compound of Formula 1 did not inhibit any megakaryocyte itor cells at 10 uM.
TABLE 6. Oral Pharmacokinetics } SCID Mouse SD Rat l Cyno Monkey l l l Dose (mpk) 3 po 30 po 3 po l , i“ l ‘ 1 2900 l 4800 3300 3 l C . ’ imaX ng/m( L ( MDu } ‘ (10) l (17) » (H) l i f 7l00 25000 12000 I AUC (ng-h/mL (uM-h) (25) i (87) (43) l in;2 (h) i 2.7 5.8 l g CLp (mLfminfkg) g l l 1 1.2 l l i I P (as) l 53 l 32 l TABLE 7. PK Profiles in Male Monkeys Dose (mg/kg) Cmax (ng/mL) AUCM (ng*h/mL) 0.3 100 (0.36 11M) 1300 (4.5 pM—h) 3 1100 (3.8 pM) 14000 (49 nM-h) 3100 (11 11M) 38000 (130 nM-h) 7700 (27 0M) 99000 (350 pM—h) TABLE 8. Pharmacokinetics in Monkeys on Day 1 Dose (mg/kg) Tmax (h) Cmax (ng/mL) AUC0-24 (ng*h/mL) 2 to 4 (ma) 36(m) 430(m) 0.15 2 (1 ) 63(1) 450(1) 2 to 4 (m) 510(m) 4600(m) 1 _ 2 (f) 680(1) 4100(1) 4 (m) 4100(m) m) 2 to 4 (i) 4200(D 380006) a. m: Male; b. f: Female.
TABLE 9. Pharmacokinetics in s on Day 27 Dose (mg/kg) Tm (h) Cm (ng/mL) AUCW, (ng*h/mL) 4 (ma) 53(m) 570(m) 0.15 2 to 4 0“) 57(1) 4500—“) 2 (m) 560(m) 5700(m) 1 5‘ 0.5 to 2 (0 . 590(1) 42000) ‘ E 2104011) 5800(m) 72000(m) E 40) L 70000) 750000) 5 a. m: Male; h. f: Female.
The oral administration of the compound of Formula I at 100, 300., and 10000 mg/kg/day for 7 consecutive days in the male CD-IGS rat ed generally in near dose tional exposure increases. The NOAEL was determined to be 1000 mg/kg/day. -87— 6.4 IN VITRO DLBCL CELL THYMIDINE INCORPORATION ASSAY A panel of DLBCL cell lines of various netic features was tested for their sensitivity to the antiproliferative activity of the compound of Formula I (. Cells were treated with the compound of Formula I for 5 days at 370C; proliferation of cells determined using 3H-thymidine incorporation . Results of 3 independent experiments are shown (mean :l: SD) in The compound starting at 0.1 —l uM significantly (p<0.05) inhibited proliferation of several lines of DLBCL cells, particularly ABC-subtype cells such as Riva, U2932, TMD8, OCI-Ly3 and OCI-Lle cells. ABC- subtype cells appear more sensitive to the antiproliferative effect than other subtype cells including BCL and PMBL cells. 6.5 TORY EFFECT ON NFKB ACTIVITY IN DLBCL CELLS DLBCL cells were treated with the compound of Formula I or an IKK1/2 dual inhibitor (used as a positive inhibitor l) for 2 days. NFKB activity was examined with Active Motif transcription factor assay using nuclear extracts from cells following treatment. Results are shown in (mean i SD). The compound of a I significantly inhibits NFKB p65 and p50 activity at concentrations of 0.1 uM, 1 uM and 10 uM. The compound of Formula I was found to inhibit the NFKB activity in some DLBCL lines of the ABC subtype, such as U2932 and e cells. These s suggest that an effect on NFKB signal transduction might be ed in the antiproliterative activity of the compound of Formula I against BCL cells, and that the baseline NFKB activity may be a predictive biomarker of lymphoma tumor response to therapy with the compound. 6.6 IN VIVO MOUSE XENOGRAPH MODEL FOR THE OCI-LYIO CELL S’UBTYPE Efficacy of the compound of Formula I t the OCI-Ly 10 cell subtype is investigated in an in viva mouse xenografi model. Female CB. 1 7 SCID mice age 6 to 12 weeks are injected with about 0.2mL/mouse of 1x107 OCI~Lyl O tumor cells in 100% Matrigel sc in flank. Treatment with the compound of Formula I begins once tumor reaches an average size of 100 to 150 mg. Body weight is measured 5/2 and then biweekly to the end of the study. Caliper measurement of the tumor is performed biweekly. The endpoint of the study is tumor growth delay (TGD). The tage TGD is calculated. Animals are monitored individually. The endpoint of the study is a tumor volume of about 1000 m3 60 days? whichever comes first. Responders to therapy may be followed longer.
Tumor collection: collect tumors in RNAse free environment (divide into 3 parts).
Part is is preserved via snap freeze as a powder for future protein analysis, shipping condition -80 OC. Part 2 is preserved in RNA later, snap freeze} shipping condition -80 °C.
Part 3 is preserved in formalin for 24 hours. then 70% ethanol, ship at room temperature to PAT for paraffin embedding. The treatment plan is shown below.
Gr. N Agent mg/kg Route Schedule 1 10 vehicle 1 ~«- po qd x 28 2 10 Compound of Formula I 3 1Wpo qd x 28 3 10 nd of Formula I 10 po qd x 28 4 10 Compound of Formula I 30 *po qd X 28 10 vincristine 1 iv q4d x 28 6.7 MULTIPLE MYELOMA MODELS The y of 3-(5~amino-2«methyl—4—oxo-4H—quinazolin—3-yl)-piperidine-2,6~dione to inhibit cancer cell growth was evaluated in a number of multiple myeloma (MM) cell lines using in vitro and in viva methods ( & SB). 3~(5~amino-2—methyloxo—4H- quinazolin—3—y1)-piperidine-2,6-dione was shown to inhibit MM cell proliferation in a number of cell lines (, 5B 8: 6). For example: the antiproliferative effect of 3—(5— amino-2—methyloxo—4H—qyuinazolinyl)~piperidine—2,6~dione was trated in a N929 xenograft model (. 6.8 CEREBLON MODELS IN ABC-DLBCL, MULTIPLE MYELOMA AND CTAL CANCER CELLS The effect of the protein cereblon (CRBN) on the efficacy of the compound of Formula I to inhibit the proliferation, cell cycle progression and/or cell on of various cancer cell lines was studied. The compound of Formula I was found to interact with endogenous myeloma CRBN and in a dose-dependent manner. The compound of Formula I also interacts with HepGZ HCC CRBN in a dose-dependent manner. In addition. the compound of Formula I was found to inhibit CRBN ubiquitination with an ngg of 208.7 ABC-DLBCL cell model The expression of cereblon was found to modulate the efficacy of the nd of Formula 1 against proliferation of ABC—DLBCL cell lines (~7C). Cerebion was ed for inhibition of each of IRE-‘4 expression, NFKB activity, and cell proliferation.
.Myeloma cell models - 89 _ The effect of cereblon in H929 myeloma cells was also ted. H929 cells were transfected with mock, negative control siRNA and CRBN-siMA-7 for 24, 48, 72 and 96 hours. Cells were d 24 h after transfection with DMSO (0.1 0/o) or the compound of Formula I for I, 2, 3 days and the effect on cell cycle and eration investigated. The compound of Formula I induced a delay of cell cycle ssion, measured as the decrease of the number of cells in S phase, in control mock and ve control siRNA—transfected cells after 72 h treatment (. Knockdown of CRBN markedly abrogated drug-induced delay in cell cycle progression in H929 cells from 65 to 22% for the nd of Formula RTsPCR and Western blot analysis was used to measure the levels of key celi cycle and apoptotic regulators in order to further investigate the effects of CRBN on the cell cycle arrest induced by the compound of Formula I . In H929 cells, the cell cycle arrest in G1 phase by the compound of Formula I coincides with a reduction of tumor suppressor, pr, phosphorylation and the oncogene and myeloma survival factor IRF4. Western blot analysis showed that the compound of Formula I decreased phosphorylation of pRB ( & 9B) and total level of protein IRF4 ( & 9D). The effect was reduced by knockdown ofCRBN suggesting that tion of cell cycle progression by the drugs requires CRBN protein.
The compound of Formula I was found to inhibit the proliferation of CRBN- ive MM cell lines U266, 100-1 and lK-2 ().
Colorectal cell model The expression of cereblon also modulates the anti—invasive activity of the compound of Formula I in HCT-IS colorectal cancer cells (). The ability of the compound of Formula I to inhibit invasion of HCT-l 5 cells was reduced by siCRBN. to U! 6.9 SOLID TUMOR MODELS The compound of a I was evaluated for its effect on solid tumor cell lines from a variety of histologies (sag, breast, ovarian, colorectal, HCC). The compound of Formula I inhibits hypoxia-induced HIFl—o expression in many such solid tumor cell lines (FIG. l2A-121). In addition, the compound of Formula I ts the on of solid tumor cells to varying degrees (Table 10) and cell colony formation (Table ll). The inhibition of solid tumor ceil colony formation was studied by a single high concentration treatment of the Compound of formula I { lOuM) on day I, followed by monitoring of cell colony formation over the course of 10 to 20 days (Table II, A & I38).
The compound of Formula I ts U87 glioiblastorna tumor cell growth at 3 and nag/kg q.d. in a xenografi model ().
TABLE 10: Effects of the Compound of Formula I on on of Solid Tumor Cells Tumor Cell Type Cell Line (stimulation) Invasion (ICSQ) Compound of Formula I HepG2 (VEGF) < 0.001 hepatocellular SK-HEP—l (VEGF) 0.0061 SNB-l9 (PDGF) 0.16 {SF—539 (PDGF) 0.025 gliohlastoma U251 (PDGF) 3.7 SF-295 (PDGF) 0.24 U87 (PDGF) 0.08 coloreotal HCTlS (bFGF) 0.0072 TABLE 11: Effects of the Compound of Formula I in Solid Tumor Cell Colony Formation Tumor Cell Type Cell Line % Inhibition of Colony Formation” HCTI 5 3 hepatocellular HCTl 16 1 3 =l< * Colo—205 l7** ovarian OVCAR—3 1 8* SK-HEP-l 6 HCC HEP—G2 6.9 SF268 0.6 SF29S 12.9 E astoma l U251 i -6 l U87 2 l ‘ } MDA-MB-453 I -7 § breast i MCF-T ; 1.4 g j 211-754 l 90M :1 prostate l PC-3 i 14.8 a: 10 pM of Compound of Formula I. *= p < 0.5 ; M: p < 0.001 (versus DMSO). 6.10 PBMC CYTOKINE PROFILING The compound of Formula I was selected for activity profiling of eleven (11) cytokines and chemokines, i.e., eukin (ID-10, IL—6, IL-8, granulocyte macrophage colony stimulating factor (GM—CSF), macrophagaderived chemokine (MDC), macrophage inflammatory protein-l alpha (MIP—lu), macrophage inflammatory protein-lbeta (MIP-l B), tumor necrosis —alpha (TNF—u), IL—IO, monocyte actic protein—l (MCP-l), and RANTES (regulated upon activation normal Tacell sed, and secreted) using lipopolysaccharide—stimulated human peripheral blood mononuclear cells s) obtained from 2 — 6 donors.
The compound of Formula I inhibited the production of (in order of potency) TNF~0L (ICSO 2 0.034 uM), > IL-lfi (1ng = 0.054 uM) >~ IL-6 (ICSU 2 0.060 uM) > MDC (K350 = 0.062 MA) > MIP—la (IC50 2 0.30 MA) > GM—CSF (Ing = 0.95 uM) > lL-S (le0 > 10 uM) > MIP-IB (ICSO > 10 MM) (Table 12). The compound of Formula I also enhanced IL- , MCP—l, and RANTES production with mean percent of control values of 480%, 236%, IS and 131%, respectively at the 0.1 uM concentration.
TABLE 12: Summary of Cytokine Inhibitory Profile of the nd of Formulal IL-6 ’IL-8 IL—10 GM-CSF MDC IMip—m MIP-lB TNF-a IC50 0.060 l>10 i0.054 0.95 #00621030 >10 0.034 6.11 VEGF-,BFGF-, AND HGF-INDUCED HUVEC TUBE FORMATION, MIGRATION, AND ON The nd of formula I trated potent inhibitory activity in an in vitro assay of human umbilical vascular endothelial cell (HUVEC) invasion. The compound of formula I strongly inhibited vascular cndothelial growth factor (VEGF)—, basic fibroblast growth factor (bFGF)-, and hepatocyte growth factor (HOD-induced invasion, weakly inhibited VEGF and bFGF-induced HUVEC tube formation and migration, and either k) U: enhanced or did not inhibit proliferation of growth factor-induced HUVEC proliferation.
The ngQ value for inhibition of VEGF-induced HUVEC invasion was 0.29 nM. The Ing value for tion of bFGF—induced HUVEC invasion was 5.5 nM. The ICSQ value for inhibition of HOP-induced HUVEC invasion was 110 nM. The compound of a I inhibited VEGF- and bFGF-induced migration 38% and 280/00 respectiver at a concentration of 1 MA. 6.12 CLINICAL PROTOCOL A Phase la/ lb, clinical study to determine the safety, tolerability, pharmacokinetics and efficacy of the the compound of Formula I when administered orally to subjects with advanced solid tumors, Non-Hodgkin’s lymphoma, or multiple myeloma is provided. The U) non-tolerated dose (NTD), the maximum tolerated dose (MTD) and the recommended phase 2 dose (RP2D) are to be defined in the study. The effect of the compound on biomarkers of angiogenesis in pre- and during treatment tumor biopsies will be evaluated.
Study Design The study is designed as a Phase la/ lb study consisting of two parts: dose escalation (Part A), and dose expansion (Part B). In Part A, subjects will receive single and multiple ascending doses of the compound of Formula I to measure pharmacokinetics (PK) and identify the m tolerated dose (MTD) and the recommended phase 2 dose (RPZD).
A standard dose (3+3) escalation design (Simon et al., 1997) will be used to fy initial toxicity. Initial cohorts of three subjects will be given the compound of Formula I (0.5 mg once daily) in dose increments of 100% until the first instance of grade 3 or higher toxicity ted to be drug—related in the first cycle, at which point the particular cohort will be expanded to a total of six subjects. This standard escalation schedule will be initiated in order to establish the non-tolerated dose (NTD) and MTD. Smaller increments and additional subjects within a dose cohort may also be evaluated for safety. Approximately 20 to 40 subjects will be treated and evaluated in Part A; however, the total number of subjects in Part A depends on the number of dose cohorts needed to establish the MTD. A dose will be considered the NTD when 2 or more out of 6 evaluable subjects in a cohort experience drug-related dose limiting toxicity (DLT) during Cycle 1. When the NTD is established, dose tion will stop. The MTD is defined as the last dose level below the NTD with 0 or 1 out of 6 evaluable subjects experiencing DLT during Cycle 1. An intermediate dose (i.e., one n the NTD and the last dose level before the NTD) or additional subjects within any dose cohort may be ed to more precisely ine the MTD and RPZD.
In Part 8, subjects may start dosing at the MTD and/or a lower dose leyei based on , PK andfor PD data from Part A. imately 100 subjects (up to 20 per cohort), stratified by tumor type will be treated and evaluated for safety and antitumor ty after Wiii also be every two cycles of therapy. The dose, doses, or schedule appropriate determined. During Part B, safety data will be reviewed regularly regarding the study continuation, as appropriate.
Study Population Men and women, 18 years or older, with advanced Solid Tumors (ST), Non- Hodgkin’s Lymphoma (NHL), Multiple Myeloma (MM), or advanced ctable solid tumors, including subjects who have ssed on (or not been able to tolerate) standard therapy or for whom no standard anticancer therapy exists. Selected tumor types include metastatic breast cancer (mBC), astoma multiforme (GBM), cellular carcinoma (HCC), diffuse large B—cell ma (DLBCL), and multiple myeloma (MM).
Dosing and Length ofStudy During the first cycle, only in Part A, each subject will be administered a single daily dose of the compound of Formula I on Day 1 followed by a 48-hour observation and PK sampling period, followed on Day 1 by daily uninterrupted dosing for 28 days (Cycle 1 2 30 days). In subsequent Part A cycles, subjects are treated in 28-day cycles with continuous dosing from Day 1 to 28. The Compound of Formula I will be given once or twice a day at a dose of 0.1, 0.5, 1, 2, 4 , 5, 7.5, 10, 20, 25, or 50 mg in an initial dose. The dose may be of 0.1, 0.5, 1, 2, 4 , 5, 7.5, 10 mg given once a day. The dose may be 50, 25, or mg given twice a day. The dose may be adjusted up, or down, from the starting dose during treatment. As described above, if needed, the drug may be given in a al manner.
In Part B, subjects receive uous dosing for 28 days from the beginning w there is no post initial, single dose r PK collection .
Therapy will be discontinued if there is evidence of disease progression, unacceptable toxicity or subject/physician decision to stop. Subjects may continue to receive compound without interruption for as long as they derive benefit as judged by the Investigator.
Enrollment is ed to occur over approximately 24 months. Completion of active treatment and subject follow—up is expected to take an additional 3-6 months Study Treatments Celgene Corporation will supply the compound of Formula I (HCl) as 0.1 mg, 0.5 mg, 1 mg and 3 mg capsules for oral administration. The compound will be ed in bottles inside boxes containing drug for 28 days.
U) In Part A (the dose escalation phase), the dose level will start at 0.5 mg once daily after the single PK dose. After the first dose is stered to the last subject in any cohort, subjects are observed for at least 30 days before the next higher, protocol—specified dose cohort can begin. Intra subject dose escalation is not permitted unless ed by the Safety Review Committee (SRC) which will consist of the principal investigator and Celgene’s medical monitor.
In Part B, subjects may receive the compound of a I at the MTD and/or a lower dose level, based on safety, PK and PD tions from Part A. Approximately 100 subjects (preselected tumor types in groups of up to 20) will be evaluated for safety and antitumor effects.
Overview acy Assessments Subjects will be ted for y after every 2 cycles. The primary efficacy variable is response. Tumor response will be based on se Evaluation Criteria in Solid Tumors (RECIST 1.1), International Workshop Criteria (IWC) for NHL, International Uniform Response Criteria for Multiple Myeloma (IURC) (Appendix A, Section 18.1), or Responses Assessment for Neuro-Oncology (RANO) Working Group for GBM.
Secondary/exploratory endpoints include biomarker measurements in blood and tumor, histopathologic response and correlations with pharmacogenomic findings.
Supplementary efficacy variables (eg, ECOG performance status, PET outcomes) will also be examined; in addition, hypovascularization changes will be ed by volume transfer constant (Ktrans) and initial AUC (IAUC) using DCE—MRIs.
Overview oqu/ety Assessments The safety variables for this study are adverse events, ciinieal laboratory les, lZ—lead ECGs (centrally ed), LVEF assessments, physical examinations and vital signs.
Overview ofPharmacokinetic Assessments The PK profiles of the compound of Formula I and its metabolites will be determined from seriai blood and urine collections during the first treatment cycle. These will be correlated with pharmacodynamic (PD) outcomes Where possible.
The examples set forth above are provided to give those 0f ordinary skill in the art with a te disclosure and description of how to make and use the claimed embodiments, and are not intended to limit the scope of What is disclosed herein.
Modifications that are obvious to persons of skill in the art are intended to be within the scope of the following claims. All publications; patents, and patent applications cited in this specification are incorporated herein by nce as if each such publication, patent or patent application were specifically and individually indicated to be incorporated herein by reference. ~96-

Claims (22)

What is claimed
1. Use of a therapeutically efl‘ective amount of minomethyl—4—oxo-4H— quinazolinyl)-piperidine-2,6-dione, which has the following structure: K INY\N NH: O Ono or an enantiomer or mixture of enantiomers thereof, or a pharmaceutically acceptable salt, solvate, hydrate, co-crystal, clathrate, or polymorph thereof in the manufacture of a medicament for treating or ng non-Hodgkin’s lymphoma, wherein the non-Hodgkin’s lymphoma is diffuse large B-cell lymphoma of the activated B-cell phenotype.
2. The use ofclaim 1, wherein the diffuse large B-cell lymphoma is characterized by the expression of one or more biomarkers overexpressed in RIVA, U2932, TMD8 or OCI-Lle cell lines.
3. The use ofclaim 1 or 2, wherein the non-Hodgkin’s lymphoma is relapsed or refractory.
4. The use of any one of claims 1 to 3, wherein the non-Hodgkin’s lymphoma is drug-resistant.
5. Use of a therapeutically effective amount of 3-(5-aminomethyloxo-4H- quinazolinyl)-piperidine-2,6-dione or a pharmaceutically acceptable salt, solvate or hydrate thereof in the preparation of a medicament for treating or ng dgkin’s lymphoma, wherein the patient is identified to have non-Hodgkin’s lymphoma sensitive to ent with 3- (5-aminomethyloxo-4H-quinazolin—3-yl)-piperidine-2,6-dione and the dgkin’s lymphoma is diffuse large B-cell lymphoma of the activated B-cell phenotype.
6. The use ofclaim 5, n the diffuse large B-cell lymphoma is characterized by the expression of one or more biomarkers pressed in RIVA, U2932, TMD8 or OCI-Lle cell lines.
7. The use of claim 5, wherein the non-Hodgkin’s lymphoma phenotype is identified in a biological sample obtained from a t having lymphoma.
8. The use of claim 7, wherein the biological sample is a lymph node biopsy, a bone marrow biopsy, or a sample of peripheral blood tumor cells.
9. The use of claim 5, n fying a patient having non-Hodgkin’s lymphoma sensitive to treatment with minomethyloxo-4H-quinazolinyl)- piperidine-2,6-dione, or a salt, solvate or hydrate thereof, comprises fication of a gene ated with the activated B-cell phenotype in a biological sample obtained from the patient.
10. The use of claim 9, wherein the gene associated with the activated B-cell phenotype is selected from the group consisting of IRF4/MUM1, FOXP1, SPIB, CARD11 and BLIMP/PDRM1.
11. The use of claim 5, wherein identifying a patient having non-Hodgkin’s lymphoma sensitive to treatment with 3-(5-aminomethyloxo-4H-quinazolinyl)- piperidine-2,6-dione, or a salt, solvate or hydrate thereof, comprises measuring the level of NF- κB activity in a biological sample obtained from the patient.
12. The use of claim 11, wherein the ical sample is a lymph node biopsy, a bone marrow biopsy, or a sample of peripheral blood tumor cells.
13. The use of claim 1, wherein the non-Hodgkin’s lymphoma phenotype of the t has been characterized as an activated B-cell subtype by measuring one or more of the following: (i) overexpression of SPIB, a hematopoietic-specific Ets family transcription factor required for survival of activated B-cell subtype cells; (ii) higher constitutive IRF4/MUM1 expression than GCB subtype cells; (iii) higher tutive FOXP1 expression up-regulated by trisomy 3; (iv) higher constitutive Blimp1, i.e., PRDM1, expression; (v) higher constitutive CARD11 gene expression; and (vi) an increased level of NF-κB activity relative to tivated B-cell subtype DLBCL cells.
14. The use of any one of claims 1-13, wherein the compound is 3-(5-amino methyloxo-4H-quinazolinyl)-piperidine-2,6-dione hydrochloride, or a solvate or hydrate thereof.
15. The use of any one of claims 1-14, wherein the ment is suitable for administering with a therapeutically effective amount of one or more additional active agents.
16. The use of claim 15, wherein the onal active agent is selected from the group consisting of an alkylating agent, an adenosine analog, a glucocorticoid, a kinase inhibitor, a SYK inhibitor, a PDE3 tor, a PDE7 inhibitor, doxorubicin, chlorambucil, vincristine, bendamustine, forskolin and mab.
17. The use of any one of claims 1-16, wherein 3-(5-aminomethyloxo-4H- quinazolinyl)-piperidine-2,6-dione, or a pharmaceutically able salt, solvate or hydrate thereof is for administering in an amount of from about 0.5 to about 50 mg per day; or about 0.5 to about 5 mg per day; or about 0.5, 1, 2, 4, 5, 10, 15, 20, 25 or 50 mg per day.
18. The use of claim 17, wherein the compound is for oral administration in a e or tablet.
19. The use of any one of claims 1-18, wherein the diffuse large B-cell lymphoma is relapsed, refractory or resistant to conventional therapy.
20. The use of any one of claims 1-19, wherein the compound is for administering for 21 days followed by seven days rest in a 28 day cycle.
21. A use according to claim 1 substantially as herein described or exemplified.
22. A use according to claim 5 substantially as herein described or exemplified.
NZ614493A 2011-03-11 2012-03-09 Methods of treating cancer using 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidine-2,6-dione NZ614493B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161451995P 2011-03-11 2011-03-11
US61/451,995 2011-03-11
US201161480272P 2011-04-28 2011-04-28
US61/480,272 2011-04-28
PCT/US2012/028498 WO2012125459A1 (en) 2011-03-11 2012-03-09 Methods of treating cancer using 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidine-2,6-dione

Publications (2)

Publication Number Publication Date
NZ614493A NZ614493A (en) 2016-01-29
NZ614493B2 true NZ614493B2 (en) 2016-05-03

Family

ID=

Similar Documents

Publication Publication Date Title
JP6348192B2 (en) Method for treating cancer using 3- (5-amino-2-methyl-4-oxo-4H-quinazolin-3-yl) -piperidine-2,6-dione
US20190388429A1 (en) Methods of treating cancer using 3-(4-((4-(morpholinomethyl)benzyl)oxy)-1-oxoisoindolin-2-yl)piperidine-2,6-dione
US9694015B2 (en) Methods for the treatment of locally advanced breast cancer
NZ614493B2 (en) Methods of treating cancer using 3-(5-amino-2-methyl-4-oxo-4h-quinazolin-3-yl)-piperidine-2,6-dione
US20170157103A1 (en) Methods of treating diffuse large b-cell lymphoma using 3-(4-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione