WO2020168106A1 - Combinaisons d'agonistes de protéine kinase c avec des stéroïdes ou des acides rétinoïques pour le traitement du cancer - Google Patents
Combinaisons d'agonistes de protéine kinase c avec des stéroïdes ou des acides rétinoïques pour le traitement du cancer Download PDFInfo
- Publication number
- WO2020168106A1 WO2020168106A1 PCT/US2020/018154 US2020018154W WO2020168106A1 WO 2020168106 A1 WO2020168106 A1 WO 2020168106A1 US 2020018154 W US2020018154 W US 2020018154W WO 2020168106 A1 WO2020168106 A1 WO 2020168106A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- bryostatin
- protein kinase
- agonist
- antigen
- combination
- Prior art date
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/045—Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
- A61K31/07—Retinol compounds, e.g. vitamin A
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/335—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
- A61K31/365—Lactones
- A61K31/366—Lactones having six-membered rings, e.g. delta-lactones
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/56—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
- A61K31/57—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
- A61K31/573—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/39558—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2887—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
Definitions
- This invention relates to novel compositions for use in methods for inducing expression of an antigen in a cancerous cell, for inhibiting the growth of a cancerous cell, and for treating proliferative diseases, more specifically, for treating hematologic cancers, such as leukemia.
- Antibody -based therapeutics provide highly targeted approaches for treating hematologic malignancies by specifically targeting therapeutic agents to cancerous cells that preferentially express a specific surface marker.
- Several antibody-based therapies including chimeric antigen receptor-T-cell (“CAR-T”) therapies, and therapies involving antibody drug conjugates (“ADCs”) and/or monoclonal antibody (“mAB”) have been developed for targeting a variety of markers across the spectrum of B-cell malignancies.
- CAR-T chimeric antigen receptor-T-cell
- ADCs antibody drug conjugates
- mAB monoclonal antibody
- B-cell malignancies are characterized by the presence of aberrant proliferative cells resembling healthy cells at different stages of the continuum of B-cell development (Blanc et al, Clin Cancer Res (2011) 17(20):6448-58).
- B-cell development surface markers like CD 19, CD20, and CD22 rise and increase in expression and these targets similarly are found on lymphoid malignancies.
- Antibody-based therapeutics targeting CD 19, CD20, and CD22 have been developed and are used to treat a wide range of lymphoid malignancies, e.g., CD19 CAR-T for ALL/DLBCL, CD20 mAB therapy for CLL/FL/DLBCL/MCL, and CD22 ADCs for ALL/HCL.
- Expression level of the target surface marker plays an important role in the effect of a given antibody -based therapeutic.
- the efficacy of Rituximab ® an anti-CD20 antibody, has been shown to be affected by expression level of CD20 antigens on target cells, with increased expression of CD20 on target cells showing enhanced Rituximab ® -mediated toxicity (Al-zoobi el al, Int Immunol (2014) 26(8):451-65).
- downregulation of CD20 antigen expression occurs in subsets of CLL patients that develop resistance to Rituximab ® (Small et al.
- the present invention provides compounds, combinations, compositions, medicaments, kits, and articles of manufacture useful for treating cancer, for inhibiting growth of cancer cells, for increasing expression of antigens on cancer cells and for sensitizing those cells for therapies. Further, the present invention provides methods for treating cancer, methods for inhibiting growth of cancer cells, methods for increasing expression of antigens on cancer cells and methods for sensitizing cancer cells for therapies using the compounds, combinations, compositions, medicaments, kits, and articles of manufacture of the present invention.
- a method for increasing expression of an antigen on the surface of a cancerous cell comprises the step of contacting the cancerous cell with an effective amount of a combination of an agonist of protein kinase C with a steroid or with an effective amount of a combination of an agonist of protein kinase C with a retinoic acid. Thereby expression of the antigen on the surface of the cancerous cell is increased.
- a cancerous cell is obtained from a subject having a cancer.
- a cancer is selected from the group consisting of lung cancer, sarcoma, gastrointestinal cancer, cancer of the genitourinary tract, liver cancer, skin cancer, gynecological cancer, bone cancer, cancer of the nervous system, cancer of adrenal gland, and hematologic cancer.
- the cancer is hematologic cancer.
- a hematologic cancer can be used to practice methods of the present invention.
- a hematologic cancer is selected from the group consisting of B-cell lymphoma, T-cell lymphoma, leukocytic leukemia, acute leukocytic leukemia, chronic leukocytic leukemia, lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, lymphocytic leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, monocytic leukemia, multiple myeloma, myelodysplastic syndrome, myelogenous leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, myeloid leukemia, acute myeloid leukemia, chronic myeloid leukemia, myeloproliferative disease, acute myelocytic leukemia, myelocytic leukemia, chronic my
- a hematologic cancer is acute lymphocytic leukemia. In some embodiments, a hematologic cancer is chronic lymphocytic leukemia. In some embodiments, a hematologic cancer is mixed lineage leukemia.
- an antigen is a lymphocyte antigen.
- a lymphocyte is selected from the group consisting of CDlb, CDlc,
- CD49f CD50, CD51, CD54, CD55, CD56, CD58, CD59, CD61, CD62L, CD62P, CD63, CD64, CD66a, CD66b, CD66c, CD68, CD71, CD73, CD74, CD79a, CD79b, CD80, CD83, CD84, CD85f, CD85i, CD85j, CD86, CD87, CD89, CD90, CD91, CD92, CD95, CD96, CD97, CD98, CD100, CD101, CD102, CD103, CD104, CD105, CD106, CD107a, CD107b, CD108, CD 109, CD111, CD112, CD113, CD116, CD117, CD118, CD119, CD120a, CD121a, CD121b, CD122, CD123, CD124, CD125, CD127, CD130, CD131, CD132, CD133, CD135, CD136, CD137, CD140a, CD140b, CD141, CD142, CD143, CD144,
- the cancerous cell is a cancerous B-lymphocyte cell having on its surface a B-lymphocyte antigen.
- a B-lymphocyte antigen is CD 19.
- a B-lymphocyte antigen is CD20.
- a B-lymphocyte antigen is CD22.
- a B-lymphocyte antigen is CD30.
- a B-lymphocyte antigen is CD38.
- a B-lymphocyte antigen is CD70.
- a B-lymphocyte antigen is CD269.
- a cancerous cell is contacted with an effective amount of a combination of an agonist of protein kinase C with a steroid or with an effective amount of a combination of an agonist of protein kinase C with a retinoic acid
- various agonists of protein kinase C can be used in combination with either a steroid or retinoic acid to increase expression of an antigen on the surface of the cancerous cell.
- an agonist of protein kinase C is selected from the group consisting of ingenol-3-angelate, prostratin, staurospine, valproate, tamoxifen, chelerythrine, miyabenol C, verbascoside, BIM- 1, indolocarbazole, isoquinolinesulfonamide, sphingosine, calphostin C (UCN-1028C), bisindolylmaleimide GF 109203X, a plant flavonoid, TNF-b, IL-Ib, a lipopolysaccharide,
- UV -light a CD3 antibody, a CD3/CD28 antibody combination, etoposide, daunorubicin, hydrogen peroxide, nocodazole, LIGHT, bleomycin, camptothecin, cisplatin, celecoxib, ciprofibrate, cycloprodigiosin, dacarbazine, Daio-Orengedeokuto, daunomycin, diazoxide, diclofenac, 5,6-dimethylxanthenone-4-acetic acid, flavone-8-acetic acid, haloperidol, imiquimod, isochamaejasmin, lithium, mitoxantrone, morphine, nipradilol, norepinephrine, nystatin, oltipraz, protocatechuic acid, SN38 (metabolite of CPT-11), Taxol (Paclitaxel), vinblastine, vincristine, WR1065, a bryo
- the agonist of protein kinase C is a bryostatin compound.
- a bryostatin compound is selected from the group consisting of Bryostatin-1, Bryostatin-2, Bryostatin-3, Bryostatin-4, Bryostatin-5, Bryostatin-6, Bryostatin- 7, Bryostatin-8, Bryostatin-9, Bryostatin- 10, Bryostatin-11, Bryostatin- 12, Bryostatin-13, Bryostatin- 14, Bryostatin-15, Bryostatin- 16, Bryostatin- 17, Bryostatin- 18, Bryostatin-19, Bryostatin-20, a bryostatin compound having Formula 1, a bryostatin compound having Formula 2, a bryostatin
- a cancerous cell is contacted with an effective amount of a combination of an agonist of protein kinase C with a steroid
- various steroids can be used in combination with an agonist of protein kinase C to increase expression of an antigen on the surface of the cancerous cell.
- a steroid is a glucocorticoid.
- glucocorticoids can be used in combination with an agonist of protein kinase C in a method of the present invention.
- a glucocorticoid is selected from the group consisting of dexamethasone, a dexamethasone compound having Formula 8, prednisolone, prednisone, methyl prednisolone, triamcinolone, hydrocortisone, deflazacort, betamethasone, budesonide, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives thereof.
- a retinoic acid is tretinoin, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives thereof.
- agonists of protein kinase C such as a bryostatin compound, and a steroid synergistically increase the expression of the antigen.
- agonists of protein kinase C such as a bryostatin compound, and a retinoic acid synergistically increase the expression of the antigen.
- the agonist of protein kinase C is Bryostatin- 1 and the steroid is dexamethasone.
- the agonist of protein kinase C is Bryostatin- 1 and the steroid is prednisolone.
- the agonist of protein kinase C is Bryostatin- 1 and the retinoic acid is tretinoin
- Methods for increasing expression of an antigen on the cell surface of a cancerous cell can be practiced in vitro, ex vivo and in vivo.
- the method further comprises administering the combination of the agonist of protein kinase C and the steroid to a subject in need of having expression of the antigen increased; or administering the combination of the agonist of protein kinase C and the retinoic acid to a subject in need of having expression of the antigen increased.
- the present invention also provides methods for treating a subject having cancer.
- a method for treating a subject having cancer comprises the step of administering to the subject a combination of an agonist of protein kinase C with a steroid or with a retinoic acid.
- the combination is being administered in an amount effective to increase expression of an antigen on the surface of a cancerous cell of the subject. Thereby expression of the antigen on the surface of the cancerous cell is increased.
- a cancer is selected from the group consisting of lung cancer, sarcoma, gastrointestinal cancer, cancer of the genitourinary tract, liver cancer, skin cancer, gynecological cancer, bone cancer, cancer of the nervous system, cancer of adrenal gland, and hematologic cancer.
- the cancer is hematologic cancer.
- a hematologic cancer can be used to practice methods of the present invention.
- a hematologic cancer is selected from the group consisting of B-cell lymphoma, T-cell lymphoma, leukocytic leukemia, acute leukocytic leukemia, chronic leukocytic leukemia, lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, lymphocytic leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, monocytic leukemia, multiple myeloma, myelodysplastic syndrome, myelogenous leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, myeloid leukemia, acute myeloid leukemia, chronic myeloid leukemia, myeloproliferative disease, acute myelocytic leukemia, myelocytic leukemia, chronic my
- a hematologic cancer is acute lymphocytic leukemia. In some embodiments, a hematologic cancer is chronic lymphocytic leukemia. In some embodiments, a hematologic cancer is mixed lineage leukemia.
- a cancer in a subject is treated by administering to the subject an effective amount of a combination of an agonist of protein kinase C with a steroid or with an effective amount of a combination of an agonist of protein kinase C with a retinoic acid
- various agonists of protein kinase C can be used in combination with either a steroid or retinoic acid.
- an agonist of protein kinase C is selected from the group consisting of ingenol-3-angelate, prostratin, staurospine, valproate, tamoxifen, chelerythrine, miyabenol C, verbascoside, BIM-1, indolocarbazole, isoquinolinesulfonamide, sphingosine, calphostin C (UCN-1028C), bisindolylmaleimide GF 109203X, a plant flavonoid, TNF-b, IL-Ib, a lipopolysaccharide, UV-light, a CD3 antibody, a CD3/CD28 antibody combination, etoposide, daunorubicin, hydrogen peroxide, nocodazole, LIGHT, bleomycin, camptothecin, cisplatin, celecoxib, ciprofibrate,
- cycloprodigiosin dacarbazine, Daio-Orengedeokuto, daunomycin, diazoxide, diclofenac, 5,6- dimethylxanthenone-4-acetic acid, flavone-8-acetic acid, haloperidol, imiquimod, isochamaejasmin, lithium, mitoxantrone, morphine, nipradilol, norepinephrine, nystatin, oltipraz, protocatechuic acid, SN38 (metabolite of CPT-11), Taxol (Paclitaxel), vinblastine, vincristine, WR1065, a bryostatin compound, single stereoisomers, mixtures of
- the agonist of protein kinase C is a bryostatin compound.
- bryostatin compounds can be used to practice methods for increasing expression of an antigen on the surface of a cancerous cell.
- a bryostatin compound is selected from the group consisting of Bryostatin-1, Bryostatin-2, Bryostatin-3, Bryostatin-4, Bryostatin-5, Bryostatin-6, Bryostatin- 7, Bryostatin-8, Bryostatin-9, Bryostatin- 10, Bryostatin-11, Bryostatin- 12, Bryostatin-13, Bryostatin- 14, Bryostatin-15, Bryostatin- 16, Bryostatin- 17, Bryostatin- 18, Bryostatin-19, Bryostatin-20, a bryostatin compound having Formula 1, a bryostatin compound having Formula 2, a bryostatin compound having Formula 3, a bryostatin compound having Formula 4, a bryostatin compound having Formula 5, a bryostatin compound having Formula 6, and a bryostatin compound having Formula 7, single stereoisomers, mixtures of stereoisomers, mixtures of stereoiso
- a steroid is a glucocorticoid.
- glucocorticoids can be used in combination with an agonist of protein kinase C in a method of the present invention.
- a glucocorticoid is selected from the group consisting of dexamethasone, a dexamethasone compound having Formula 8, prednisolone, prednisone, methyl prednisolone, triamcinolone, hydrocortisone, deflazacort, betamethasone, budesonide, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives thereof.
- a retinoic acid is tretinoin, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives thereof.
- the agonist of protein kinase C is Bryostatin-1 and the steroid is dexamethasone.
- the agonist of protein kinase C is Bryostatin-1 and the steroid is prednisolone.
- the agonist of protein kinase C is Bryostatin-1 and the retinoic acid is tretinoin.
- agonists of protein kinase C such as a bryostatin compound, and a steroid synergistically increase the expression of the antigen.
- agonists of protein kinase C such as a bryostatin compound, and a retinoic acid
- an antigen is a lymphocyte antigen.
- a lymphocyte is selected from the group consisting of CDlb, CDlc,
- the cancerous cell is a cancerous B-lymphocyte cell having on its surface a B-lymphocyte antigen.
- a B-lymphocyte antigen is CD 19.
- a B-lymphocyte antigen is CD20.
- a B-lymphocyte antigen is CD22.
- a B-lymphocyte antigen is CD30.
- a B-lymphocyte antigen is CD38.
- a B-lymphocyte antigen is CD70.
- a B-lymphocyte antigen is CD269.
- a method for treating a subject having cancer further comprises administering to the subject a therapeutic agent.
- the therapeutic agent is capable of specifically binding to an antigen on the cell surface of a subject’s cancerous cell. More specifically, the therapeutic agent is capable of specifically binding to the antigen on the cell surface of a subject’s cancerous cell, the expression of which had been increased by administering to the subject having cancer a combination of an agonist of protein kinase C with a steroid or a combination of an agonist of protein kinase C with a retinoic acid.
- a therapeutic agent is selected from the group consisting of an antibody mediating antibody-dependent cellular cytotoxicity, a monoclonal antibody, a humanized antibody, a chimeric antibody, a bi-specific antibody, an ADC, an antibody fragment, a recombinant antibody, and an immunotoxin.
- the therapeutic agent is a chimeric antibody.
- the chimeric antibody is Rituximab ® .
- monoclonal antibodies can be used as therapeutic agents.
- the chimeric antibody is Ofatumumab ® .
- Compounds, combinations and compositions of the present invention may be administered simultaneously or sequentially.
- a cancer in a subject is treated by administering to the subject an effective amount of a combination of an agonist of protein kinase C with a steroid
- the agonist of protein kinase C and the steroid are administered
- a cancer in a subject is treated by administering to the subject an effective amount of a combination of an agonist of protein kinase C with a retinoic acid
- the agonist of protein kinase C and the retinoic acid are administered simultaneously.
- a cancer in a subject is treated by administering to the subject an effective amount of a combination of an agonist of protein kinase C with a steroid
- the agonist of protein kinase C and the steroid are administered sequentially.
- the agonist of protein kinase C and the retinoic acid are administered sequentially.
- a cancer in a subject is treated by administering to the subject an effective amount of a combination of an agonist of protein kinase C with a steroid, and a therapeutic agent
- the agonist of protein kinase C, the steroid, and the therapeutic agent are administered simultaneously.
- the agonist of protein kinase C, the retinoic acid, and the therapeutic agent are administered simultaneously.
- a cancer in a subject is treated by administering to the subject an effective amount of a combination of an agonist of protein kinase C with a steroid, and a therapeutic agent
- the agonist of protein kinase C, the steroid, and the therapeutic agent are administered sequentially.
- the agonist of protein kinase C, the retinoic acid, and the therapeutic agent are administered sequentially.
- the present invention further provides use of a first composition comprising an agonist of protein kinase C in combination with a second composition comprising a steroid or with a third composition comprising a retinoic acid in a method for increasing expression of an antigen on the cell surface of a cancerous cell.
- a first composition comprising an agonist of protein kinase C in combination with a second composition comprising a steroid or with a third composition comprising a retinoic acid in a method for increasing expression of an antigen on the cell surface of a cancerous cell.
- Useful agonists of protein kinase C, steroids, retinoic acids and cancerous cells are provided herein and can be used in a method for increasing expression of an antigen on the cell surface of a cancerous cell.
- the present invention further provides use of a first composition comprising an agonist of protein kinase C in combination with a second composition comprising a steroid or with a third composition comprising a retinoic acid in a method for treating a subject having cancer.
- a first composition comprising an agonist of protein kinase C
- a second composition comprising a steroid
- a third composition comprising a retinoic acid
- the present invention further provides use of a combination of an agonist of protein kinase C with a steroid or with a retinoic acid in a method for increasing expression of an antigen on the cell surface of a cancerous cell.
- Useful agonists of protein kinase C, steroids, retinoic acids for preparing the combinations and cancerous cells are provided herein and can be used in a method for increasing expression of an antigen on the cell surface of a cancerous cell.
- the present invention further provides use of a combination of an agonist of protein kinase C with a steroid or with a retinoic acid in a method for treating a subject having cancer.
- a combination of an agonist of protein kinase C with a steroid or with a retinoic acid in a method for treating a subject having cancer.
- Useful agonists of protein kinase C, steroids, retinoic acids for preparing the combinations and cancers are provided herein and can be used in a method for treating a subject having cancer.
- the present invention provides pharmaceutical compositions.
- compositions may comprise an agonist of protein kinase C for use in combination with a steroid or a retinoic acid.
- Other pharmaceutical compositions comprise a steroid for use in combination with an agonist of protein kinase C.
- Yet other pharmaceutical compositions comprise a retinoic acid for use in combination with an agonist of protein kinase C.
- Useful agonists of protein kinase C, steroids and retinoic acids for preparing pharmaceutical compositions are described herein.
- an article of manufacture comprises an agonist of protein kinase C for use in combination with a steroid or for use in combination with a retinoic acid and a package insert comprising instructions for administering to a subject in need thereof the combination of the agonist of protein kinase C with the steroid or the combination of the agonist of protein kinase C with the retinoic acid.
- a package insert comprising instructions for administering to a subject in need thereof the combination of the agonist of protein kinase C with the steroid or the combination of the agonist of protein kinase C with the retinoic acid.
- an article of manufacture comprises a steroid for use in combination with an agonist of protein kinase C and a package insert comprising instructions for administering to a subject in need thereof the steroid and the agonist of protein kinase C.
- a package insert comprising instructions for administering to a subject in need thereof the steroid and the agonist of protein kinase C.
- an article of manufacture comprises a retinoic acid for use in combination with an agonist of protein kinase C and a package insert comprising instructions for administering to a subject in need thereof the retinoic acid and the agonist of protein kinase C.
- retinoic acids and agonists of protein kinase C for preparing articles of manufacture are described herein.
- the present invention further provides various uses of agonists of protein kinase C in combination with either a steroid or a retinoic acid.
- the use of an agonist of protein kinase C and a steroid or retinoic acid is for the manufacture of a medicament for use in a method for increasing expression of an antigen on the cell surface of a cancerous cell.
- the use of an agonist of protein kinase C and a steroid or retinoic acid is for the manufacture of a medicament for use in a method for treating a subject having cancer.
- Useful agonists of protein kinase C, steroids and retinoic acids for the above uses are described herein.
- a product of manufacture comprises an effective amount of a combination of an agonist of protein kinase C with a steroid.
- the combination of the agonist of protein kinase C with the steroid is in a sterile vial, ampoule or syringe.
- a product of manufacture comprises an effective amount of a combination of an agonist of protein kinase C with a retinoic acid.
- the combination of the agonist of protein kinase C with the retinoic acid is in a sterile vial, ampoule or syringe. Useful agonists of protein kinase C and retinoic acid for producing the above products of manufacture are described herein.
- FIG. 1 schematically depicts subtypes of B-cell malignancies as characterized by the stage of B-cell development as aberrant cell most resembles (see, Blanc et al, Clin Cancer Res (2011) 17(20):6448-58). Details are described herein.
- FIG. 2 shows the bryostatin family of natural products (for details, see,
- FIGs. 3A and 3B schematically depict an exemplary result of CD20 priming of abnormal B cells (defined herein as positive for B-cell marker CD 19 as well as abnormal B-cell marker CD5) obtained from a CLL patient and treated as indicated with
- FIG. 3A depicts a histogram plot of CD20 staining intensity. The dotted line represents the gMFI of the CD20 staining for the DMSO control.
- FIG. 3B depicts a graphic representation. Error bars represent the standard error of the mean (SEM). Experimental details are described herein and in Example B.
- FIGs. 4A and 4B schematically depict CD20 priming of abnormal CD20 negative B cells obtained from a CLL patient and treated as indicated with Dexamethasone (50 nM), Bryostatin-1 (5 nM), Dexamethasone (50 nM) + Bryostatin-1 (5 nM) or untreated (“Control”).
- FIG. 4A depicts a histogram plot of CD20 staining intensity. The dotted line represents the gMFI of the CD20 staining for the DMSO control.
- FIG. 4B depicts a graphic representation. Error bars represent the standard error of the mean (SEM). Experimental details are described herein and in Example C.
- FIGs. 5A-5D schematically depict CD20 priming of abnormal CD20 dim B cells obtained from dim CLL patients and treated as indicated with Dexamethasone (200 nM), Bryostatin-1 (1 nM), Dexamethasone (200 nM) + Bryostatin-1 (1 nM) or untreated (“Control”).
- FIGs. 5A-5B CD20 priming in cells obtained from Patient I (Sample ID No. 671CLL1)
- FIGs. 5C-5D CD20 priming in cells obtained from Patient II (Sample ID No. 725CLL1).
- the dotted line represents the CD20 gMFI of the vehicle control.
- error bars represent the standard error of the mean (SEM). Experimental details are described herein and in Example D.
- FIGs. 6A and 6B schematically depict CD20 priming of abnormal CD20 positive B cells obtained from a CLL patient and treated as indicated with Dexamethasone (200 nM), Bryostatin-1 (1 nM), Dexamethasone (200 nM) + Bryostatin-1 (1 nM) or untreated (“Control”).
- the dotted line represents the CD20 gMFI of the vehicle control.
- error bars represent the standard error of the mean (SEM). Experimental details are described herein and in Example E.
- FIGs. 7A and 7B schematically depict CD20 priming of abnormal B cells obtained from CLL patients and treated with Dexamethasone (200 nM), Bryostatin-1 (1 nM), Dexamethasone (200 nM) + Bryostatin-1 (1 nM) or untreated control. Untreated control is shown as a base line value for each patient sample. Patient samples are identified beneath each data bars.
- CD20 priming is expressed as log of the normalized CD20 gMFI (geometric mean fluorescent intensity; gMFI of the treated condition is divided by gMFI of the vehicle (control) treated condition). Experimental details are described herein and in Example F.
- FIGs. 8A and 8B schematically depict CD19 priming of abnormal B cells obtained from CLL patients and treated with Dexamethasone (200 nM), Bryostatin-1 (1 nM), Dexamethasone (200 nM) + Bryostatin-1 (1 nM) or untreated (“Control”). Control is shown as a base line value for each patient sample. Patient samples are identified beneath each data bars.
- CD 19 priming is expressed as log gMFI, which means the log of the normalized CD 19 gMFI (gMFI of the treated condition is divided by gMFI of the vehicle (control) treated condition). Experimental details are described herein and in Example G.
- FIGs. 9A and 9B schematically depict CD 19 priming of abnormal B cells obtained from a CLL patient and treated as indicated with Dexamethasone (200 nM), Bryostatin-1 (1 nM), Dexamethasone (200 nM) + Bryostatin-1 (1 nM) or untreated
- Control (“Control”).
- the dotted line represents the CD 19 gMFI of the vehicle control.
- error bars represent the standard error of the mean (SEM). Experimental details are described herein and in Example H.
- FIGs. 10A and 10B schematically depict CD 19 priming of abnormal B cells obtained from a B-ALL patient and treated as indicated with Dexamethasone (200 nM), Bryostatin-1 (1 nM), Dexamethasone (200 nM) + Bryostatin-1 (1 nM) or untreated
- Control (“Control”).
- the dotted line represents the CD 19 gMFI of the vehicle control.
- error bars represent the standard error of the mean (SEM). Experimental details are described herein and in Example I.
- FIGs. 11 A-l 1C schematically depict CD20 priming of CD20 positive abnormal B cells obtained from a CLL patient with a various dosing regimen of ND 1000 (as indicated).
- FIG. 11A schematically depicts a line chart.
- FIG. 11B shows fold increase of CD20 as a result of priming with the concentrations of Dexamethasone and Bryostatin-1.
- FIG. l lC schematically depicts a graphic representation.
- error bars represent the standard error of the mean (SEM). Experimental details are described herein and in Example J.
- FIGs. 12A and 12B schematically depict an unresponsive CD20 priming of abnormal CD20 dim B cells obtained from a CLL patient and treated as indicated with Dexamethasone (200 nM), Bryostatin-1 (1 nM), Dexamethasone (200 nM) + Bryostatin-1 (1 nM) or untreated (“Control”).
- the dotted line represents the CD20 gMFI of the vehicle control.
- error bars represent the standard error of the mean (SEM). Experimental details are described herein and in Example K.
- FIG. 13 schematically depicts CD22 priming of abnormal B-cells (positive for
- CD5 and CD 19 cells obtained from MCL and CLL patients and treated as indicated with Dexamethasone (200 nM), Bryostatin-1 (5 nM), Dexamethasone (200 nM) + Bryostatin-1 (5 nM) or untreated control. Untreated control is shown as a base line value for each patient sample. Patient samples are identified beneath each data bars. FIGs. 7A and 7B, CD20 priming is expressed as gMFI. Experimental details are described herein and in Example L.
- FIGs. 14A and 14B schematically depict CD22 priming of abnormal B-cells
- FIG. 15 schematically depicts CD22 priming of abnormal B-cells (positive for
- CD5 and CD19 cells obtained from an ALL patient (patient sample 825ALL1) and treated as indicated with dimethyl sulfoxide, Dexamethasone (200 nM), Tretinoin (50 nM), Bryostatin- 1 (5 nM), Bryostatin-1 (5 nM) + Dexamethasone (200 nM), Bryostatin-1 (5 nM) + Tretinoin (50 nM), or untreated control. Untreated control is shown as a base line value for each patient sample. Experimental details are described herein and in Example N.
- FIG. 16 schematically depicts CD20 priming of abnormal B-cells (positive for
- CD5 and Cdel9 cells obtained from a CLL patient and treated as indicated with
- Dexamethasone 200 nM
- Prednisolone (220 nM)
- Bryostatin-1 5 nM
- Bryostatin-1 5 nM
- Dexamethasone 200 nM
- Bryostatin-1 5 nM + Prednisolone (50 nM)
- Untreated control is shown as a base line value for each patient sample.
- compositions comprising, “include,” and“have” and variations thereof such as “comprises,” “comprising,” “includes,” “including,”“has,” and“having” are to be interpreted inclusively. That is, these words are intended to convey the possible, however, not mandatory, inclusion of other elements or integers not specifically recited, where the context allows.
- the term “consisting essentially of means that the composition, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure. No language in the specification should be construed as indicating any non- claimed element essential to the practice of the invention.
- each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value 10 is disclosed, then “about 10 is also disclosed. It is also understood that when a value is disclosed that is “less than or equal to the value” or “greater than or equal to the value” possible ranges between these values are also disclosed, as appropriately understood by the skilled artisan. For example, if the value “ 10" is disclosed the “less than or equal to 10 "as well as "greater than or equal to 10" is also disclosed.
- first feature or a first element can be the same as a second feature or a second element.
- a first feature or first element discussed herein could be termed a second feature or second element, and similarly, a second feature or second element described herein could be termed a first feature or first element without departing from the teachings of the present invention.
- the terms "to administer, " “administering,” “administered” or grammatical equivalents thereof mean the actual physical introduction of a composition into or onto (as appropriate) a host or a cell. Any and all methods of introducing the composition into the host or cell are contemplated according to the invention; the method is not dependent on any particular means of introduction and is not to be so construed. Means of introduction, such as injecting, inhaling, ingesting, absorbing, implanting, and the like, are well-known to those skilled in the art, and also are exemplified herein.
- contacting in“combination,”“contacting a combination,” administration "in combination,”“administering a combination,” “co-administration” or grammatical equivalents thereof refer to both simultaneous and sequential contacting or both simultaneous and sequential administration of two or more compounds or two or more compositions.
- Concurrent or combined administration to a subject means that two or more compounds or two or more compositions are administered to a subject either (a) simultaneously, or (b) at different times during the course of a common treatment schedule. In the latter case, the two or more compounds or compositions are administered sufficiently close in time to achieve the intended effect. Concurrent or combined
- compositions comprising one or more agonists of protein kinase C, a composition comprising one or more steroid compounds, or a composition comprising one or more retinoic acid compounds can be administered in combination with a another composition comprising one or more therapeutic agents.
- Concurrent or combined administration also means that a one or more agonists of protein kinase C, one or more steroid compounds, or one or more retinoic acid compounds can be administered in combination with one or more therapeutic agents.
- agent and “compound” are used interchangeably herein and mean any chemical compound, for example, a macromolecule or a small molecule disclosed herein.
- An agent can have a formula weight of less than about 100,000 grams per mole, less than about 50,000 grams per mole, less than about 10,000 grams per mole, less than 5,000 grams per mole, less than 1,000 grams per mole, or less than about 500 grams per mole.
- An agent can be naturally occurring (e.g., a herb or a nature product), synthetic, or both.
- macromolecules are proteins, protein complexes, and glycoproteins, nucleic acids, e.g., DNA, RNA and PNA (peptide nucleic acid).
- small molecules are peptides, peptidomimetics (e.g., peptoids), amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, nucleotides, nucleotide analogs, organic or inorganic compounds, e.g., heteroorganic or organometallic compounds.
- An agent can be the only substance used in a method described herein. Alternatively, a collection of agents can be used either consecutively or concurrently in methods described herein. Likewise, an agent can be the only substance used in a composition or kit described herein. Alternatively, a collection of agents can be used in a combination, a composition, a kit or an article of manufacture as described herein.
- agonist refers to an agent that causes an increase in the expression or activity of a target gene or protein, respectively.
- An agonist can bind to and activate its cognate target in some fashion, which directly or indirectly brings about a physiological effect on the target gene or protein.
- Agonists include agents that, e.g., induce or activate the expression of a polypeptide of the invention or bind to, stimulate, increase, open, activate, facilitate, or enhance activation, sensitize or up-regulate the activity of a polypeptide of the invention.
- protein kinase C agonist refers to a compound that causes activation of protein kinase C.
- Such agonists include nucleic acids such as siRNA, antisense RNA, and ribozymes that increase or activate the expression of protein kinase C as well as naturally occurring and synthetic compounds and agents, small chemical molecules and the like.
- Preferred protein kinase C agonists are the compounds described herein.
- an activation or increased activity of protein kinase C by a disclosed composition can be determined by assaying the activity of protein kinase C in the presence of the composition and comparing it to the activity of protein kinase C in the absence of the composition.
- the composition can be said to increase the activity of protein kinase C.
- samples or assays comprising e.g., a protein kinase C polypeptide that are treated with an agonist (or a potential agonist) are compared to control samples without the agonist to examine the extent of the effect.
- Control samples (untreated with agonists) are assigned a relative activity value of 100%.
- Activation of the protein kinase C polypeptide is achieved when the level or activity value relative to a control is increased by about 20%, by about 30%, by about 40%, by about 50%, by about 60%, by about 70%, by about 80%, by about 90, by about 100%, by about 150%, by about 200%, by about 250%, by about 300%, by about 350%, by about 400%, by about 500%, by about 750%, by about 1000% or more.
- activation of the protein kinase C polypeptide is achieved when the level or activity value relative to a control is increased by at least about 20%, by at least about 30%, by at least about 40%, by at least about 50%, by at least about 60%, by at least about 70%, by at least about 80%, by at least about 90, by at least about 100%, by at least about 150%, by at least about 200%, by at least about 250%, by at least about 300%, by at least about 350%, by at least about 400%, by at least about 500%, by at least about 750%, by at least about 1000% or more.
- Assays for agonists of protein kinase C activity are known in the art.
- ALL refers to acute lymphocytic leukemia and includes B-cell ALL (B-ALL) and T-cell ALL (T-ALL).
- AML refers to acute myelocytic leukemia and includes B-cell AML (B-AML) and T-cell AML (T-AML).
- B-AML B-cell AML
- T-AML T-cell AML
- antigen refers to a molecule or composition of matter, which induces an immune response in an animal and which interacts specifically with antigen-recognizing components of an animal's immune system. Included within the term are endogenous antigens (self-antigens, i.e., antigens expressed by an isolated animal cell or by a cell within an animal) and exogenous antigens.
- An exogenous antigen refers to a substance that, upon introduction into a vertebrate animal, stimulates the production of antibodies.
- an "assay” refers to a test performed on a sample or a component of a sample.
- An assay can test for the presence of a component, the amount or concentration of a component, the composition of a component, the activity of a component, etc.
- Assays that can be performed in conjunction with the compositions and methods of the present invention include, but are not limited to, immunocytochemical assays, flow cytometry, fluorescence-activated cell sorting (FACS), interphase FISH (fluorescence in situ hybridization), karyotyping, immunological assays, biochemical assays, binding assays, cellular assays, genetic assays, gene expression assays and protein expression assays.
- a preferred component assayed for is an antigen, preferably, a lymphocyte antigen, more preferably, a B-lymphocyte antigen.
- biologically active when referring to an agent is art- recognized and refers to a form of an agent that allows for it, or a portion of the amount of agent administered, to be absorbed by, incorporated to, or otherwise physiologically available to a subject or patient to whom it is administered.
- biological sample means a sample of biological tissue, cells or fluid that contains nucleic acids or polypeptides. Such samples are typically from humans, but include tissues, cells or fluids isolated from non-human primates, or rodents, e.g., mice, and rats. Biological samples may also include sections of tissues such as biopsy and autopsy samples, frozen sections taken for histological purposes, cerebral spinal fluid, blood, plasma, serum, sputum, stool, tears, mucus, hair, skin, etc. Biological samples also include explants and primary and/or transformed cell cultures derived from patient tissues. A “biological sample” also refers to a cell or population of cells or a quantity of tissue or fluid from an animal.
- biological sample can also refer to cells or tissue analyzed in vivo, i.e., without removal from the animal.
- a biological sample will contain cells from the animal, but the term can also refer to non-cellular biological material, such as non-cellular fractions of blood, serum, saliva, cerebral spinal fluid or urine, that can be used to measure expression level of a polynucleotide or polypeptide.
- non-cellular biological material such as non-cellular fractions of blood, serum, saliva, cerebral spinal fluid or urine, that can be used to measure expression level of a polynucleotide or polypeptide.
- Numerous types of biological samples can be used in the present invention, including, but not limited to, a tissue biopsy or a blood sample.
- tissue biopsy refers to an amount of tissue, such as a lung tissue, removed from an animal, preferably a human, for diagnostic analysis.
- tissue biopsy can refer to any type of biopsy, such as needle biopsy, fine needle biopsy, surgical biopsy, etc.
- providing a biological sample means to obtain a biological sample for use in a method described herein. Most often, this will be done by removing a sample of tissue, cells, or fluid from a subject, but can also be accomplished by using previously isolated tissue(s), cell(s) or fluid(s) (e.g., isolated by another person, at another time, and/or for another purpose), or by performing a method of the invention in vivo. Archival tissues, cells or fluids having treatment or outcome history, will be particularly useful.
- B-lymphocyte cell refers to a lymphocyte cell that is produced in the bone marrow, produces
- a B-lymphocyte cell includes a B stem cell, a pro-B cell, a pre-B cell, a naive B cell, an activated B cell, a memory GC B cell, a late plasmablast cell and a plasma cell.
- B-lymphocyte antigen As used herein, the terms "B-lymphocyte antigen” "antigen in a B-lymphocyte cell” or “antigen on the cell surface of a B-lymphocyte cell” refer to an antigen expressed in a B-lymphocyte cell.
- a preferred B-lymphocyte antigen is an antigen located on the cell surface of a B-lymphocyte cell, which may also be referred to as B-lymphocyte surface marker.
- Exemplary B-lymphocyte cell surface markers include, but are not limited to, CD 10, CD 19, CD20, CD21, CD22, CD23, CD24, CD37, CD53, CD72, CD73, CD74, CDw75, CDw76, CD77, CDw78, CD79a, CD79b, CD80, CD81, CD82, CD83, CDw84, CD85 and CD86 leukocyte surface markers.
- a B-lymphocyte surface marker of particular interest is preferentially expressed on B-cells compared to other non-B-cell tissues of a mammal and may be expressed on both precursor B-cells and mature B-cells.
- a B-cell surface marker is one, like CD20 or CD 19, which is found on B- cells throughout differentiation of the lineage from the stem cell stage up to a point just prior to terminal differentiation into plasma cells.
- Preferred B-cell surface markers herein are CD 19 and CD20.
- cancer refers to a cell that exhibits deregulated growth and, in most cases, has lost at least one of its differentiated properties, such as, but not limited to, characteristic morphology, non-migratory behavior, cell-cell interaction and cell-signaling behavior, protein expression and secretion pattern, and the like.
- a cancer is "responsive" to a therapeutic agent if its rate of growth is inhibited as a result of contact with the therapeutic agent, compared to its growth in the absence of contact with the therapeutic agent.
- Growth of a cancer can be measured in a variety of ways, for instance, the size of a tumor, the number of tumor cells in a sample, or the expression of tumor markers appropriate for that tumor type may be measured. Still further, measures of responsiveness can be assessed using additional criteria beyond growth size of a tumor, including patient quality of life, degree of metastases, etc.
- clinical prognostic markers and variables can be assessed (e.g., M protein in myeloma, PSA levels in prostate cancer) in applicable situations.
- a cancer is "non-responsive" to a therapeutic agent if its rate of growth is not inhibited, or inhibited to a very low degree, as a result of contact with the therapeutic agent when compared to its growth in the absence of contact with the therapeutic agent.
- growth of a cancer can be measured in a variety of ways, for instance, the size of a tumor, the number of tumor cells in a sample, or the expression of tumor markers appropriate for that tumor type may be measured.
- carrier in the context of “pharmaceutically acceptable carrier” refers to an inert substance used as a diluent, adjuvant, excipient or vehicle with which a compound, agent, drug, medicament, or vaccine is administered.
- cell surface marker refers to any antigen or receptor on the surface of a cell to which an antibody, an antibody fragment or ligand specifically binds.
- the term "cellular assay” refers to an assay that tests for a cellular process, such as, but not limited to, a metabolic activity, a catabolic activity, an ion channel activity, an intracellular signaling activity, a receptor-linked signaling activity, a transcriptional activity, a translational activity, or a secretory activity.
- CLL refers to chronic lymphocytic leukemia and includes B-cell CLL and T-cell CLL.
- CML chronic myelocytic leukemia and includes B-cell CML and T-cell CML.
- contacting can include contacting a substance, such as a cell, to a composition or therapeutic agent as described herein.
- a cell can be contacted with the composition or therapeutic agent, for example, by adding the composition or therapeutic agent to the culture medium (e.g., by continuous infusion, by bolus delivery, or by changing the medium to a medium that contains the composition or therapeutic agent) or by adding the composition or therapeutic agent to an extracellular fluid in vivo (e.g., by local delivery, systemic delivery, intravenous injection, bolus delivery, or continuous infusion).
- the duration of contact with a cell or group of cells may be determined by one of ordinary skill in the art by the time the composition or therapeutic agent is present at physiologically effective levels or at presumed physiologically effective levels in the medium or extracellular fluid bathing the cell.
- a lymphocyte cell expressing a lymphocyte antigen or a B-lymphocyte cell expressing a B-lymphocyte antigen is contacted with a composition or therapeutic agent in an effective amount.
- the term“contacting” is used herein interchangeably with the following: combined with, added to, mixed with, passed over, incubated with, flowed over, placed in direct physical association with another substance, and the like.
- contacting "in combination” refers to both simultaneous and sequential exposure of two or more substances to another substance.
- Concurrent or combined contacting of a cell means that two or more agents are contacted to a cell either (a) simultaneously, or (b) at different times during the course of a common schedule.
- Concurrent or combined contacting of a cell also means that a first composition comprising one or more bryostatin compounds and one or more dexamethasone compounds can be contacted to a cell in combination with a second composition comprising one or more therapeutic agents.
- control refers to a biological sample from a healthy patient or a biological sample from a patient not having the disease.
- cytotoxic agent refers to a substance that inhibits or prevents the function of a cell and/or causes destruction of a cell.
- the term is intended to include radioactive isotopes (e.g., I 113 , Y 90 , Ar 211 , P 32 , Re 188 , Re 186 , SM 153 , B 212 and others) and toxins, such as enzymatically active toxins of bacterial, fungal, plant or animal origin, or cytotoxic fragments thereof.
- cytotoxic T-lymphocyte refers to a type of lymphocyte that matures in the thymus and has the ability to recognize specific peptide antigens, or specific peptide antigens complexed with a major histocompatibility complex protein (MHC), through the receptors on its cell surface.
- CTLs can kill cells that are infected by a specific virus or other intracellular microbe or cells that present a specific antigen on their surface.
- the term "derivative” refers to a compound that is derived from a similar compound or a compound that can be imagined by one of ordinary skill in the art to arise from another compound, if one atom or group of atoms is replaced with another atom or group of atoms.
- a hydrogen atom of a compound may be substituted by an alkyl, an acyl, an amino, a hydroxyl, a halo, a haloalkyl, and the like, to produce a derivative of that compound or a derivatized compound.
- a “functional derivative” refers to a derivative of a compound that substantially functions as the compound from which it was derived.
- a “physiologically functional derivative” refers to any pharmaceutically acceptable derivative of a compound of the present invention, for example an ester or an amide thereof, and includes any pharmaceutically acceptable salt, ester, or salt of such ester of a compound which, upon administration to a mammal, such as a human, is capable of providing (directly or indirectly) a compound as described herein or an active metabolite or residue thereof. It will be appreciated by those skilled in the art that compounds described herein may be modified to provide physiologically functional derivatives thereof at any of the functional groups in those compounds, and that the compounds described herein may be so modified at more than one position.
- Synonyms of the term "determining" are contemplated within the scope of the present invention and include, but are not limited to, detecting, measuring, assaying, or testing for the presence, absence, amount or concentration of a nucleic acid, a polypeptide, a molecule, a label, or a small molecule of the invention and the like. The term refers to both qualitative and quantitative determinations.
- determining an amount refers to detecting, measuring, testing the presence, absence, amount or concentration of a molecule.
- a preferred molecule is an antigen, preferably a lymphocyte antigen, more preferably, a B-lymphocyte antigen.
- determining an effect or “determining the functional effect” means assaying for an agent that increases or decreases a parameter that is indirectly or directly under the influence of the agent, e.g., functional, enzymatic, physical and chemical effects.
- Such effects can be measured by any means known to those skilled in the art, e.g., changes in spectroscopic characteristics (e.g., fluorescence, absorbance, refractive index), hydrodynamic (e.g., shape), chromatographic, or solubility properties for the protein, measuring inducible markers or transcriptional activation of a gene, such as a gene encoding a DNA repair enzyme, measuring binding activity, measuring cellular proliferation, measuring apoptosis, measuring subcellular localization of a polypeptide, such as a DNA repair enzyme, or the like.
- spectroscopic characteristics e.g., fluorescence, absorbance, refractive index
- hydrodynamic e.g., shape
- chromatographic, or solubility properties for the protein e.g., changes in spectroscopic characteristics (e.g., fluorescence, absorbance, refractive index), hydrodynamic (e.g., shape), chromatographic, or solubility properties for the protein, measuring inducible markers or transcriptional activation of
- Determination of the functional effect of an agent on a disease, disorder, cancer or other pathology can also be performed using assays known to those of skill in the art such as in vitro assays, e.g., cellular proliferation; growth factor or serum dependence; mRNA and protein expression in cells, and other characteristics of cells.
- assays known to those of skill in the art such as in vitro assays, e.g., cellular proliferation; growth factor or serum dependence; mRNA and protein expression in cells, and other characteristics of cells.
- the effects can be evaluated by many means known to those skilled in the art, e.g., microscopy for quantitative or qualitative measures of alterations in morphological features, measurement of changes in RNA or protein levels, measurement of RNA stability, identification of downstream or reporter gene expression (CAT, luciferase, b-gal, GFP and the like), e.g., via chemiluminescence, fluorescence, colorimetric reactions, antibody binding, inducible markers, ligand binding assays, apoptosis assays, measuring the production of acetyl-CoA and AMP, and the like.
- “Functional effects” include in vitro, in vivo, and ex vivo activities.
- the term “different” means not the same, not of the same identity.
- the terms “disorder”, “disease” or “pathological condition” are used inclusively and refer to any deviation from the normal structure or function of any part, organ or system of the body (or any combination thereof).
- a specific disease is manifested by characteristic symptoms and signs, including biological, chemical and physical changes, and is often associated with a variety of other factors including, but not limited to, demographic, environmental, employment, genetic and medically historical factors. Certain characteristic signs, symptoms, and related factors can be quantitated through a variety of methods to yield important diagnostic information.
- a preferred “disorder,” “disease,” or “pathological condition” amenable to prevention and/or treatment using compositions and methods described herein, is cancer.
- the terms "dosage unit,” or simply “dose” or “dosage” refer to discrete, predetermined quantities of a compound that can be administered as unitary dosages to a subject.
- a predetermined quantity of an active compound can be selected to produce a desired therapeutic effect and can be administered with a pharmaceutically acceptable carrier.
- the predetermined quantity in each unit dosage can depend on factors that include, but are not limited to, (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of creating and administering such dosage units.
- the terms "effective amount,” “effective dose,” “sufficient amount,” “amount effective to,” “therapeutically effective amount,” “amount effective,” “dose effective,” “amount sufficient,” “amount therapeutically effective,” or grammatical equivalents thereof mean a dosage sufficient to produce a desired result, to ameliorate, or in some manner, reduce a symptom or stop or reverse progression of a condition and provide either a subjective relief of a symptom(s) or an objectively identifiable improvement as noted by a clinician or other qualified observer.
- Amelioration of a symptom of a particular condition by administration of a pharmaceutical composition described herein refers to any lessening, whether permanent or temporary, lasting or transit that can be associated with the administration of the pharmaceutical composition.
- an “effective amount” can be administered in vivo, ex vivo, and in vitro.
- excipient or“carrier” refer to an inert substance used as a diluent or vehicle in a pharmaceutical composition or for administering a pharmaceutical composition.
- the term "immunotoxin” refers to a molecule comprising a targeted toxin and a targeting moiety.
- the toxin is targeted to a desired cell by the targeting moiety, which retains antigen recognition and binding capability.
- the terms “increasing,” “increased,” and grammatical equivalents thereof refer to a level, amount, concentration of a parameter, such as a chemical compound, a metabolite, a nucleic acid, a polypeptide or a physical parameter (absorption, half-life, pH, temperature, viscosity, etc.) measured in a sample, such as a biological sample.
- a parameter such as a chemical compound, a metabolite, a nucleic acid, a polypeptide or a physical parameter (absorption, half-life, pH, temperature, viscosity, etc.) measured in a sample, such as a biological sample.
- such level, amount, or concentration of a parameter has an increase of at least about 10%, at least about 20%, at least about 30%, preferably at least about 50%, more preferable at least about 75%, and still more preferably an increase of more than 100% when compared to the level, amount, or concentration of the same chemical compound, metabolite, nucleic acid, polypeptide or physical parameter in a control sample.
- such level, amount, or concentration of a parameter has an increase in the range of at least about 2-fold up to more than about 1,000-fold when compared to the level, amount, or concentration of the same chemical compound, metabolite, nucleic acid, polypeptide or physical parameter in a control sample.
- such level, amount, or concentration of a parameter has an increase at least about 3 -fold, at least about 4-fold, at least about 5-fold, at least about 10-fold, at least about 15-fold, at least about 20-fold, at least about 30-fold, at least about 40-fold, at least about 50-fold, at least about 60-fold, at least about 70-fold, at least about 80-fold, at least about 90-fold, at least about 100-fold, at least about 200-fold, at least about 300-fold, at least about 400-fold, at least about 500-fold, at least about 600-fold, at least about 700-fold, at least about 800-fold, at least about 900-fold, at least about 1,000-fold, when compared to the level, amount, or concentration of the same chemical compound, metabolite, nucleic acid, polypeptide or physical parameter in a control sample.
- a preferred polypeptide is an antigen, preferably, a lymphocyte antigen, more preferably, a B-lymphocyte antigen.
- the terms “inhibition,” “inhibiting,” “inhibits” or grammatical equivalents thereof means to reduce an activity as compared to a control (e.g., activity or parameter in the absence of such inhibition). It is understood that inhibition can mean a slight reduction in activity to the complete ablation of all activity.
- the terms "inhibiting the growth of a cell,” “inhibiting the growth of a population of cells” or grammatical equivalents thereof refer to inhibiting cell division and may include destruction of the cell.
- the terms also refer to any inhibition in cell growth and proliferation characteristics in vitro, ex vivo, or in vivo of a cell, preferably a cancerous cell, such as inhibiting formation of foci, inhibiting anchorage independence, inhibiting semi-solid or soft agar growth, inhibiting loss of growth factor or serum requirements, inhibiting changes in cell morphology, inhibiting immortalization, inhibiting expression of tumor specific markers, and/or inhibiting formation of tumors of the cell.
- Preferred herein is inhibiting the growth of a cancerous cell, more preferred, is inhibiting the growth of a cancerous lymphocyte cell, and even more preferred is inhibiting the growth of a cancerous B-lymphocyte cell.
- the terms also refer to inhibiting an increase in cellular volume of a cell or inhibiting an increase in cell size of a cell.
- in vitro means outside the body of the organism from which a cell or cells is obtained or from which a cell line is isolated.
- Cell culture, or culture typically refers to the maintenance of cells in an artificial, in vitro environment, or the maintenance of cells in an external, ex vivo environment (i.e., outside of an organism), and can include the cultivation of individual cells and tissues.
- Certain cell culture systems described herein may be an ex vivo environment and/or an in vitro environment.
- in vivo means within the body of the organism from which a cell or cells is obtained or from which a cell line is isolated.
- the term "isomer” refers to a compound that has the same molecular formula as another compound, but differs therefrom in the nature or sequence of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed “stereoisomers.” “Stereoisomer” and “stereoisomers” refer to compounds that exist in different stereoisomeric forms if they possess one or more asymmetric centers or a double bond with asymmetric substitution and, therefore, can be produced as individual stereoisomers or as mixtures. Stereoisomers include enantiomers and diastereomers.
- stereoisomers that are not mirror images of one another are termed “diastereomers” and those that are non-superimposable mirror images of each other are termed “enantiomers.”
- enantiomers When a compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible.
- An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as (+) or (-)-isomers respectively).
- a chiral compound can exist as either individual enantiomer or as a mixture thereof.
- a mixture containing equal proportions of the enantiomers is called a "racemic mixture.”
- the description of compounds herein is intended to include individual stereoisomers as well as mixtures. Methods for determination of stereochemistry and separation of stereoisomers are well-known in the art (see, discussion in Chapter 4 of Advanced Organic Chemistry, 4th edition J. March, John Wiley and Sons, New York, 1992).
- level of an mRNA in a biological sample refers to the amount of mRNA transcribed from a gene that is present in a cell or a biological sample.
- the mRNA generally encodes a functional protein, although mutations may be present that alter or eliminate the function of the encoded protein.
- a "level of mRNA” need not be quantified, but can simply be detected, e g., a subjective, visual detection by a human, with or without comparison to a level from a control sample or a level expected of a control sample.
- a preferred mRNA is an mRNA transcribed from a gene encoding an antigen, preferably, a lymphocyte antigen, more preferably, preferably, a B-lymphocyte antigen.
- level of a polypeptide in a biological sample refers to the amount of polypeptide translated from an mRNA that is present in a cell or biological sample.
- the polypeptide may or may not have protein activity.
- a "level of a polypeptide” need not be quantified, but can simply be detected, e g., a subjective, visual detection by a human, with or without comparison to a level from a control sample or a level expected of a control sample.
- a preferred polypeptide is an antigen, preferably, a lymphocyte antigen, more preferably, a B-lymphocyte antigen.
- lymphocyte cell refers to a subtype of white blood cell in a vertebrate's immune system. Lymphocytes include natural killer (NK) cells, which function in cell-mediated, cytotoxic innate immunity, T-cells for cell- mediated, cytotoxic adaptive immunity, and B-cells for humoral, antibody-driven adaptive immunity.
- NK natural killer
- B-cell, B-lymphocyte, and B-lymphocyte cell are used herein interchangeably.
- T-cell, T-lymphocyte, and T-lymphocyte cell are used herein interchangeably.
- mammalian means or relates to the class mammalia including the orders carnivore (e.g., dogs and cats), rodentia (e.g., mice, guinea pigs, and rats), and primates (e.g., humans, chimpanzees, and monkeys).
- carnivore e.g., dogs and cats
- rodentia e.g., mice, guinea pigs, and rats
- primates e.g., humans, chimpanzees, and monkeys.
- mammalian cell refers to a cell derived from a mammal including, but not limited to, human, rat, mouse, guinea pig, chimpanzee, or macaque.
- a mammalian cell may be cultured in vivo, ex vivo, or in vitro.
- modulate As used herein, the terms “modulate,” “modulation,” “modulating” and grammatical equivalents thereof are art-recognized and refer to up-regulation (i.e., activation, stimulation, increase), or down regulation (i.e., inhibition, suppression, reduction, or decrease) of a response, or the two in combination or apart.
- up-regulation i.e., activation, stimulation, increase
- down regulation i.e., inhibition, suppression, reduction, or decrease
- the term “modulate” encompasses “increase,” stimulate,” or “activate” and “decrease,” “inhibit,” “suppress,” “reduce,” or “decrease.”
- agents that increase gene expression of an antigen preferably, a lymphocyte antigen, more preferably, a B- lymphocyte antigen, and/or which increase a level of an antigen, preferably, a lymphocyte antigen polypeptide in the lymphocyte cell or on the cell surface of the lymphocyte cell, more preferably, a B-lymphocyte antigen polypeptide in a B-lymphocyte cell or on the cell surface of the B-lymphocyte cell, and/or which increase a level of an antigen mRNA, preferably, a lymphocyte antigen mRNA, and more preferably, a B-lymphocyte antigen mRNA, in a cell.
- a "modulator" of the level or activity of an antigen preferably, a lymphocyte antigen, more preferably, a B-lymphocyte antigen, or of a gene encoding an antigen, preferably, a lymphocyte antigen, more preferably, a B-lymphocyte antigen, includes an activator and/or inhibitor of that gene or polypeptide and is used to refer to a compound that activates or inhibits the level of expression of the gene or polypeptide or an activity of the gene or polypeptide.
- the term “pharmaceutically acceptable” refers to compositions that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction when administered to a subject, preferably a human subject.
- pharmaceutically acceptable means approved by a regulatory agency of a federal or state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
- polypeptide and “protein” are used interchangeably herein to refer to a polymer of amino acid residues.
- the terms also apply to amino acid polymers in which one or more amino acid residues is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers, those containing modified residues, and non-naturally occurring amino acid polymers.
- Preferred polypeptides are antigens, preferably, lymphocyte antigens, even more preferred are El ly mphocyte antigens.
- the term "population of cells” refers to cells, preferably mammalian cells, more preferably human cells, grown in vitro, ex vivo, or in vivo.
- the term also refers to cells within a host and may comprise a mixture of cells, such as lymphocyte cells and non-lymphocyte cells or cancer cells and non-cancer cells.
- Preferred population of cells include, but are not limited to, e.g., a population of lymphocyte cells, a population of lymphocyte cells within a host, a population of lymphocyte cells expressing a lymphocyte antigen, a population of lymphocyte cells expressing a lymphocyte antigen and within a host, a population of B-lymphocyte cells, a population of B-lymphocyte cells within a host, a population of B-lymphocyte cells expressing a B-lymphocyte antigen, a population of B- lymphocyte cells expressing a B-lymphocyte antigen and within a host, a population of B- lymphocyte cells expressing CD19, a population of B-lymphocyte cells expressing CD19 and within a host, a population of B-lymphocyte cells expressing CD20, a population of B- lymphocyte cells expressing CD20 and within a host, a population of B-lymphocyte cells expressing CD22, and a population of B-lymphocyte cells expressing CD22 and within a host.
- precancerous cell refers to a cell characterized by uncontrolled, abnormal growth or a cell derived from such a cell.
- precancerous cell includes, for example, a primary precancerous cell obtained from a patient with precancerous disorder or cell line derived from such a cell or a cancer stem cell.
- a hematological precancerous cell refers to a precancerous cell deriving from a blood cell or bone marrow cell. In one embodiment, the hematological precancerous cell is a
- precancerous disorder refers to a group of hyperproliferative disorders that can develop into cancer, including for example precancerous blood disorders, such as myeloproliferative disease or myelodysplastic syndrome which is a premalignant condition that is related to and/or can develop into acute myeloid leukemia (AML).
- precancerous blood disorders such as myeloproliferative disease or myelodysplastic syndrome which is a premalignant condition that is related to and/or can develop into acute myeloid leukemia (AML).
- AML acute myeloid leukemia
- the terms“priming,”“to prime,”“primed,” or grammatical equivalents thereof refer to contacting a cell with a compound resulting in the up-regulation of a gene or of a polypeptide, preferably the up-regulation of a gene or of a polypeptide to which a therapeutic agent can bind.
- prodrug refers to a compound, which is a drug precursor and which, following administration and absorption, releases the drug in vivo via some metabolic process.
- Prodrugs of compounds described herein include, but are not limited to, e.g., phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, 3-lactam-containing prodrugs, optionally substituted phenoxyacetamide-containing prodrugs or optionally substituted phenylacetamide-containing prodrugs, 5 fluorocytosine and other 5-fluorouridine prodrugs.
- a "prodrug” as used in this application when referring to a precursor or derivative form of a pharmaceutically active substance is less cytotoxic to tumor cells compared to the parent drug and is capable of being enzymatically activated or converted into the more active parent form.
- the term “salt” refers to a salt of a compound which may be prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds described herein contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
- Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
- acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
- Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric,
- monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like.
- salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see.
- a preferred salt is a pharmaceutically acceptable salt.
- the neutral forms of a compound may be regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner.
- the parent form of the compound may differ from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
- binding partner e.g., an antibody, and the like
- a binding partner e.g., an antibody, and the like
- selective reactivity may be distinguished as mediated through specific recognition of the antigen.
- specific binding results in a much stronger association between the antibody (or other binding partners) and cells bearing the antigen than between the bound antibody (or other binding partners) and cells lacking the antigen.
- Specific binding typically results in greater than 2 fold, preferably, greater than 5 fold, more preferably, greater than 10 fold, and, most preferably, greater than 100-fold increase in amount of bound antibody (or other binding partners), per unit time, to a cell or tissue bearing a target antigen as compared to a cell or tissue lacking the target antigen.
- Specific binding to a protein under such conditions requires an antibody (or other binding partner) that is selected for its specificity for a particular protein.
- a variety of immunoassay formats are appropriate for selecting antibodies specifically immunoreactive with a particular protein.
- solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies (or other binding partners) specifically immunoreactive with a protein. See, Harlow & Lane, ANTIBODIES, A LABORATORY MANUAL, Cold Spring Harbor Publications, New York (1988), for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
- small molecule refers to a molecule having a molecular weight of less than about 5,000, less than about 2,000, less than about 1,000, or less than about 500 Da (or g/mol). In many embodiments, such small molecules do not include a peptide bond or a phosphodiester bond. For example, they can be non-polymeric. In some embodiments, a small molecule has a molecular weight of at least about 50, at least about 100, at least about 200, or at least about 400 Dalton. A preferred small molecule is dexamethasone having a molecular weight of about 392 g/mol. Another preferred small molecule is bryostatin having a molecular weight of about 905 g/mol.
- the terms "subject,” “host,” “individual,” and “patient,” are used interchangeably and refer to a mammal, including, but not limited to, murines, simians, felines, canines, equines, bovines, mammalian farm animals, mammalian sport animals, and mammalian pets and humans. Preferred is a human.
- "subject” or “patient” to be treated for a pathological condition, disorder, or disease by a subject method means either a human or non-human animal in need of treatment for a pathological condition, disorder, or disease.
- Subjects include, males and females of any age group (infant, child, adolescent, young adult, middle-aged adult, or senior adult).
- “synergizing,” “synergistically,” or grammatical equivalents thereof mean an interaction of two or more agents such that the effect when combined is greater than the predicted effect based on the response of each agent applied separately.
- a therapeutic synergism means a therapeutic effect achieved with a tolerated regimen of the combined agents that exceeds the optimal effect achieved at any tolerated dose of monotherapy.
- a synergistic effect can be about two-fold, about three-fold, about five-fold, about ten-fold or more when compared to the additive effect of the individual agents.
- Synergy, synergism or synergistic effects of a combination of agents can be determined as described in Ting-Chao Chou,“ Theoretical Basis, Experimental Design, and Computerized Simulation of Synergism and Antagonism in Drug Combination Studies”, Pharmacol. Rev. (2006) 58:621-681, which is incorporated herewith by reference in its entirety.
- a determination of a synergistic interaction between a compound or a pharmaceutically acceptable salt thereof and one or more agents may be based on the results obtained from assays described herein.
- the results of those assays can be analyzed, e.g., using the Chou and Talalay combination method and Dose-Effect Analysis with CalcuSyn software in order to obtain a Combination Index (Chou and Talalay, Adv Enzyme Regul (1984) 22:27-55).
- the combinations of compounds provided herein can be evaluated in several assay systems, and the data can be analyzed utilizing a standard program for quantifying synergism, additivism, and antagonism among agents.
- a preferred program is that described by Chou and Talalay, in " New Avenues in Developmental Cancer
- a combination therapy may provide “synergy” and prove “synergistic,” i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
- a synergistic effect may be attained when the active ingredients are: (i) co formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (ii) delivered by alternation or in parallel as separate formulations; or (iii) by some other regimen.
- a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes.
- an effective dosage of each active ingredient is administered sequentially, i.e., serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
- the term "targeting moiety” refers to the portion of an immunotoxin intended to target a toxin to a cell of interest.
- the targeting moiety is an antibody, or a fragment of an antibody that retains antigen recognition capability, such as a scFv, a dsFv, an Fab, or an F(ab’)2, but it can also be, for example, a cytokine (e.g., IL-13), or other protein (such as a transferrin) that binds a specific antigen or receptor, preferably an antigen or a receptor on a cell surface.
- cytokine e.g., IL-13
- transferrin protein that binds a specific antigen or receptor, preferably an antigen or a receptor on a cell surface.
- targeted toxin refers to a cytotoxic agent, which is covalently linked to targeting moiety and targeted to a desired cell by the targeting moiety, which binds to a specific receptor or antigen present on the surface of such cell.
- T-lymphocyte cell As used herein, the terms "T-lymphocyte cell,” “T-lymphocyte,” or simply, “T-cell” refer to any lymphocyte that matures in the thymus and has the ability to recognize specific peptide antigens, or specific peptide antigens complexed with a major
- MHC histocompatibility complex protein
- the term "therapeutic agent” refers to any number of compounds that is administered to a patient to induce a desired therapeutic effect in a patient.
- a therapeutic effect intended is, for example, the killing of a cancer cell.
- Another therapeutic effect is to treat a patient.
- the terms “treat,” “treating,” and “treatment” or grammatical equivalents thereof refer to both therapeutic treatment and prophylactic or preventative measures.
- Those in need of treatment include subjects already afflicted with a pathological condition, disorder, or disease as well as those in which a pathological condition, disorder, or disease is to be prevented.
- a subject may have been diagnosed as having a pathological condition, disorder, or disease or may be predisposed or susceptible to a pathological condition, disorder, or disease.
- the terms include: (1) preventing a pathological condition, disorder, or disease, i.e.
- Treatment means any manner in which the symptoms of a pathological condition, disorder, or disease are ameliorated or otherwise beneficially altered.
- the subject in need of such treatment is a mammal, more preferable a human.
- This disclosure describes a variety of compounds useful for practicing combinations of compounds, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention.
- Applicants herein describe innovative approaches for (i) increasing expression of an antigen in a cancerous cell, preferably, an antigen in a cancerous lymphocyte cell, more preferably, an antigen in a cancerous B-lymphocyte cell, and (ii) for increasing expression of an antigen on the cell surface of a cancerous cell, preferably, an antigen on the cell surface of a cancerous lymphocyte cell, more preferably, an antigen on the cell surface of a cancerous B-lymphocyte cell.
- Increased expression of the antigen is achieved by contacting a cancerous cell in vitro or in vivo with a first composition comprising an agonist of protein kinase C, preferably, a bryostatin compound, and a second composition comprising a steroid or a third composition comprising a retinoic acid.
- a first composition comprising an agonist of protein kinase C, preferably, a bryostatin compound
- a second composition comprising a steroid or a third composition comprising a retinoic acid.
- compositions in particular compositions comprising an agonist for protein kinase C, preferably a bryostatin compound, to (i) increase expression of an antigen in a cancerous cell, preferably, to increase expression of an antigen in a lymphocyte cell, more preferably, to increase expression of an antigen in a B-lymphocyte cell, and (ii) to increase expression of an antigen on the cell surface of a cancerous cell, preferably, to increase expression of an antigen on the cell surface of a lymphocyte cell, more preferably, to increase expression of an antigen on the cell surface of a B-lymphocyte cell, in combination with a composition comprising a steroid or a retinoic acid.
- an agonist for protein kinase C preferably a bryostatin compound
- compositions comprising an agonist of protein kinase C, and in particular compositions comprising a bryostatin compound, (ii) compositions comprising a steroid, and (iii) compositions comprising a therapeutic compound for inhibiting growth of a cancerous cell.
- compositions comprising an agonist of protein kinase C, and in particular compositions comprising a bryostatin compound, (ii) compositions comprising a steroid, and (iii) compositions comprising a therapeutic compound for the treatment of a subject having cancer.
- compositions comprising an agonist of protein kinase C, and in particular compositions comprising a bryostatin compound, (ii) compositions comprising a retinoic acid, and (iii) compositions comprising a therapeutic compound for inhibiting growth of a cancerous cell.
- compositions comprising an agonist of protein kinase C, and in particular compositions comprising a bryostatin compound, (ii) compositions comprising a retinoic acid, and (iii) compositions comprising a therapeutic compound for the treatment of a subject having cancer.
- This disclosure provides combination of compounds and compositions that are useful in a wide range of methods. These methods include, but are not limited to, a method for increasing expression of an antigen in a cancerous cell, a method for increasing expression of an antigen in a cancerous lymphocyte cell, a method for increasing expression of an antigen in a cancerous B-lymphocyte cell, a method for increasing expression of an antigen on the cell surface of a cancerous cell, a method for increasing expression of an antigen on the cell surface of a cancerous lymphocyte cell, a method for increasing expression of an antigen on the cell surface of a cancerous B-lymphocyte cell, a method for preventing the growth of a cancerous cell, a method for preventing the growth of a cancerous lymphocyte cell, a method for preventing the growth of a cancerous B-lymphocyte cell, and a method for treating a subject having cancer. Given the guidance of this disclosure, other methods of use of the compositions of the present invention will become apparent to those skilled in the art.
- This invention discloses the surprising finding that combination of compounds (agonists of protein kinase C, steroids, and retinoic acids) and compositions comprising an agonist of protein kinase C, in particular, a bryostatin compound, when used as described herein, in combination with a steroid or retinoic acid or compositions comprising a steroid or a retinoic acid, lead to an increased expression of an antigen in a cancerous cell, lead to an increased expression of an antigen in a cancerous lymphocyte cell, lead to an increased expression of an antigen in a cancerous B-lymphocyte cell.
- This invention further discloses the unexpected and surprising finding that combinations of agonists of protein kinase C with a steroid or with a retinoic acid or compositions comprising an agonist of protein kinase C, in particular, a bryostatin compound, and compositions comprising a steroid or a retinoic acid synergize to achieve the described effect, i.e., synergistically increase expression of an antigen in a cancerous cell, synergistically increase expression of an antigen in a cancerous lymphocyte cell, synergistically increase expression of an antigen in a cancerous B- lymphocyte cell.
- any agonist of protein kinase C can be used in combination with a steroid or retinoic acid to achieve effects as described herein.
- compositions comprising an agonist of protein kinase C preferably, compositions comprising a bryostatin compound, and compositions comprising a steroid or a retinoic acid can be used in combination with compositions comprising a therapeutic agent to prevent the growth of a cancerous cell.
- compositions comprising an agonist of protein kinase C preferably, compositions comprising a bryostatin compound, and compositions comprising a steroid or a retinoic acid can be used in combination with compositions comprising a therapeutic agent to treat a subject having cancer.
- a combination or a composition is used for increasing expression of an antigen, preferably, an antigen in a cancerous cell, more preferably, an antigen on the cell surface of a cancerous cell.
- a combination or a composition is used for increasing expression of an antigen in a lymphocyte cell.
- a combination or a composition is used for increasing expression of an antigen on the cell surface of a lymphocyte cell.
- a combination or a composition is used for increasing expression of an antigen in a B-lymphocyte cell.
- a combination or a composition is used for increasing expression of an antigen on the cell surface of a B-lymphocyte cell.
- composition comprises an agonist of protein kinase C (PKC), i.e., a compound increasing PKC activity.
- PKC protein kinase C
- EC 2.7.11.13 is a family of protein kinase enzymes that are involved in controlling the function of other proteins through the phosphorylation of hydroxyl groups of serine and threonine amino acid residues on these proteins, or a member of this family. PKC enzymes play important roles in several signal transduction cascades and regulate growth factor response, proliferation, and apoptosis. Its central role in these processes, which are closely involved in tumor initiation, progression, and response to antitumor agents, makes it an attractive therapeutic target in cancer.
- a PKC agonist may be any molecule that effects an increase in the activity of PKC. This includes proteins, peptides, DNA molecules (including antisense), RNA molecules (including RNAi and antisense) and small molecules.
- PKC agonists can be used to practice combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention.
- an agonist of protein kinase C is selected from the group consisting of ingenol-3-angelate, prostratin, staurospine, valproate, tamoxifen, chelerythrine, miyabenol C, verbascoside, BIM-1, indolocarbazole, isoquinolinesulfonamide, sphingosine, calphostin C (UCN-1028C), bisindolylmaleimide GF 109203X, a plant flavonoid, and a bryostatin compound.
- TNF-b Methyl NF-b
- IL-Ib Olet al, Cytokine (1990) 2(6):389-97
- IL-Ib Olet al, Proc Natl Acad Sci USA (1989) 86(7):2336-40
- lipopolysaccharide Sen et al., Cell (1986) 47(6):921-8
- UV-light Step et al., Mol Cell Biol (1989) 9(11):5169-81
- CD3 antibodies Teong-Starkesen et al., J Immunol (1989)
- daunorubicin (Wang et al., Science (1996) 274(5288):784-7); hydrogen peroxide (Shreck at al, 1991, EMBO 10(8): 2247-58); nocodazole (Rosette et al, 1995, J Cell Biol 128(6): 1111- 9); LIGHT (Zou et al, 2005, J Cell Physiol 205(3):437-43); bleomycin (Ishii et al, 2002, Toxicol Appl Pharmicol 184(2): 88-97); camptothecin (Piret et al, 1996 Nucleic Acids Res 24(20:4242-8); cisplatin (Nie et al, 1998, Mol Pharmacol 53(4):663-9); celecoxib (Kim e/ al, 2004, J Cancer Res Clin Oncol 130(9):551-60); ciprofibrate (Li et al, 1996,
- an agonist of protein kinase C is selected from the group consisting of ingenol-3-angelate, prostratin, staurospine, valproate, tamoxifen, chelerythrine, miyabenol C, verbascoside, BIM-1, indolocarbazole,
- cycloprodigiosin dacarbazine, Daio-Orengedeokuto, daunomycin, diazoxide, diclofenac, 5,6- dimethylxanthenone-4-acetic acid, flavone-8-acetic acid, haloperidol, imiquimod, isochamaejasmin, lithium, mitoxantrone, morphine, nipradilol, norepinephrine, nystatin, oltipraz, protocatechuic acid, SN38 (metabolite of CPT-11), Taxol (Paclitaxel), vinblastine, vincristine, WR1065, a bryostatin compound, single stereoisomers, mixtures of
- a preferred PKC agonist is a bryostatin compound.
- an agonist of protein kinase C is a bryostatin compound.
- composition comprising an agonist of protein kinase C is also referred to herein as a first composition.
- a composition comprising an agonist of protein kinase C is also referred to herein as a first composition.
- composition comprising an agonist of protein kinase C is also referred to herein as a first pharmaceutical composition.
- a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method comprises a bryostatin compound.
- a variety of bryostatin compounds can be used in a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention.
- a bryostatin compound is a naturally occurring bryostatin. a. Naturally Occurring Bryostatins
- a bryostatin compound is a naturally occurring bryostatin compound.
- the bryostatins are a family of structurally complex natural products isolated from the marine bryozoan Bugula neritina. 20 bryostatin compounds have been isolated from B. neritina (Wender et al., Isr J Chem (2011) 51(3- 4):453-472). Recent studies have demonstrated that the biological source of the bryostatins is Endobugula sertula, a bacterial symbiont to B.
- bryostatins render host larvae unpalatable to predators (Lopanik et al, Oecologia (2004) 139: 131-139). Structural formula of those naturally occurring bryostatins are shown in FIG. 2. Naturally occurring bryostatins are also useful for practicing compositions, systems, and methods of the present invention.
- a bryostatin compound is Bryostatin-1 (see, FIG. 2).
- Bryostatin-1 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-1.
- a bryostatin compound is Bryostatin-2 (see, FIG. 2).
- Bryostatin-2 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-2.
- a bryostatin compound is Bryostatin-3 (see, FIG. 2).
- Bryostatin-3 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-3.
- a bryostatin compound is Bryostatin-4 (see, FIG. 2).
- Bryostatin-4 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-4.
- a bryostatin compound is Bryostatin-5 (see, FIG .2). Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-5.
- a bryostatin compound is Bryostatin-6 (see. FIG. 2).
- Bryostatin-6 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-6.
- a bryostatin compound is Bryostatin-7 (see. FIG. 2).
- Bryostatin-7 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-7.
- a bryostatin compound is Bryostatin-8 (see. FIG. 2).
- Bryostatin-8 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-8.
- a bryostatin compound is Bryostatin-9 (see. FIG. 2).
- Bryostatin-9 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-9.
- a bryostatin compound is Bryostatin-10 (see. FIG. 2).
- Bryostatin-10 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-10.
- a bryostatin compound is Bryostatin-11 (see. FIG. 2).
- Bryostatin-11 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-11.
- a bryostatin compound is Bryostatin-12 (see. FIG. 2). Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-12.
- a bryostatin compound is Bryostatin-13 (see. FIG. 2). Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-13.
- a bryostatin compound is Bryostatin-14 (see. FIG. 2). Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-14.
- a bryostatin compound is Bryostatin-15 (see. FIG. 2).
- Bryostatin-15 Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-15.
- a bryostatin compound is Bryostatin-16 (see. FIG. 2). Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-16.
- a bryostatin compound is Bryostatin-17 (see. FIG. 2).
- Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-17.
- a bryostatin compound is Bryostatin-18 (see. FIG. 2). Also useful for combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-18.
- a bryostatin compound is Bryostatin-19 (see. FIG. 2).
- Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-19.
- a bryostatin compound is Bryostatin-20 (see. FIG. 2). Also useful for practicing combinations, compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-20. b. Bryostatin Compound Having Formula 1
- a bryostatin compound is a bryostatin analog.
- a bryostatin analog retains some function of a naturally occurring bryostatin; however, when compared to a naturally occurring bryostatin, a bryostatin analog may also demonstrate certain improved characteristics, such as longer half-life, more or less potent activity against a target, such as protein kinase C, easier synthesis, etc.
- Some exemplary, non-limiting bryostatin analogs are described herein and are presented by their structural formula.
- a bryostatin compound for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention is a compound described in published US2017/0239212 A1 (herewith incorporated by reference in its entirety).
- Preferred compounds described in published US2017/0239212 A1 for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention are referred to herein as bryostatin compound having Formula 1 :
- R 1 is hydrogen, an alkyl group, an aryl group, an amino group, a cycloalkyl group, an alkenyl, or an alkynyl group;
- R 2 is hydrogen, an alkyl group or aryl group
- X 1 and X 2 are, independently, hydrogen, an alkyl group, a hydroxyl, or a substituted hydroxyl group;
- X 3 is hydrogen, hydroxyl, an alkyl group, an alkoxy group, or a halide
- Y 1 , Y 2 , and Y 3 are, independently, hydrogen, an alkyl group, a hydroxyl group, a substituted hydroxyl group, an oxo group, a substituted or unsubstituted alkylene group, or— 0C(0)R 3 , where R 3 is an alkyl group;
- Z 1 and Z 2 are, independently, hydrogen, an alkyl group, a hydroxyl group, a substituted hydroxyl group, or collectively form a cycloalkyl group; wherein when C7, C9, C13, C20, or C26 is a chiral center, the chiral center is the substantially pure enantiomer; and wherein the compound is not Bryostatin-1.
- a bryostatin compound for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention is a compound described in published US2011/0269713 Al (herewith incorporated by reference in its entirety).
- Preferred compounds described in published US2011/0269713 Al for use in a composition, a pharmaceutical composition, a medicament, a kit, an article, of manufacture, or a method of the present invention are referred to herein as bryostatin compound having Formula 2:
- R 1 is hydrogen, an alkyl group, an aryl group, a cycloalkyl group, an alkenyl, or an alkynyl group;
- R 2 is an alkyl group or aryl group
- X 1 and X 2 are, independently, hydrogen, an alkyl group, a hydroxyl, or a substituted hydroxyl group;
- X 3 is hydrogen, hydroxyl, an alkyl group, an alkoxy group, or a halide
- Y 1 , Y 2 , and Y 3 are, independently, hydrogen, an alkyl group, a hydroxyl group, a substituted hydroxyl group, an oxo group, a substituted or unsubstituted alkylene group, or— 0C(0)R 3 , where R 3 is an alkyl group;
- Z 1 and Z 2 are, independently, hydrogen, an alkyl group, a hydroxyl group, a substituted hydroxyl group, or collectively form a cycloalkyl group; wherein when C7, C9, C13, C20, or C26 is a chiral center, the chiral center is the
- substantially pure enantiomer and wherein the compound is not Bryostatin-1, and wherein X 3 , Y 1 , Y 2 , Z 1 , and Z 2 are simultaneously hydrogen.
- a bryostatin compound for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention is a compound described in published US2010/0280262 Al and US2013/0123518 Al (herewith incorporated by reference in its entirety).
- Preferred compounds described in published US2010/0280262 Al and US2013/0123518 Al for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention are referred to herein as bryostatin compound having Formula 3:
- R is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heteroaralkyl, optionally substituted heteroalkyl, optionally substituted alkyl(cycloheteroalkyl);
- R3 is independently H,— OH, or 0(C0)R;
- R a and R b are independently H,— COOR' ,— CONR c R d or R’;
- R c and R d are independently H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, (CH 2 ) t CONH 2 R', or (CH 2 ),COOR' where t is 1, 2 or 3;
- R 6 is H,— OH, or R';
- R' is optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heteroaralkyl, optionally substituted heteroalkyl, optionally substituted alkyl(cycloheteroalkyl), (CO)R", or (COO)R";
- R" is optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heteroaralkyl, optionally substituted heteroalkyl, or optionally substituted alkyl(cycloheteroalkyl);
- Xi; X2, X3, and X4 are independently C(R I ) 2 , O, S, or N(Ri);
- Y is O or N(R t ); m is 0 or 1; n is 0, 1, 2, or 3; and p is 0, 1,2, 3, or 4; with the proviso that the compound does not have the structure of Formula A
- a bryostatin compound for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention is a compound described in published US2009/0270492 Al (herewith incorporated by reference in its entirety).
- Preferred compounds described in published US2009/0270492 Al for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention are referred to herein as bryostatin compound having Formula 4:
- X is O, S or NR'
- R 3 is H or OH
- R 20 is H, OH, or -T-U— V— R' where:
- T is selected from— O— ,— S— ,— N(H)— or— N(Me)— ;
- U is absent or is selected from— C(O)— ,— C(S)— ,— S(O)— or— S(0) 2— ;
- V is absent or is selected from— O— ,— S— ,— N(H)— or— N(Me)— , provided that V is absent when U is absent;
- R a and R b are independently H, CQ2R', CONR c R d or R'; R c and R d are independently H, alkyl, alkenyl or alkynyl, or (CH 2 ) n C0 2 R' where n is 1, 2 or
- R 26 is H or R'
- R' is independently selected from the group consisting of H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl and optionally substituted heteroaralkyl.
- a bryostatin compound having Formula 4 is not
- R'" is C7H15CH3C13H27, Ph
- R" is selected from the group consisting of CH3, Ph, C13H27, C7H15,
- a bryostatin compound for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention is a compound described in published US2009/0270492 A1 (herewith incorporated by reference in its entirety).
- Preferred compounds described in published US2009/0270492 A1 for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention are referred to herein as bryostatin compound having Formula 5:
- R 20 is H, OH, or -T-U— V— R' where:
- T is selected from— O— ,— S— ,— N(H)— or— N(Me)— ;
- U is absent or is selected from— C(O)— ,— C(S)— ,— S(O)— or— S(0) 2— ;
- V is absent or is selected from— O— ,— S— ,— N(H)— or— N(Me)— , provided that V is absent when U is absent;
- R 21 CR a R b or R 21 represents independent moieties R c and R d where:
- R a and R b are independently H, CQ2R', CONR c R d or R';
- R c and R d are independently H, alkyl, alkenyl or alkynyl, or (Cty n CC R 1 where n is 1, 2 or
- R 26 is H or R'
- R' is independently selected from the group consisting of H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl and optionally substituted heteroaralkyl.
- a bryostatin compound having Formula 5 is not
- R'" is C7H15CH3C13H27, Ph
- R" is selected from the group consisting of CH3, Ph, C13H27, C7H15,
- a bryostatin compound for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention is a compound described in published US2009/0270492 A1 (herewith incorporated by reference in its entirety).
- Preferred compounds described in published US2009/0270492 A1 for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention are referred to herein as bryostatin compound having Formula 6:
- R 3 is H or OH
- R 8 is selected from the group consisting of H, OH, R',— (CH 2 ) n 0 2 CR', and— (CH 2 ) n 0 2 C- haloalkyl; n is 0, 1,2, 3, 4, or 5;
- R 9 is H or OH
- R 20 is H, OH, or -T-U— V— R' where:
- T is selected from— O— ,— S— ,— N(H)— or— N(Me)— ;
- U is absent or is selected from— C(O)— ,— C(S)— ,— S(O)— or— S(0) 2— ;
- V is absent or is selected from— O— ,— S— ,— N(H)— or— N(Me)— , provided that V is absent when U is absent;
- R c and R d are independently H, alkyl, alkenyl or alkynyl, or (CH 2 ) n C0 2 R' where n is 1, 2 or
- R 26 is H or R'
- R' is independently selected from the group consisting of H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl and optionally substituted heteroaralkyl.
- a bryostatin compound having Formula 6 is not
- a bryostatin compound for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, a use or a method of the present invention is a compound described in published US2009/0270492 Al (herewith incorporated by reference in its entirety).
- Preferred compounds described in published US2009/0270492 Al for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention are referred to herein as bryostatin compound having Formula 7:
- R 3 is H or OH
- R 20 is H, OH, or -T-U— V— R' where:
- T is selected from— O— ,— S— ,— N(H)— or— N(Me)— ;
- U is absent or is selected from— C(O)— ,— C(S)— ,— S(O)— or— S(0)2— ;
- V is absent or is selected from— O— ,— S— ,— N(H)— or— N(Me)— , provided that V is absent when U is absent;
- R 21 is H, alkyl, alkenyl or alkynyl, or (CH2) n C02R' where n is 1, 2 or 3; R 26 is H or R'; and
- R' is independently selected from the group consisting of H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl and optionally substituted heteroaralkyl.
- a bryostatin compound for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, a use or a method of the present invention is a compound described in published US2011/0014699 Al (herewith incorporated by reference in its entirety).
- a bryostatin compound is selected from the group consisting of Bryostatin- 1, bryostatin-2, bryostatin-3, bryostatin-4, bryostatin-5, bryostatin-6, bryostatin-7, bryostatin-8, bryostatin-9, Bryostatin- 10, Bryostatin- 11, Bryostatin- 12, Bryostatin-13, Bryostatin- 14, Bryostatin- 15, Bryostatin- 16, Bryostatin- 17, Bryostatin- 18, Bryostatin- 19, bryostatin-20, a bryostatin compound having Formula 1, a bryostatin compound having Formula 2, a bryostatin compound having Formula 3, a bryostatin compound having Formula 4, a bryostatin compound having Formula 5, a bryostatin compound having Formula 6, a bryostatin compound having Formula 7, single stereoisomers, mixtures of
- Various steroids can be used in a combination, a composition, a
- compositions comprising a steroid
- a pharmaceutical composition comprising a steroid is also referred to herein as a second pharmaceutical composition.
- a steroid is a biologically active organic compound with four rings arranged in a specific molecular configuration. Steroids have two principal biological functions: as important components of cell membranes which alter membrane fluidity; and as signaling molecules. Hundreds of steroids are found in plants, animals and fungi. All steroids are manufactured in cells from the sterols lanosterol (opisthokonts) or cycloartenol (plants). Lanosterol and cycloartol are derived from the cyclization of the triterpene squalene.
- a steroid is selected from the group consisting of a corticosteroid, a sex steroid, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters or prodrugs thereof.
- a corticosteroid comprises glucocorticoids and mineralocorticoids.
- a corticosteroid is selected from the group consisting of a glucocorticoid, a mineralocorticoid, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters or prodrugs thereof.
- Glucocorticoids include cortisol, corticosterone, cortisone, and synthetic glucocorticoids, such as dexamethasone.
- a glucocorticoid is selected from the group consisting of cortisol, corticosterone, cortisone, and a synthetic glucocorticoid, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters or prodrugs thereof.
- Mineralocorticoids include aldosterone.
- a mineralocorticoid is aldosterone, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters or prodrugs thereof.
- Sex steroids include progestagens (e.g., progesterone), androgens (e.g., testosterone) and estrogens (e.g., estradiol).
- progestagens e.g., progesterone
- androgens e.g., testosterone
- estrogens e.g., estradiol
- a sex steroid is selected from the group consisting of progestogen, androgen, estrogen, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters or prodrugs thereof.
- a steroid is a steroid analog as described in U.S. Patent Publication No. 20180221390 (incorporated herewith by reference in its entirety), or single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters or prodrugs thereof.
- a steroid is a combination of two or more of any of the aforementioned steroids or single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters or prodrugs thereof.
- a steroid is a glucocorticoid.
- a glucocorticoid is selected from the group consisting of a dexamethasone compound, prednisolone, prednisone, methyl prednisolone, triamcinolone, hydrocortisone, deflazacort, betamethasone, budesonide, or single
- stereoisomers mixtures of stereoisomers, pharmaceutically acceptable salts, esters or prodrugs thereof.
- a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method comprises a dexamethasone compound.
- a dexamethasone compound is selected from the group consisting of dexamethasone and a dexamethasone analog, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives thereof.
- a dexamethasone compound is dexamethasone.
- Dexamethasone is known for a number of uses, including treatment of inflammatory and autoimmune diseases such as rheumatoid arthritis.
- dexamethasone has not been recognized as an effective treatment for acute or chronic lymphocytic leukemia or for any other leukemia or for a hematologic cancer.
- Dexamethasone also has not been recognized for synergistically activating an antigen in combination with a protein kinase C agonist, such as a bryostatin compound, and more specifically, in combination with Bryostatin-1.
- Dexamethasone is an anti-inflammatory glucocorticoid. Its chemical names include (11 b, 16a)-9-Fluoro-l 1 , 17,21 -trihydroxy-16-methylpregna-l ,4-diene-3,20-dione; 9a- Fluoro-16a-methyl prednisolone and 16a-methyl-9a-riuoro- 1 4-pregnadiene- 1 1 b 17 a21 -triol- 3,20-dione.
- Dexamethasone is also known as (8S, 9R, 10S, 11S, 13S, 14S, 16R, 17R)-9- Fluoro-l l,17-dihydroxy-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7- ,8,11,12,14,15,16- octahydro-cyclopenta[a]phenanthren-3-one, and is available from various pharmaceutical manufacturers.
- Two known commercially available forms are dexamethasone acetate and dexamethasone sodium phosphate. It’s structural Formula is
- a dexamethasone compound is dexamethasone, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives thereof.
- dexamethasone examples include dexamethasone, dexamethasone acetate and dexamethasone sodium phosphate.
- dexamethasone refers to dexamethasone salts, acids, variants, polymorphs, derivatives, prodrugs and metabolites.
- Dexamethasone formulations and delivery systems useful for practicing combinations, compositions, pharmaceutical compositions, medicaments, kits, articles of manufacture, uses and methods of the present invention are described, e.g., in US2018/0303851, US2017/0304319, US2015/0342964, US2015/0065474, US2015/0024034, US2014/0315867, US2013/0310352, US2010/0082095, US2008/0241231, US2008/0004694, US2006/0034915, US2005/0014701 (each of which is incorporated by reference herewith in its entirety).
- the amount of dexamethasone or a dexamethasone compound can be described as the total amount of dexamethasone.
- a combination or a composition comprises from about 10 ng to about 10 mg of dexamethasone. More preferably,
- dexamethasone can be present at about 100 ng to about 1 mg. Even more preferably, dexamethasone can be present at about 1 pg to about 500 pg. Still more preferably, dexamethasone can be present from about 10 pg to about 250 pg. Still more preferably, dexamethasone can be present at about 100 pg to about 200 pg. Most preferably,
- dexamethasone can be present at about 150 ug.
- a combination or a composition comprises dexamethasone at a concentration of about 10 ng/ml to about 10 mg/ml. More preferably, dexamethasone can be present at a concentration of about 100 ng/ml to about 1 mg/ml. Even more preferably, dexamethasone can be present at a concentration of about 1 ug/ml to about 100 ug/ml. Still more preferably, dexamethasone can be present at a concentration of about 5 ug/ml to about 50 ug/ml.
- dexamethasone can be present at a concentration of about 8 ug/ml to about 25 ug/ml. Most preferably, dexamethasone can be present at a concentration of about 10 ug/ml. Also useful for practicing combinations, compositions, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of
- a dexamethasone compound is a dexamethasone analog.
- a dexamethasone analog retains some function of dexamethasone; however, when compared to dexamethasone, a dexamethasone analog may also demonstrate certain improved characteristics, such as longer half-life, more or less potent activity against a target, easier synthesis, etc.
- Some exemplary, non-limiting dexamethasone analogs are described herein and are presented by their structural formula.
- a dexamethasone compound for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention is a compound described in published US2010/0082095 A1 (herewith incorporated by reference in its entirety).
- Preferred compounds described in published US2010/0082095 A1 for use in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention are referred to herein as bryostatin compound having Formula 8:
- Ri is selected from the group consisting of the following:
- R2 is selected from the group consisting of (Cl-C16)alkyl, (C2-C16)alkenyl, (C2-C16)alkenyl, (C3-C10)cycloalkyl, phenyl, ester, carbonate, ether, and ketone, wherein any one or more hydrogen atoms on the cycloalkyl or phenyl; ring is optionally substituted with a substituent selected from the group consisting of alkyl, alkenyl, and alkynyl; and one or more hydrogen atoms on R2 are optionally substituted with chlorine, fluorine or a combination thereof; and the total number of carbon atoms in R2 is not more than 16.
- the amount of dexamethasone or a dexamethasone compound can be described as the total amount of dexamethasone.
- a combination or a composition comprises from about 10 ng to about 10 mg of dexamethasone. More preferably,
- dexamethasone can be present at about 100 ng to about 1 mg. Even more preferably, dexamethasone can be present at about 1 pg to about 500 pg. Still more preferably, dexamethasone can be present from about 10 pg to about 250 pg. Still more preferably, dexamethasone can be present at about 100 pg to about 200 pg. Most preferably, dexamethasone can be present at about 150 ug. In some embodiments of the present invention, a combination or a composition comprises dexamethasone at a concentration of about 10 ng/ml to about 10 mg/ml.
- dexamethasone can be present at a concentration of about 100 ng/ml to about 1 mg/ml. Even more preferably, dexamethasone can be present at a concentration of about 1 ug/ml to about 100 ug/ml. Still more preferably, dexamethasone can be present at a concentration of about 5 ug/ml to about 50 ug/ml. Still more preferably, dexamethasone can be present at a concentration of about 8 ug/ml to about 25 ug/ml. Most preferably, dexamethasone can be present at a concentration of about 10 ug/ml. Also useful for practicing combinations, compositions, kits, articles of manufacture, uses and methods of the present invention are single stereoisomers, mixtures of
- a dexamethasone compound is a dexamethasone compound having Formula 8, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives thereof.
- a dexamethasone compound is selected from the group consisting of dexamethasone and a dexamethasone compound having Formula 8, single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives thereof. ii. Prednisolone
- a steroid is prednisolone, a single stereoisomer, a mixture of stereoisomers, a pharmaceutically acceptable salt, an ester, a prodrug or a functional derivative thereof.
- a steroid is prednisone, a single stereoisomer, a mixture of stereoisomers, a pharmaceutically acceptable salt, an ester, a prodrug or a functional derivative thereof.
- retinoid acids can be used in a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention.
- a composition comprising a retinoid is also referred to herein as a third composition.
- a pharmaceutical composition comprising a retinoid is also referred to herein as a third pharmaceutical composition.
- a retinoid acid is tretinoin.
- Tretinoin also known as all-trans retinoic acid (ATRA) is medication used for the treatment of acne and acute promyelocytic leukemia.
- ATRA all-trans retinoic acid
- a combination or a composition comprises a bryostatin compound and a steroid. Any bryostatin compound can be combined with any steroid. a. Combinations and compositions comprising a bryostatin compound and a glucocorticoid
- a combination or a composition comprises a bryostatin compound and a glucocorticoid. Any bryostatin compound can be combined with any glucocorticoid. i. Combinations and compositions comprising a
- a combination or a composition comprises a bryostatin compound and a dexamethasone compound. Any bryostatin compound can be combined with any dexamethasone compound.
- a combination or a combination comprises a bryostatin compound and a dexamethasone compound. Any bryostatin compound can be combined with any dexamethasone compound.
- composition comprises Bryostatin-1 and dexamethasone.
- Combinations and compositions comprising (i) Bryostatin-1 and (ii) dexamethasone are referred to herein, from time to time, as ND 1000.
- a ND1000 combination or composition comprising Bryostatin-1 and dexamethasone may comprise Bryostatin-1 at a concentration ranging from about .01 nanomolar to about 10 millimolar.
- a ND1000 combination or composition comprising Bryostatin-1 and dexamethasone comprises
- Bryostatin-1 at a concentration ranging from about .1 nanomolar to about 10 nanomolar.
- a ND1000 combination or composition comprising Bryostatin-1 and dexamethasone may comprise dexamethasone at a concentration ranging from about 2 nanomolar to about 20 millimolar.
- a NDlOOO combination or composition comprising Bryostatin-1 and dexamethasone comprises dexamethasone at a concentration ranging from about 20 nanomolar to about 200 nanomolar.
- composition comprises a bryostatin compound and prednisolone. Any bryostatin compound can be combined with any prednisolone.
- a preferred combination comprises Bryostatin-1 and prednisolone.
- a combination or composition comprising Bryostatin-1 and prednisolone, may from time to time, referred to herein, as ND1000B.
- a ND1000B combination or composition comprising Bryostatin-1 and prednisolone may comprise Bryostatin-1 at a concentration ranging from about .01 nanomolar to about 10 millimolar.
- a ND1000B combination or composition comprising Bryostatin-1 and prednisolone comprises Bryostatin-1 at a concentration ranging from about .1 nanomolar to about 10 nanomolar.
- a ND1000B combination or composition comprising Bryostatin-1 and prednisolone may comprise prednisolone at a concentration ranging from about 1 nanomolar to about 20 millimolar.
- a ND1000B combination or composition comprising Bryostatin-1 and prednisolone comprises prednisolone at a concentration ranging from about 50 nanomolar to about 500 nanomolar.
- composition comprises a bryostatin compound and a retinoic acid. Any bryostatin compound can be combined with any retinoic acid.
- a preferred combination or composition comprises Bryostatin-1 and a retinoic acid. a. Combinations and compositions comprising a bryostatin compound and tretinoin
- composition comprises a bryostatin compound and tretinoin. Any bryostatin compound can be combined with tretinoin.
- a preferred combination or composition comprises Bryostatin-1 and tretinoin.
- a combination or composition comprising Bryostatin-1 and tretinoin may from time to time, referred to herein, as ND1000C.
- a ND1000C combination or composition comprising Bryostatin-1 and tretinoin may comprise Bryostatin-1 at a concentration ranging from about .01 nanomolar to about 10 millimolar.
- a ND1000C combination or composition comprising Bryostatin-1 and tretinoin comprises Bryostatin-1 at a concentration ranging from about .1 nanomolar to about 10 nanomolar.
- a ND1000C combination or composition comprising Bryostatin-1 and tretinoin may comprise tretinoin at a concentration ranging from about 0.1 nanomolar to about 50 millimolar.
- a ND1000C may comprise tretinoin at a concentration ranging from about 0.1 nanomolar to about 50 millimolar.
- combination or composition comprising Bryostatin-1 and tretinoin comprises tretinoin at a concentration ranging from about 5 nanomolar to about 50 nanomolar.
- a combination or a composition is used to contact a cancerous cell.
- Various cancerous cells can be used to practice a combination, a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention.
- a cancerous cell is obtained from a subject having a cancer selected from the group consisting of a lung cancer, a sarcoma, a gastrointestinal cancer, a cancer of the genitourinary tract, a liver cancer, a skin cancer, a gynecological cancer, a bone cancer, a cancer of the nervous system, a cancer of adrenal gland, and a hematologic cancer.
- cancerous cells can be maintained and cultured in vitro or ex vivo.
- a cancerous cell is obtained from a subject having a hematologic cancer.
- Various cancerous cells obtained from a subject having a hematologic cancer can be used to practice a composition, a pharmaceutical composition, a medicament, a kit, an article of manufacture, or a method of the present invention.
- cancerous cells are from a subject having a hematologic cancer selected from the group consisting of B-cell lymphoma, T-cell lymphoma, leukocytic leukemia, acute leukocytic leukemia, chronic leukocytic leukemia, lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, lymphocytic leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, monocytic leukemia, multiple myeloma, myelodysplastic syndrome, myelogenous leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, myeloid leukemia, acute myeloid leukemia, chronic myeloid leukemia, myeloproliferative disease, acute myelocytic leukemia, myelocytic leukemia, chronic myelocytic leukemia, promyelocytic leukemia,
- a cancerous cell is a cancerous lymphocyte cell. In some embodiments of the present invention, a cancerous cell is a cancerous B-lymphocyte cell.
- a cancerous cell is an acute lymphocytic leukemia cell. In some embodiments of the present invention, a cancerous cell is a chronic lymphocytic leukemia cell.
- a combination ora composition is used to increase expression of an antigen in a cancerous cell.
- combination or a composition is used to increase expression of an antigen located on the cell surface of a cancerous cell.
- antigens described herein one of ordinary skill in the art will appreciate that expression of a variety of antigens known in the art can be increased.
- combination or a composition is used to increase expression of an antigen in a cancerous lymphocyte cell. In some embodiments of the present invention, combination or a composition is used to increase expression of an antigen located on the cell surface of a cancerous lymphocyte cell.
- Combinations and compositions of the present invention can be used to increase expression of a variety of lymphocyte antigens.
- combination or a composition increases expression of an antigen selected from the group consisting of CDlb, CDlc, CDld, CD2, CD3d, CD3e, CD3g, CD4, CD5, CD6, CD7, CD8a, CD8b, CD10, CDl la, CDl lb, CDl lc, CD13, CD14, CD16a, CD16b, CD18, CD19, CD20, CD21, CD22, CD23, CD25, CD26, CD27, CD28, CD29, CD30, CD31, CD32, CD33, CD34, CD35, CD40, CD41, CD42d, CD44, CD45, CD46, CD47, CD48, CD49A, CD49b, CD49c, CD49d, CD49e, CD49f, CD50, CD51, CD54, CD55, CD56, CD58, CD59, CD
- combination or a composition is used to increase expression of a B-lymphocyte antigen in a cancerous B-lymphocyte cell.
- combination or a composition is used to increase expression of a B-lymphocyte antigen located on the cell surface of a cancerous B- lymphocyte cell.
- Combinations and compositions of the present invention can be used to increase expression of a variety of B-lymphocyte antigens.
- combination or a composition increases the expression of a B-lymphocyte antigen selected from the group consisting of CD19, CD20, CD22, CD30, CD38, CD70, and CD269.
- combination or a composition increases expression of CD 19.
- An exemplary and non-limiting combination or composition that synergistically increases expression of CD 19 comprises Bryostatin-1 and dexamethasone (see. FIGs. 8A-B, 9A-B, 10A-B). Such composition is referred to herein, from time to time, as ND1000.
- compositions increases expression of CD20.
- a combination or a composition increases expression of CD20.
- a combination or a composition synergistically increases expression of CD20.
- An exemplary and non-limiting combination or composition that synergistically increases expression of CD20 comprises Bryostatin-1 and dexamethasone (see. FIGs. 3A-B, 4A-B, 5A-D, 6A-B, 7A-B, 11A-C, 16). Such combination or composition is referred to herein, from time to time, as ND1000.
- Another exemplary and non-limiting combination or composition that synergistically increases expression of CD20 comprises Bryostatin-1 and prednisolone (see, FIG. 16). Such composition is referred to herein, from time to time, as ND1000B.
- composition increases expression of CD22.
- a combination or a composition increases expression of CD22.
- a combination or a composition synergistically increases expression of CD22.
- An exemplary and non-limiting combination or composition that synergistically increases expression of CD22 comprises Bryostatin-1 and dexamethasone (see, FIGs. 13, 14A-B, 15). Such combination or composition is referred to herein, from time to time, as ND1000.
- CD22 synergistically increases expression of CD22 comprises Bryostatin-1 and tretinoin (see, FIGs. 14A-B, 15). Such composition is referred to herein as ND1000C.
- combination or a composition comprises a therapeutic agent.
- a combination or a composition comprising a therapeutic agent is also referred to herein as a fourth composition, merely to distinguish it from a first composition comprising an agonist for protein kinase C, from a second composition comprising a steroid or from a third composition comprising a retinoic acid as described herein.
- a pharmaceutical composition comprising a therapeutic agent is also referred to herein as a fourth pharmaceutical composition.
- a therapeutic agent is an agent that specifically binds to an antigen in a cancerous cell, preferably to an antigen on the cell surface of a cancerous cell.
- a therapeutic agent is an agent that specifically binds to an antigen in a cancerous lymphocyte cell, preferably to an antigen on the cell surface of a cancerous lymphocyte cell.
- a therapeutic agent is an agent that specifically binds to an antigen in a cancerous B-lymphocyte cell, preferably to an antigen on the cell surface of a cancerous B-lymphocyte cell.
- a therapeutic agent is an agent that specifically binds to an antigen in a cancerous cell, preferably to an antigen on the cell surface of a cancerous cell, the expression of which increased upon contacting the cancerous cell with a first composition of the present invention comprising an agonist of protein kinase C and with a second composition of the present invention comprising a steroid or with a first composition of the present invention comprising an agonist of protein kinase C and a third composition of the present invention comprising a retinoic acid.
- a therapeutic agent is an agent that specifically binds to an antigen in a cancerous lymphocyte cell, preferably to an antigen on the cell surface of a cancerous lymphocyte cell, the expression of which increased upon contacting the cancerous lymphocyte cell with a first composition of the present invention comprising an agonist of protein kinase C and with a second composition of the present invention comprising a steroid or with a first composition of the present invention comprising an agonist of protein kinase C and with a third composition of the present invention comprising a retinoic acid.
- a therapeutic agent is an agent that specifically binds to an antigen in a cancerous B-lymphocyte cell, preferably to an antigen on the cell surface of a cancerous B-lymphocyte cell, the expression of which increased upon contacting the cancerous B-lymphocyte cell with a first composition of the present invention comprising an agonist of protein kinase C and with a second composition of the present invention comprising a steroid or with a first composition of the present invention comprising an agonist of protein kinase C and with a third composition of the present invention comprising a retinoic acid.
- a variety of therapeutic agents may be used to achieve a desired effect upon binding of the therapeutic agent to an antigen in a cancerous cell, preferably, to an antigen in a cancerous lymphocyte cell, and more preferably, to an antigen in a cancerous B-lymphocyte cell.
- a variety of therapeutic agents may be used to achieve a desired effect upon binding of the therapeutic agent to an antigen on the cell surface of a cancerous cell, preferably, to an antigen on the cell surface of a cancerous lymphocyte cell, and more preferably, to an antigen on the cell surface of a cancerous B-lymphocyte cell.
- any one of the agonists for protein kinase C, any one of the steroid compounds, and any one of the retinoic acid compounds can be combined with a therapeutic agent and thus produce a variety of combinations.
- Preferred therapeutic agents include, but are not limited to, antibodies and immunotoxins.
- a therapeutic agent is an antibody.
- Various antibodies can be used to prepare a composition of the present invention or to use such composition in a method of the present invention.
- an antibody is selected from the group consisting of an antibody mediating antibody-dependent cellular cytotoxicity, a monoclonal antibody, a humanized antibody, a chimeric antibody, a bi-specific antibody, an ADC, an antibody fragment, and a recombinant antibody.
- an antibody is an antibody for mediating antibody-dependent cellular cytotoxicity (ADCC), also called antibody-dependent cell-mediated cytotoxicity.
- ADCC is an immune mechanism through which Fc receptor bearing effector cells (e.g., natural killer (NK) cells) can recognize and kill antibody-coated target cells expressing tumor antigens on their surface.
- Fc receptor bearing effector cells e.g., natural killer (NK) cells
- NK natural killer
- an antibody is a monoclonal antibody.
- a monoclonal antibody is Ofatumumab ® .
- Ofatumumab ® is also known as ARZERRA ® .
- Ofatumumab ® is an anti- CD20 IgGlK human monoclonal antibody.
- the candidate targets the same antigen as rituximab, but Ofatumumab ® binds a novel, membrane-proximal epitope, and dissociates from its target at a slower rate compared to rituximab (Zhang, MAbs (2009) 1(4):326-331).
- an antibody is a humanized antibody. iv. Chimeric Antibody
- an antibody is a chimeric antibody.
- a chimeric antibody is Rituximab ® .
- Rituximab ® a human-mouse chimeric monoclonal antibody (mAh) that targets CD20 and inducers apoptosis in targeted cells.
- Rituximab ® is a prescription medicine made by Roche and is used to treat (i) Low-grade or follicular CD20-positive non-Hodgkin’s lymphoma as a single-agent therapy in patients whose disease recurred or did not respond to initial treatment, (ii) Follicular CD20-positive non-Hodgkin’s lymphoma as an initial treatment with chemotherapy, and in patients whose initial treatment was successful, as a single-agent follow-up therapy, (iii) Low-grade CD20-positive non-Hodgkin’s lymphoma as a single-agent follow-up therapy for patients who did not progress on initial treatment with CVP chemotherapy, (iv) CD20-positive diffuse large B-cell non-Hodgkin’s lymphoma as an initial treatment in combination with CHOP chemotherapy, and (v) CD20-positive chronic lymphocytic leukemia in combination with FC chemotherapy as an initial treatment or as a treatment after disease has recurred.
- an antibody is an antibody fragment. vi. Recombinant Antibody
- an antibody is a recombinant antibody.
- an antibody is a bi-specific antibody.
- a therapeutic agent is an immunotoxin.
- Various immunotoxins can be used to prepare a composition of the present invention or to use such composition in a method of the present invention.
- An immunotoxin typically comprises two parts - a targeting moiety and a cytotoxic moiety.
- the targeting moiety facilitates the transfer and internalization of the immunotoxin into a cell, e.g., a cancerous cell.
- the cytotoxic moiety kills the cell, typically by enzymatically inhibiting protein synthesis.
- an immunotoxin is selected from the group consisting of a recombinant immunotoxin, a PE-based immunotoxin, a DT- based immunotoxin, and combinations thereof.
- cells are exposed to or contacted with an effective amount of a composition comprising an immunotoxin wherein the contacting results in killing of the cells.
- the contacting results in killing of the cells and treatment of the subject.
- an immunotoxin is a recombinant immunotoxin.
- Recombinant immunotoxins are proteins that contain a toxin fused to an antibody (e.g., ADCs) or small molecules and are constructed by the genetic engineering technique. RITs can bind to and be internalized by cells and kill cancerous or non-cancerous cells by inhibiting protein synthesis.
- ADCs antibody-derived immunotoxins
- RITs can bind to and be internalized by cells and kill cancerous or non-cancerous cells by inhibiting protein synthesis.
- a wide variety of RITs have been tested against different cancers in cell culture, xenograft models, and human patients during the past several decades. RITs have shown activity in therapy of several kinds of cancers, but different levels of side effects, mainly related to vascular leak syndrome, were also observed in the treated patients.
- an immunotoxin is a PE-based immunotoxin.
- a PE-based immunotoxin comprises a Pseudomonas exotoxin (PE).
- PE is a single-chain protein, consisting of 613 amino acids in length, which is further composed of 3 functional domains. Domain la (amino acids 1-252) is the binding domain, domain II (amino acids 253-364) mediates translocating the toxin to the cytosol, and domain III (amino acids 400-613) contains the ADP-ribosylating enzyme that inactivates EF-2 in the cytosol. (See.
- an immunotoxin is a DT-based immunotoxin.
- a DT-based immunotoxin comprises a Diphtheria toxin (DT).
- DT Diphtheria toxin
- Full-length 535-amino acid DT is a single-chain protein containing an enzymatic A domain (amino acids 1-193) and a binding B domain (amino acids 482-535)
- a translocation or transmembrane (T) domain is located in the center of the molecule.
- the present invention provides pharmaceutical compounds, pharmaceutical combinations and pharmaceutical compositions and medicaments.
- a pharmaceutical compound, a pharmaceutical combination, a pharmaceutical composition or a medicament are useful for practicing a method of the present invention.
- the present invention provides pharmaceutical combinations and
- a first pharmaceutical composition comprises (i) an agonist of protein kinase C in an amount sufficient to achieve a desired effect, and (ii) a first pharmaceutically acceptable carrier.
- a first pharmaceutical compound is an agonist of protein kinase C in an amount sufficient to achieve a desired effect.
- Useful agonists of protein kinase C and pharmaceutically acceptable carriers are described herein.
- Preferred agonists of protein kinase C are bryostatin compounds.
- a preferred agonist of protein kinase C is Bryostatin- 1.
- the present invention provides pharmaceutical combinations and
- a second pharmaceutical composition comprises (i) a steroid in an amount sufficient to achieve a desired effect, and (ii) a second pharmaceutically acceptable carrier.
- a second pharmaceutical compound is a steroid in an amount sufficient to achieve a desired effect.
- Useful steroids and pharmaceutically acceptable carriers are described herein.
- a preferred steroid is a glucocorticoid.
- a preferred glucocorticoid is dexamethasone. Another preferred glucocorticoid is prednisolone.
- the first and second pharmaceutically acceptable carriers may be the same or different.
- the present invention provides pharmaceutical combinations and
- a third pharmaceutical composition comprises (i) a retinoic acid in an amount sufficient to achieve a desired effect, and (ii) a third pharmaceutically acceptable carrier.
- a third pharmaceutical compound is a retinoic acid in an amount sufficient to achieve a desired effect.
- Useful retinoic acids and pharmaceutically acceptable carriers are described herein.
- a preferred retinoic acid is tretinoin.
- the third pharmaceutically acceptable carries may be the same or different than the first or second pharmaceutically acceptable carrier.
- the present invention also provides pharmaceutical combinations and pharmaceutical compositions or medicaments comprising a therapeutic agent and, optionally, a fourth pharmaceutically acceptable carrier.
- a fourth pharmaceutical composition comprises (i) a therapeutic agent in an amount sufficient to achieve a desired effect, and (ii) a fourth pharmaceutically acceptable carrier.
- a fourth pharmaceutical compound is a therapeutic agent in an amount sufficient to achieve a desired effect.
- Useful therapeutic agents and pharmaceutically acceptable carriers are described herein.
- the fourth pharmaceutically acceptable carrier may be the same or different than the first, second or third
- the present invention provides pharmaceutical compositions or medicaments comprising an agonist of protein kinase C, a steroid, and, optionally, a first pharmaceutically acceptable carrier.
- a pharmaceutical composition comprises (i) an agonist of protein kinase C, (ii) a steroid, and (iii) a first pharmaceutically acceptable carrier.
- the agonist of protein kinase C and the steroid are provided in an amount sufficient to achieve a desired effect.
- Useful agonists of protein kinase C, steroids, and pharmaceutically acceptable carriers are described herein.
- Preferred agonists of protein kinase C are bryostatin compounds.
- a preferred agonist of protein kinase C is Bryostatin- 1.
- Preferred steroids are glucocorticoids.
- a preferred glucocorticoid is dexamethasone.
- Another preferred glucocorticoid is prednisolone.
- the present invention provides pharmaceutical compositions or medicaments comprising an agonist of protein kinase C, a retinoic acid, and, optionally, a second pharmaceutically acceptable carrier.
- a pharmaceutical composition comprises (i) an agonist of protein kinase C, (ii) a retinoic acid, and (iii) a second pharmaceutically acceptable carrier.
- the agonist of protein kinase C and the retinoic acid are provided in an amount sufficient to achieve a desired effect.
- Useful agonists of protein kinase C, retinoic acids, and pharmaceutically acceptable carriers are described herein.
- Preferred agonists of protein kinase C are bryostatin compounds.
- a preferred agonist of protein kinase C is Bryostatin-1.
- a preferred retinoic acid is tretinoin.
- the second pharmaceutically acceptable carrier may be the same or different than the first pharmaceutically acceptable carrier.
- the present invention provides pharmaceutical compositions or medicaments comprising an agonist of protein kinase C, a steroid or a retinoic acid, and, optionally, a third pharmaceutically acceptable carrier.
- a pharmaceutical composition comprises (i) an agonist of protein kinase C, (ii) a steroid or a retinoic acid, and (iii) a third pharmaceutically acceptable carrier.
- the agonist of protein kinase C and the steroid or the agonist of protein kinase C and the retinoic acid are provided in an amount sufficient to achieve a desired effect.
- agonists of protein kinase C are bryostatin compounds.
- a preferred agonist of protein kinase C is Bryostatin-1.
- Preferred steroids are glucocorticoids.
- a preferred glucocorticoid is dexamethasone. Another preferred
- glucocorticoid is prednisolone.
- a preferred retinoic acid is tretinoin.
- pharmaceutically acceptable carrier may be the same or different than the first and second pharmaceutically acceptable carrier.
- the present invention provides pharmaceutical compositions or medicaments comprising an agonist of protein kinase C, a steroid, a therapeutic agent, and, optionally, a first pharmaceutically acceptable carrier.
- a pharmaceutical composition comprises (i) an agonist of protein kinase C, (ii) a steroid, (iii) a therapeutic agent, and (iv) a first pharmaceutically acceptable carrier.
- the agonist of protein kinase C, the steroid and the therapeutic agent are provided in an amount sufficient to achieve a desired effect.
- Useful agonists of protein kinase C, steroids, therapeutic agents, and pharmaceutically acceptable carriers are described herein.
- Preferred agonists of protein kinase C are bryostatin compounds.
- a preferred agonist of protein kinase C is Bryostatin-1.
- Preferred steroids are glucocorticoids.
- a preferred glucocorticoid is dexamethasone.
- Another preferred glucocorticoid is prednisolone.
- Preferred therapeutic agents are an antibody mediating antibody-dependent cellular cytotoxicity, a monoclonal antibody, a humanized antibody, a chimeric antibody, a bi- specific antibody, an ADC, an antibody fragment, a recombinant antibody, or an
- the present invention provides pharmaceutical compositions or medicaments comprising an agonist of protein kinase C, a retinoic acid, a therapeutic agent, and, optionally, a second pharmaceutically acceptable carrier.
- a pharmaceutical composition comprises (i) an agonist of protein kinase C, (ii) a retinoic acid, (iii) a therapeutic agent, and (iv) a second pharmaceutically acceptable carrier.
- the agonist of protein kinase C, the retinoic acid and the therapeutic agent are provided in an amount sufficient to achieve a desired effect.
- Preferred agonists of protein kinase C are bryostatin compounds.
- a preferred agonist of protein kinase C is Bryostatin- 1.
- a preferred retinoic acid is tretinoin.
- Preferred therapeutic agents are an antibody mediating antibody-dependent cellular cytotoxicity, a monoclonal antibody, a humanized antibody, a chimeric antibody, a bi-specific antibody, an ADC, an antibody fragment, a recombinant antibody, or an immunotoxin.
- the second pharmaceutically acceptable carrier may be the same or different than the first pharmaceutically acceptable carrier.
- the present invention provides pharmaceutical compositions or medicaments comprising an agonist of protein kinase C, a steroid or a retinoic acid, a therapeutic agent, and, optionally, a third pharmaceutically acceptable carrier.
- a pharmaceutical composition comprises (i) an agonist of protein kinase C, (ii) a steroid or a retinoic acid, (iii) a therapeutic agent, and (iv) a third pharmaceutically acceptable carrier.
- the agonist of protein kinase C and the steroid or the agonist of protein kinase C and the retinoic acid and the therapeutic agent are provided in an amount sufficient to achieve a desired effect.
- agonists of protein kinase C are bryostatin compounds.
- a preferred agonist of protein kinase C is Bryostatin- 1.
- Preferred steroids are glucocorticoids.
- a preferred glucocorticoid is dexamethasone.
- Another preferred glucocorticoid is prednisolone.
- a preferred retinoic acid is tretinoin.
- Preferred therapeutic agents are an antibody mediating antibody-dependent cellular cytotoxicity, a monoclonal antibody, a humanized antibody, a chimeric antibody, a bi-specific antibody, an ADC, an antibody fragment, a recombinant antibody, or an immunotoxin.
- the third pharmaceutically acceptable carrier may be the same or different than the first and second pharmaceutically acceptable carrier.
- the present invention also provides an effective amount of a combination of an agonist of protein kinase C with a retinoic acid.
- the present invention also provides an effective amount of a combination of an agonist of protein kinase C with a steroid or with a retinoic acid.
- the present invention also provides an effective amount of a combination of an agonist of protein kinase C with a steroid and with a therapeutic agent.
- the present invention also provides an effective amount of a combination of an agonist of protein kinase C with a retinoic acid and with a therapeutic agent.
- the present invention also provides an effective amount of a combination of an agonist of protein kinase C with a steroid or with a retinoic acid and with a therapeutic agent.
- the present invention also provides pharmaceutical compositions or medicaments comprising (i) Bryostatin-1, (ii) dexamethasone and (iii) a pharmaceutically acceptable carrier.
- Compositions comprising (i) Bryostatin-1 and (ii) dexamethasone are referred to herein, from time to time, as ND1000.
- the present invention also provides an effective amount of a combination of Bryostatin-1 with a steroid.
- the present invention also provides an effective amount of a combination of Bryostatin-1 with a glucocorticoid.
- the present invention also provides an effective amount of a combination of Bryostatin-1 with dexamethasone.
- the present invention also provides pharmaceutical compositions or medicaments comprising (i) Bryostatin-1, (ii) prednisolone and (iii) a pharmaceutically acceptable carrier.
- Compositions comprising (i) Bryostatin-1 and (ii) prednisolone are referred to herein, from time to time, as ND1000B.
- the present invention also provides an effective amount of a combination of Bryostatin-1 with prednisolone.
- the present invention also provides pharmaceutical compositions or medicaments comprising (i) Bryostatin-1, (ii) tretinoin and (iii) a pharmaceutically acceptable carrier.
- Compositions comprising (i) Bryostatin-1 and (ii) tretinoin are referred to herein, from time to time, as ND1000C.
- the present invention also provides an effective amount of a combination of Bryostatin-1 with a retinoic acid.
- the present invention also provides an effective amount of a combination of Bryostatin-1 with tretinoin.
- a pharmaceutical compound, a pharmaceutical combination, a pharmaceutical composition or medicament as provided and described herein can be administered to a subject for the treatment of, for example, a condition or disease as described herein.
- a pharmaceutical combination, a pharmaceutical composition or a medicament may include any combination of agonists of protein kinase C, steroids, retinoic acids, or therapeutic agents.
- Compounds of the present invention are useful in the manufacture of a pharmaceutical combination, a pharmaceutical composition or a medicament comprising an effective amount thereof, optionally, in conjunction or in a mixture with excipients or carriers suitable for either enteral or parenteral application.
- compositions or medicaments for use in the present invention can be formulated by standard techniques using one or more
- physiologically acceptable carriers or excipients are described herein and in “ Remington's Pharmaceutical Sciences " by E. W. Martin.
- the small molecule compounds of the present invention and their physiologically acceptable salts and solvates can be formulated for administration by any suitable route, including via inhalation, topically, nasally, orally, parenterally, or rectally.
- the administration of a suitable route including via inhalation, topically, nasally, orally, parenterally, or rectally.
- compositions may be made by intradermal, subdermal, intravenous, intramuscular, intranasal, intracerebral, intratracheal, intraarterial, intraperitoneal, intravesical, intrapleural, intracoronary or intratumoral injection, with a syringe or other devices.
- Transdermal administration is also contemplated, as are inhalation or aerosol administration. Tablets and capsules can be administered orally, rectally or vaginally.
- a pharmaceutical compound, a pharmaceutical combination, a pharmaceutical composition or a medicament can take the form of, for example, a tablet or a capsule prepared by conventional means with a pharmaceutically acceptable excipient.
- binders e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose,
- polyvinylpyrrolidone and/or hydroxypropyl methylcellulose if desired (d) disintegrants, e.g., starches (e.g., potato starch or sodium starch), glycolate, agar, alginic acid or its sodium salt, or effervescent mixtures; (e) wetting agents, e.g., sodium lauryl sulphate, and/or (1) absorbents, colorants, flavors and sweeteners.
- disintegrants e.g., starches (e.g., potato starch or sodium starch), glycolate, agar, alginic acid or its sodium salt, or effervescent mixtures
- wetting agents e.g., sodium lauryl sulphate, and/or (1) absorbents, colorants, flavors and sweeteners.
- Tablets may be either film coated or enteric coated according to methods known in the art.
- Liquid preparations for oral administration can take the form of, for example, solutions, syrups, or suspensions, or they can be presented as a dry product for reconstitution with water or other suitable vehicle before use.
- Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives, for example, suspending agents, for example, sorbitol syrup, cellulose derivatives, or hydrogenated edible fats; emulsifying agents, for example, lecithin or acacia; non-aqueous vehicles, for example, almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils; and preservatives, for example, methyl or propyl-p-hydroxybenzoates or sorbic acid.
- the preparations can also contain buffer salts, flavoring, coloring, and/or sweetening agents as appropriate. If desired, preparations for oral administration can be suitably formulated to give controlled release of the active compound.
- Compounds, combinations, compositions, and medicaments of the present invention i.e., a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent
- a small molecule compound of the present invention an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent
- parenteral administration for injection, for example by bolus injection or continuous infusion.
- Formulations for injection can be presented in unit dosage
- Injectable compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are preferably prepared from fatty emulsions or suspensions.
- the combinations and compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers.
- the active ingredient can be in powder form for constitution with a suitable vehicle, for example, sterile pyrogen-free water, before use. In addition, they may also contain other therapeutically valuable substances.
- the combinations and compositions may be prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1 to 75%, preferably about 1 to 50%, of the active ingredient.
- compositions, and medicaments i.e., a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent
- a suitable propellant for example, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or other suitable gas.
- the dosage unit can be determined by providing a valve to deliver a metered amount.
- Capsules and cartridges of, for example, gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound and a suitable powder base, for example, lactose or starch.
- Suitable formulations for transdermal application include an effective amount of a compound, a combination, a composition or a medicament of the present invention (i.e., a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent) with a carrier or excipient.
- Preferred carriers and excipients include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
- transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
- Matrix transdermal formulations may also be used.
- Suitable formulations for topical application are preferably aqueous solutions, ointments, creams or gels well-known in the art. Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
- Compounds, combinations, compositions, and medicaments of the present invention i.e., a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent
- rectal compositions for example, suppositories or retention enemas, for example, containing conventional suppository bases, for example, cocoa butter or other glycerides.
- compounds, combinations, compositions, and medicaments of the present invention i.e., a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent
- a depot preparation i.e., a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent
- Such long-acting formulations can be administered by implantation (for example, subcutaneously or intra
- the compounds, combinations, compositions, and medicaments can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
- Compounds, combinations, compositions and medicaments of the present invention i.e., a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent
- a pack or dispenser device can contain one or more unit dosage forms containing the active ingredient.
- the pack can, for example, comprise metal or plastic foil, for example, a blister pack.
- the pack or dispenser device can be accompanied by instructions for administration.
- a pharmaceutical combination, a pharmaceutical composition or a medicament comprises an effective amount of an agonist of protein kinase C and a dexamethasone compound (as described herein, e.g., for the synergistic increase of expression of an antigen), and another therapeutic agent, such as a therapeutic agent binding to that antigen, as further described herein.
- a therapeutic agent may be used individually, sequentially, or in combination with one or more agonists of protein kinase C and a dexamethasone compound of the invention.
- Administration may be by the same or different route of administration or together in the same pharmaceutical formulation.
- a pharmaceutical composition comprises (i), an agonist of protein kinase C, (ii) a steroid, and (iii) a pharmaceutically acceptable carrier.
- a pharmaceutical combination is a combination of an agonist of protein kinase C with a steroid.
- composition comprises (i), an agonist of protein kinase C, (ii) a steroid, (iii) a therapeutic agent, and (iv) a pharmaceutically acceptable carrier.
- a pharmaceutical combination is a combination of an agonist of protein kinase C with a steroid and with a therapeutic agent.
- a pharmaceutical composition comprises (i), an agonist of protein kinase C, (ii) a retinoic acid, and (iii) a pharmaceutically acceptable carrier.
- a pharmaceutical combination is a combination of an agonist of protein kinase C with a retinoic acid.
- composition comprises (i), an agonist of protein kinase C, (ii) a retinoic acid, (iii) a therapeutic agent, and (iv) a pharmaceutically acceptable carrier.
- a pharmaceutical combination is a combination of an agonist of protein kinase C with a retinoic acid and with a therapeutic agent.
- a pharmaceutical combination or a pharmaceutical composition or medicament is administered to a subject, preferably a human, at a therapeutically effective dose to prevent, treat, or control a condition or disease as described herein, such as cancer.
- the pharmaceutical combination, pharmaceutical composition or medicament is administered to a subject in an amount sufficient to elicit an effective therapeutic response in the subject.
- An effective therapeutic response is a response that at least partially arrests or slows the symptoms or complications of the condition or disease. An amount adequate to accomplish this is defined as "therapeutically effective dose.”
- the dosage of active compounds, combinations and compositions of the present invention administered is dependent on the species of warm-blooded animal (mammal), the body weight, age, individual condition, surface area of the area to be treated and on the form of administration.
- the size of the dose also will be determined by the existence, nature, and extent of any adverse effects that accompany the administration of a particular small molecule compound in a particular subject.
- a unit dosage for oral administration to a mammal of about 50 to 70 kg may contain between about 5 and 500 mg of the active ingredient.
- a dosage of the active compounds of the present invention is a dosage that is sufficient to achieve the desired effect.
- Optimal dosing schedules can be calculated from measurements of compound accumulation in the body of a subject. In general, dosage may be given once or more daily, weekly, or monthly. Persons of ordinary skill in the art can easily determine optimum dosages, dosing methodologies and repetition rates.
- a pharmaceutical combination or a pharmaceutical composition or a medicament comprising a compound or compounds of the present invention is administered in a daily dose in the range from about 0.1 mg of each compound per kg of subject weight (0.1 mg/kg) to about 1 g/kg for multiple days.
- the daily dose is a dose in the range of about 5 mg/kg to about 500 mg/kg.
- the daily dose is about 10 mg/kg to about 250 mg/kg.
- the daily dose is about 25 mg/kg to about 150 mg/kg.
- a preferred dose is about 10 mg/kg.
- the daily dose can be administered once per day or divided into subdoses and administered in multiple doses, e.g., twice, three times, or four times per day.
- a small molecule compound of the present invention an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a
- glucocorticoid a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent may be administered in different amounts and at different times.
- compounds, combinations, and compositions of the present invention i.e., a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent
- a small molecule compound of the present invention i.e., an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent
- a small molecule compound of the present invention i.e., an agonist of protein kinase C, a
- therapeutically effective administration of compounds, combinations, and compositions of the present invention to treat a condition or disease described herein in a subject requires periodic (e.g., daily) administration that continues for a period ranging from three days to two weeks or longer.
- compounds, combinations, or compositions will be administered for at least three consecutive days, often for at least five consecutive days, more often for at least ten, and sometimes for 20, 30, 40 or more consecutive days. While consecutive daily doses are a preferred route to achieve a therapeutically effective dose, a therapeutically beneficial effect can be achieved even if the compounds are not administered daily, so long as the administration is repeated frequently enough to maintain a therapeutically effective concentration of the compounds in the subject.
- a preferred dosing schedule for example, is administering daily for a week, one week off and repeating this cycle dosing schedule for 3-4 cycles.
- Optimum dosages, toxicity, and therapeutic efficacy of such compounds, combinations and compositions may vary depending on the relative potency of individual compounds and can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, by determining the LD50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
- the dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio, LD 50 /ED 50 .
- Compounds, combinations and compositions that exhibit large therapeutic indices are preferred. While compounds, combinations and compositions that exhibit toxic side effects can be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue to minimize potential damage to normal cells and, thereby, reduce side effects.
- Data obtained from, for example, cell culture assays and animal studies can be used to formulate a dosage range for use in humans.
- the dosage of such small molecule compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
- the dosage can vary within this range depending upon the dosage form employed and the route of administration.
- the therapeutically effective dose can be estimated initially from cell culture assays.
- a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture.
- IC 50 the concentration of the test compound that achieves a half-maximal inhibition of symptoms
- levels in plasma can be measured, for example, by high performance liquid chromatography (HPLC).
- HPLC high performance liquid chromatography
- the dose equivalent of compounds is from about 1 ng/kg to 100 mg/kg for a typical subject.
- kits are also provided by the present invention.
- kits and components can be prepared according to the present invention, depending upon the intended user of the kit and the particular needs of the user. As such, a skilled artisan will appreciate that the below description of kits is not limiting.
- kits may include any or all of the following: assay reagents, buffers, a compound of the present invention (i.e., a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent), an antigen polypeptide, an antigen nucleic acid, an anti-antigen polypeptide antibody, hybridization probes and/or primers, expression constructs for e.g., a lymphocyte antigen, an antigen expressed in a cancerous cell, etc.
- a therapeutic product may include sterile saline or another pharmaceutically acceptable emulsion and suspension base.
- a kit comprises one or more of a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, prednisolone, a retinoic acid, tretinoin, or a therapeutic agent.
- these compounds are provided in a container.
- kits for practicing methods of the present invention including, but not limited to, increasing expression of an antigen in a cancerous cell and inhibiting the growth of a cancerous cell.
- the kits comprise compositions described herein for practicing a method of the present invention.
- a kit comprises (i) a first container containing an agonist of protein kinase C in an amount sufficient to achieve a desired effect as described herein, (ii) a second container containing a steroid in an amount effective to achieve a desired effect as described herein; (iii) a third container containing a therapeutic agent in an amount effective to achieve a desired effect as described herein; and (iv) an instruction for using (i) - (iii) for practicing methods of the present invention, including, but not limited to, increasing expression of an antigen in a cancerous cell, and inhibiting the growth of a cancerous cell.
- a kit comprises (i) a first container containing an agonist of protein kinase C in an amount sufficient to achieve a desired effect as described herein, (ii) a second container containing a retinoic acid in an amount effective to achieve a desired effect as described herein; (iii) a third container containing a therapeutic agent in an amount effective to achieve a desired effect as described herein; and (iv) an instruction for using (i) - (iii) for practicing methods of the present invention, including, but not limited to, increasing expression of an antigen in a cancerous cell, and inhibiting the growth of a cancerous cell.
- kits for practicing methods of the present invention including, but not limited to, increasing expression of an antigen in a cancerous cell and inhibiting the growth of a cancerous cell.
- the kits comprise compositions described herein for practicing a method of the present invention.
- a kit comprises (i) a first container containing an agonist of protein kinase C in an amount sufficient to achieve a desired effect as described herein, (ii) a second container containing a steroid or a retinoic acid in an amount effective to achieve a desired effect as described herein; (iii) a third container containing a therapeutic agent in an amount effective to achieve a desired effect as described herein; and (iv) an instruction for using (i) - (iii) for practicing methods of the present invention, including, but not limited to, increasing expression of an antigen in a cancerous cell, and inhibiting the growth of a cancerous cell.
- the instructional materials may contain directions (i.e., protocols) for the practice of methods of this invention.
- the instructions may be present in the subject kits in a variety of forms, one or more of which may be present in the kit. While the instructional materials typically comprise written or printed materials they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated by this invention. Such media include, but are not limited to electronic storage media (e.g., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. Such media may include addresses to internet sites that provide such instructional materials.
- the instructional material comprises warnings of possible side effects and drug-drug or drug-food interactions.
- a kit is a pharmaceutical kit and comprises a pharmaceutical composition comprising (i) a small molecule compound of the present invention, an agonist of protein kinase C, a bryostatin compound, Bryostatin-1, a steroid, a glucocorticoid, a dexamethasone compound, dexamethasone, a retinoic acid, tretinoin, and/or a therapeutic agent, and (ii) a pharmaceutical acceptable carrier.
- kits optionally comprise an instruction stating that the pharmaceutical composition can or should be used for treating a condition or disease described herein.
- Additional kit embodiments of the present invention include optional functional components that would allow one of ordinary skill in the art to perform any of the method variations described herein.
- the present invention describes a variety of methods using compounds, combinations, compositions, medicaments, and kits of the invention.
- a compound, a combination, a composition, a medicament, or a kit as described herein is used in a method for increasing expression of an antigen in a cancerous cell.
- a compound, a combination, a composition, a medicament, a kit or an article of manufacture as described herein is used in a method for increasing expression of an antigen on the cell surface of a cancerous cell.
- both methods will be referred to herein as “increasing expression of an antigen” and specifics will be pointed out and/or will be apparent to one of skill in the art.
- various cancerous cells can be contacted with one or more compositions or with one or more combinations of the present invention to increase expression of the antigen.
- a cancerous cell is obtained from a subject having a hematologic cancer.
- a cancerous cell obtained from a subject having a hematologic cancer may be contacted with one or more compositions of the present invention.
- a cancerous cell obtained from a subject having a hematologic cancer is selected from the group consisting of B-cell lymphoma, T-cell lymphoma, leukocytic leukemia, acute leukocytic leukemia, chronic leukocytic leukemia, lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, lymphocytic leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, monocytic leukemia, multiple myeloma, myelodysplastic syndrome, myelogenous leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, myeloid leukemia, acute myeloid leuk
- compositions and methods of the present invention can be used by one of ordinary skill in the art to increase expression of a variety of antigens, e.g., the expression of an antigen in a cancerous cell, preferably, the expression of an antigen in a lymphocytic cancerous cell, and even more preferably, the expression of an antigen in a B-lymphocytic cell.
- antigens e.g., the expression of an antigen in a cancerous cell, preferably, the expression of an antigen in a lymphocytic cancerous cell, and even more preferably, the expression of an antigen in a B-lymphocytic cell.
- compositions and methods of the present invention can be used by one of ordinary skill in the art to increase expression of a variety of antigens on the cell surface of a cancerous cell, preferably, the expression of an antigen on the cell surface of a lymphocytic cancerous cell, and even more preferably, the expression of an antigen on the cell surface of a B-lymphocytic cell.
- the antigen is selected from the group consisting of CDlb, CDlc, CDld, CD2, CD3d, CD3e, CD3g, CD4, CD5, CD6, CD7, CD8a, CD8b, CD10, CDl la, CDl lb, CDl lc, CD13, CD14, CD 16a, CD 16b, CD18, CD19, CD20, CD21, CD22, CD23, CD25, CD26, CD27, CD28, CD29, CD30, CD31, CD32, CD33, CD34, CD35, CD40, CD41, CD42d, CD44, CD45,
- CD213al CD213a2, CD217, CD218a, CD220, CD221, CD222, CD226, CD228, CD229, CD230, CD232, CD239, CD243, CD244, CD265, CD269, CD272, CD273, CD274, CD275,
- an antigen is CD 19. In some embodiments of the present invention, an antigen is CD 20.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a first composition comprising an agonist of protein kinase C.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with an agonist of protein kinase C.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a first composition comprising a bryostatin compound.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a bryostatin compound.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a first composition comprising Bryostatin-1.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with Bryostatin-1.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising a steroid.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a steroid.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising a glucocorticoid.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a glucocorticoid.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising a dexamethasone compound.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a dexamethasone compound.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising dexamethasone.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with dexamethasone.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising prednisolone.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with prednisolone.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with the first composition and the second composition in an amount effective to increase expression of the antigen in the cancerous cell, preferably, to increase expression of the antigen on the surface of the cancerous cell. Thereby expression of the antigen in the cancerous cell, preferably on the cell surface of the cancerous cell is increased.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with the an effective amount of a combination with an agonist of protein kinase C with a steroid or with a retinoic acid. Thereby expression of the antigen in the cancerous cell, preferably on the cell surface of the cancerous cell is increased.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a third composition comprising a retinoic acid.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a retinoic acid.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a third composition comprising tretinoin.
- a method for increasing expression of an antigen in a cancerous cell preferably, of a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with tretinoin.
- the step of contacting a cancerous cell includes contacting a plurality of cancerous cells. Cancerous cells that can be contacted with combinations or compositions of the present invention are described herein. Further, it will be apparent to one of ordinary skill in the art that the step of contacting a cancerous cell with a first composition may be performed prior to contacting the cancerous cell with a second composition or third composition. Alternatively, the step of contacting a cancerous cell with a first composition may be performed after the step of contacting the cancerous cell with a second or third composition. It is also possible to perform both steps at the same time. As explained herein, the terms first, second, third, or fourth, as used in the context of a composition is merely for convenience of description and distinction and does in no way imply any order.
- a method for increasing expression of an antigen comprises the step of contacting a cancerous cell with an agonist of protein kinase C or with a first composition comprising an agonist of protein kinase C and wherein the agonist of protein kinase C isselected from the group consisting of ingenol-3-angelate, prostratin, staurospine, valproate, tamoxifen, chelerythrine, miyabenol C, verbascoside, BIM-1, indolocarbazole, isoquinolinesulfonamide, sphingosine, calphostin C (UCN-1028C), bisindolylmaleimide GF 109203X, a plant flavonoid, TNF-b, IL-Ib, a lipopolysaccharide,
- UV -light a CD3 antibody, a CD3/CD28 antibody combination, etoposide, daunorubicin, hydrogen peroxide, nocodazole, LIGHT, bleomycin, camptothecin, cisplatin, celecoxib, ciprofibrate, cycloprodigiosin, dacarbazine, Daio-Orengedeokuto, daunomycin, diazoxide, diclofenac, 5,6-dimethylxanthenone-4-acetic acid, flavone-8-acetic acid, haloperidol, imiquimod, isochamaejasmin, lithium, mitoxantrone, morphine, nipradilol, norepinephrine, nystatin, oltipraz, protocatechuic acid, SN38 (metabolite of CPT-11), Taxol (Paclitaxel), vinblastine, vincristine, WR1065, a bryo
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a bryostatin compound or with a first composition comprising a bryostatin compound.
- a bryostatin compound or with a first composition comprising a bryostatin compound.
- a bryostatin compound is selected from the group consisting of Bryostatin-1, Bryostatin-2, Bryostatin-3, Bryostatin-4, Bryostatin-5, Bryostatin-6, Bryostatin-7, Bryostatin-8, Bryostatin-9, Bryostatin- 10, Bryostatin-11, Bryostatin- 12, Bryostatin-13, Bryostatin- 14, Bryostatin- 15, Bryostatin- 16, Bryostatin- 17, Bryostatin-18, Bryostatin-19, Bryostatin-20, a bryostatin compound having Formula 1, a bryostatin compound having Formula 2, a bryostatin compound having Formula 3, a bryostatin compound having Formula 4, a bryostatin compound having Formula 5, a bryostatin compound having Formula 6, a bryostatin compound having Formula 7,
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with Bryostatin- 1 or with a first composition Bryostatin-1.
- Bryostatin- 1 or with a first composition Bryostatin-1.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-1.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising a steroid.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a steroid.
- Useful steroids are described herein. Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of those steroids.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising a glucocorticoid.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a glucocorticoid.
- Useful glucocorticoids are described herein. Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of those glucocorticoids.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising a dexamethasone compound.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a dexamethasone compound.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising dexamethasone.
- a method for increasing expression of an antigen in a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with dexamethasone.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of dexamethasone.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a second composition comprising prednisolone.
- a method for increasing expression of an antigen in a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with prednisolone.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of prednisolone.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a third composition comprising a retinoic acid.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a retinoic acid.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of retinoic acid.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of contacting a cancerous cell with a third composition comprising tretinoin.
- a method for increasing expression of an antigen in a cancerous cell comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with tretnoin.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of tretinoin.
- a method for increasing expression of an antigen on the cell surface of a cancerous cell comprises the step of synergistically increasing the expression of the antigen. This is achieved by contacting the cancerous cell with both the first composition comprising an agonist of protein kinase C and the second composition comprising a steroid or with both the first composition comprising an agonist of protein kinase C and the third composition comprising a retinoic acid.
- this is achieved by contacting the cancerous cell with a combination of an agonist of protein kinase C with a steroid or with a combination of an agonist of protein kinase C with a retinoic acid. Likewise, this is achieved by contacting the cancerous cell with a combination of an agonist of protein kinase C with a steroid and with a retinoic acid.
- a method for increasing expression of an antigen in a cancerous cell preferably, a method for increasing expression of an antigen on the cell surface of a cancerous cell
- the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C, with a steroid and with a retinoic acid.
- a method for increasing expression of an antigen in a cancerous cell preferably, a method for increasing expression of an antigen on the cell surface of a cancerous cell, the method is practiced in vitro.
- a method for increasing expression of an antigen in a cancerous cell preferably, a method for increasing expression of an antigen on the cell surface of a cancerous cell
- the method is practiced in vivo.
- the method comprises the step of administering a combination or a first composition comprising an agonist of protein kinase C and a second composition comprising a steroid or a first composition comprising an agonist of protein kinase C and a third composition comprising a retinoic acid to a subject in need of having expression of the antigen increased.
- a compound, a combination, a composition, a medicament, a kit or an article of manufacture as described herein is used in a method for inhibiting the growth of a cancerous cell.
- a compound, a combination, a composition, a medicament, a kit or an article of manufacture as described herein is used in a method for inhibiting the growth of a cancerous lymphocyte cell.
- a compound, a combination, a composition, a medicament, a kit or an article of manufacture as described herein is used in a method for inhibiting the growth of a cancerous B-lymphocyte cell.
- those methods will be referred to herein as“inhibiting the growth of a cancerous cell” and specifics will be pointed out and/or will be apparent to one of skill in the art.
- the growth of a variety of cancerous cells can be inhibited using a compound, a combination, a composition, a medicament, a kit, an article of manufacture, or method of the present invention.
- various cancerous cells can be contacted with one or more compounds, combinations or compositions of the present invention to inhibit their growth.
- a cancerous cell is from a subject having a cancer selected from the group consisting of a lung cancer, a sarcoma, a gastrointestinal cancer, a cancer of the genitourinary tract, a liver cancer, a skin cancer, a gynecological cancer, a bone cancer, a cancer of the nervous system, a cancer of adrenal gland, and a hematologic cancer.
- a cancerous cell is obtained from a subject having a hematologic cancer.
- a cancerous cell obtained from a subject having a hematologic cancer may be contacted with one or more compositions of the present invention.
- a cancerous cell obtained from a subject having a hematologic cancer is selected from the group consisting of B-cell lymphoma, T-cell lymphoma, leukocytic leukemia, acute leukocytic leukemia, chronic leukocytic leukemia, lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, lymphocytic leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, monocytic leukemia, multiple myeloma, myelodysplastic syndrome, myelogenous leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, myeloid leukemia, acute my
- compositions and methods of the present invention can be used by one of ordinary skill in the art to inhibit the growth of a cancerous cell.
- Those cancerous cells may express a variety of antigens.
- a cancerous cell expresses an antigen selected from the group consisting of CDlb, CDlc, CDld, CD2, CD3d, CD3e, CD3g, CD4, CD5, CD6, CD7, CD8a, CD8b, CD10, CDl la, CDl lb, CDl lc, CD13, CD14, CD16a, CD16b, CD18, CD 19, CD20, CD21, CD22, CD23, CD25, CD26, CD27, CD28, CD29, CD30, CD31, CD32, CD33, CD34, CD35, CD40, CD41, CD42d, CD44, CD45, CD46, CD47, CD48, CD49A, CD49b, CD49c,
- an antigen is CD 20.
- the method comprises the step of contacting a cancerous cell with a first composition comprising an agonist of protein kinase C. In some embodiments of a method for inhibiting the growth of a cancerous cell, the method comprises the step of contacting a cancerous cell with an agonist of protein kinase C.
- the method comprises the step of contacting a cancerous cell with a second composition comprising a steroid or with a third composition comprising a retinoic acid. In some embodiments of a method for inhibiting the growth of a cancerous cell, the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a steroid or with a combination of an agonist of protein kinase C with a retinoic acid.
- the method comprises the step of contacting a cancerous cell with the first composition and the second composition or with the first composition and the third composition in an amount effective to inhibit the growth of the cancerous cell.
- the method comprises the step of contacting a cancerous cell with an effective amount of a combination of an agonist of protein kinase C with a steroid or with a combination of an agonist of protein kinase C with a retinoic acid. Thereby growth of the cancerous cell is inhibited.
- the step of contacting a cancerous cell includes contacting a plurality of cancerous cells. Cancerous cells that can be contacted with a compound, a combination or a composition of the present invention that are described herein. Further, it will be apparent to one of ordinary skill in the art that the step of contacting a cancerous cell with a first compound or first composition may be performed prior to contacting the cancerous cell with a second or third compound or composition. Alternatively, the step of contacting a cancerous cell with a first compound or composition may be performed after the step of contacting the cancerous cell with a second or third compound or composition. It is also possible to perform both steps at the same time. A cancerous cell can also be contacted with a combination described herein.
- a method for inhibiting the growth of a cancerous cell comprises the step of contacting a cancerous cell with an agonist of protein kinase C or with a first composition comprising an agonist of protein kinase C and wherein the agonist of protein kinase C is selected from the group consisting of ingenol-3-angelate, prostratin, staurospine, valproate, tamoxifen, chelerythrine, miyabenol C, verbascoside, BIM- 1, indolocarbazole, isoquinolinesulfonamide, sphingosine, calphostin C (UCN-1028C), bisindolylmaleimide GF 109203X, a plant flavonoid, TNF-b, IL-Ib, a lipopolysaccharide,
- UV -light a CD3 antibody, a CD3/CD28 antibody combination, etoposide, daunorubicin, hydrogen peroxide, nocodazole, LIGHT, bleomycin, camptothecin, cisplatin, celecoxib, ciprofibrate, cycloprodigiosin, dacarbazine, Daio-Orengedeokuto, daunomycin, diazoxide, diclofenac, 5,6-dimethylxanthenone-4-acetic acid, flavone-8-acetic acid, haloperidol, imiquimod, isochamaejasmin, lithium, mitoxantrone, morphine, nipradilol, norepinephrine, nystatin, oltipraz, protocatechuic acid, SN38 (metabolite of CPT-11), Taxol (Paclitaxel), vinblastine, vincristine, WR1065, a bryo
- the method comprises the step of contacting a cancerous cell with a bryostatin compound or with a first composition comprising a bryostatin compound.
- a bryostatin compound is selected from the group consisting of
- the method comprises the step of contacting a cancerous cell with Bryostatin-1 or with a first composition comprising Bryostatin-1.
- Bryostatin-1 or with a first composition comprising Bryostatin-1.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of Bryostatin-1.
- the method comprises the step of contacting a cancerous cell with a second composition comprising a steroid. In some embodiments of a method for inhibiting the growth of a cancerous cell, the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a steroid.
- Useful steroids are described herein. Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of those steroids.
- the method comprises the step of contacting a cancerous cell with a second composition comprising a glucocorticoid.
- the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a glucocorticoid.
- Useful glucocorticoids are described herein. Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of those glucocorticoids.
- the method comprises the step of contacting a cancerous cell with a second composition comprising a dexamethasone compound.
- the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a dexamethasone compound.
- Useful dexamethasone compounds are described herein. Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of those dexamethasone compounds.
- the method comprises the step of contacting a cancerous cell with a second composition comprising dexamethasone.
- the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with dexamethasone.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of dexamethasone.
- the method comprises the step of contacting a cancerous cell with a second composition comprising prednisolone.
- the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with prednisolone.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of prednisolone.
- the method comprises the step of synergistically increasing expression of an antigen. This is achieved by contacting the cancerous cell with both the first composition comprising an agonist of protein kinase C and the second composition comprising a steroid. Likewise, this is achieved by contacting the cancerous cell with a combination of an agonist of protein kinase C with a steroid.
- the method comprises the step of contacting a cancerous cell with a third composition comprising a retinoic acid.
- the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with a retinoic acid.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of retinoic acids.
- the method comprises the step of contacting a cancerous cell with a third composition comprising tretinoin.
- the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C with tretinoin.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of tretinoin.
- the method comprises the step of synergistically increasing expression of an antigen.
- the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C, with a steroid and with a retinoic acid.
- the method comprises the step of contacting a cancerous cell with a fourth composition comprising a therapeutic agent.
- the method comprises the step of contacting a cancerous cell with a combination of an agonist of protein kinase C, a steroid and a therapeutic agent or with a combination of an agonist of protein kinase C, a retinoic acid and a therapeutic agent or with a combination of an agonist of protein kinase C, a steroid, a retinoic acid, and a therapeutic agent.
- Useful therapeutic agents are described herein.
- the method comprises the step of contacting a cancerous cell with a therapeutic agent or a fourth composition comprising a therapeutic agent and wherein the therapeutic compound is selected from the group consisting of an antibody mediating antibody-dependent cellular cytotoxicity, a monoclonal antibody, a humanized antibody, a chimeric antibody, a bi-specific antibody, an ADC, an antibody fragment, a recombinant antibody, and an immunotoxin.
- the method comprises the step of contacting a cancerous cell with a monoclonal antibody or a fourth composition comprising a monoclonal antibody.
- a monoclonal antibody or a fourth composition comprising a monoclonal antibody.
- a variety of monoclonal antibodies may be used.
- the method comprises the step of contacting a cancerous cell with a fourth composition comprising a monoclonal antibody wherein the monoclonal antibody is Ofatumumab ® .
- the method comprises the step of contacting a cancerous cell with a chimeric antibody or a fourth composition comprising a chimeric antibody.
- a variety of chimeric antibodies may be used.
- the method comprises the step of contacting a cancerous cell with Rituximab ® or a fourth composition comprising Rituximab ® .
- a cancerous cell may first be contacted with an agonist of protein kinase C or with a first composition comprising an agonist of protein kinase C. Thereafter, the cancerous cell may be contacted with a second composition comprising a steroid. After that step, the cancerous cell may be contacted with a fourth composition comprising a therapeutic agent.
- a cancerous cell is first contacted with steroid or with a second composition comprising a steroid.
- the cancerous cell is contacted with an agonist of protein kinase C or with a first composition comprising an agonist of protein kinase C.
- the cancerous cell may be contacted with a therapeutic agent or with a fourth composition comprising a therapeutic agent.
- a cancerous cell may first be contacted with an agonist of protein kinase C or with a first composition comprising an agonist of protein kinase C. Thereafter, the cancerous cell may be contacted with a retinoic acid or with a third composition comprising a retinoic acid. After that step, the cancerous cell may be contacted with a therapeutic agent or with a fourth composition comprising a therapeutic agent.
- a cancerous cell is first contacted with a retinoic acid or with a third composition comprising a retinoic acid. Thereafter, the cancerous cell is contacted with an agonist of protein kinase C or with a first composition comprising an agonist of protein kinase C. After that step, the cancerous cell may be contacted with a therapeutic agent or with a fourth composition comprising a therapeutic agent.
- a cancerous cell is first contacted with an agonist of protein kinase C or with a first composition comprising an agonist of protein kinase C. Thereafter, the cancerous cell is contacted with a steroid or with a second composition comprising a steroid. Thereafter, the cancerous cell is contacted with a retinoic acid or with a third composition comprising a retinoic acid. After those steps, the cancerous cell is contacted with a therapeutic agent or with a fourth composition comprising a therapeutic agent.
- a therapeutic agent or with a fourth composition comprising a therapeutic agent.
- a cancerous cell is first contacted with a retinoic acid or with a third composition comprising a retinoic acid, then with a steroid or with a second composition comprising a steroid. Thereafter, the cancerous cell is contacted with an agonist of protein kinase C or with a first composition comprising an agonist of protein kinase C. After that step, the cancerous cell is contacted with a therapeutic agent or with a fourth composition comprising a therapeutic agent.
- a cancerous cell may also be contacted simultaneously or sequentially with (i) a combination of an agonist of protein kinase C with a steroid, (ii) a combination of an agonist of protein kinase C with a retinoic acid, (iii) a combination of an agonist of protein kinase C with a steroid and with a therapeutic agent, (iv) a combination of an agonist of protein kinase C with a retinoic acid and with a therapeutic agent, and (v) a combination of an agonist of protein kinase C with a steroid, with a retinoic acid, and with a therapeutic agent.
- a cancerous cell may also be contacted simultaneously or sequentially with (i) a combination of one or more agonists of protein kinase C with one or more steroids, (ii) a combination of one or more agonists of protein kinase C with one or more retinoic acids, (iii) a combination of one or more agonists of protein kinase C with one or more steroids and with one or more therapeutic agents, (iv) a combination of one or more agonists of protein kinase C with one or more retinoic acids and with one or more therapeutic agents, and (v) a combination of one or more agonists of protein kinase C with one or more steroids, with one or more retinoic acids, and with one or more therapeutic agents.
- the method comprises the step of combining a first composition comprising an agonist of protein kinase C and a second composition comprising a steroid to form a single composition.
- the method comprises the step of contacting a cancerous cell with a first composition comprising an agonist of protein kinase C and a second composition comprising a steroid at about the same time. This permits the first and the second composition to be administered in combination.
- the method comprises the step of combining a first composition comprising an agonist of protein kinase C and a third composition comprising a retinoic acid to form a single composition.
- the method comprises the step of contacting a cancerous cell with a first composition comprising an agonist of protein kinase C and a third composition comprising a retinoic acid at about the same time. This permits the first and the third composition to be administered in combination.
- the method comprises the step of combining a first composition comprising an agonist of protein kinase C, a second composition comprising a steroid, and a third composition comprising a retinoic acid to form a single composition.
- the method comprises the step of contacting a cancerous cell with a first composition comprising an agonist of protein kinase C, with a second composition comprising a steroid, and a third composition comprising a retinoic acid at about the same time. This permits the first, the second, and the third composition to be administered in combination.
- the method comprises the step of combining a first composition comprising an agonist of protein kinase C, a second composition comprising a steroid, and a fourth composition comprising a therapeutic agent to form a single composition.
- a first composition comprising an agonist of protein kinase C
- a second composition comprising a steroid
- a fourth composition comprising a therapeutic agent
- a method for inhibiting the growth of a cancerous cell comprises the step of contacting a cancerous cell with a first composition comprising an agonist of protein kinase C, with a second composition comprising a steroid and with a fourth composition comprising a therapeutic agent at about the same time. This permits the first, the second, and the fourth composition to be administered in combination.
- the method comprises the step of combining a first composition comprising an agonist of protein kinase C, a third composition comprising a retinoic acid, and a fourth composition comprising a therapeutic agent to form a single composition.
- a first composition comprising an agonist of protein kinase C
- a third composition comprising a retinoic acid
- a fourth composition comprising a therapeutic agent
- a method for inhibiting the growth of a cancerous cell comprises the step of contacting a cancerous cell with a first composition comprising an agonist of protein kinase C, with a third composition comprising a retinoic acid and with a fourth composition comprising a therapeutic agent at about the same time. This permits the first, the third, and the fourth composition to be administered in combination.
- the method comprises the step of combining a first composition comprising an agonist of protein kinase C, a second composition comprising a steroid, a third composition comprising a retinoic acid, and a fourth composition comprising a therapeutic agent to form a single composition.
- the method comprises the step of contacting a cancerous cell with a first composition comprising an agonist of protein kinase C, with a second composition comprising a steroid, with a third composition comprising a retinoic acid, and with a fourth composition comprising a therapeutic agent at about the same time.
- a first composition comprising an agonist of protein kinase C
- a second composition comprising a steroid
- a third composition comprising a retinoic acid
- a fourth composition comprising a therapeutic agent at about the same time.
- the method is practiced in vitro. In some embodiments of a method for inhibiting the growth of a cancerous cell, the method is practiced ex vivo.
- the method is practiced in vivo.
- the method comprises the step of administering a first composition comprising an agonist of protein kinase C to a subject in need of having the growth of a cancerous cell inhibited.
- the method comprises the step of administering a second composition comprising a steroid to a subject in need of having the growth of a cancerous cell inhibited.
- the method comprises the step of administering a third composition comprising a retinoic acid to a subject in need of having the growth of a cancerous cell inhibited.
- the method comprises the step of administering a fourth composition comprising therapeutic agent to a subject in need of having the growth of a cancerous cell inhibited.
- inhibition of the growth of the cancerous cell is achieved when the growth relative to a control is reduced by about 10%, by about 20%, by about 30%, by about 40%, by about 50%, by about 60%, by about 70%, by about 80%, by about 90, or by about 100%. Further, inhibition of the growth of a cancerous cell is achieved when the growth relative to a control is reduced by at least about 10%, by at least about 20%, by at least about 30%, by at least about 40%, by at least about 50%, by at least about 60%, by at least about 70%, by at least about 80%, or by at least about 90. Assays for determining inhibition of the growth of a cancerous cell are known in the art.
- Methods of the present invention can be practiced in vitro, ex vivo and in vivo.
- methods for treating a subject suffering from a cancerous condition i.e., methods for treating a subject having cancer
- a cancerous condition typically is characterized by unwanted growth or proliferation of a cell (a cancerous cell).
- a cancerous cell expresses on its cell surface one or more antigens, also referred to as, cell surface markers, which can be targeted using a composition of the present invention.
- a cancer is selected from the group consisting of a lung cancer, a sarcoma, a gastrointestinal cancer, a cancer of the genitourinary tract, a liver cancer, a skin cancer, a gynecological cancer, a bone cancer, a cancer of the nervous system, a cancer of adrenal gland, and a hematologic cancer.
- a cancer is selected from the group consisting of a lung cancer, a sarcoma, a gastrointestinal cancer, a cancer of the genitourinary tract, a liver cancer, a skin cancer, a gynecological cancer, a bone cancer, a cancer of the nervous system, a cancer of adrenal gland, and a hematologic cancer, each expressing one or more antigens or cell surface markers to which a therapeutic agent can bind.
- a preferred cancer is a hematologic cancer.
- a compound, a combination or a composition of the present invention is used to treat a subject having a lung cancer.
- a lung cancer includes, but is not limited to, bronchogenic carcinoma [squamous cell,
- a cancerous lung cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a lung cancer is treated with one or more compounds, combinations or compositions of the present invention.
- a compound, a combination, or a composition of the present invention is used to treat a subject having a sarcoma.
- a sarcoma includes, but is not limited to, cancers such as angiosarcoma, fibrosarcoma,
- rhabdomyosarcoma liposarcoma, myxoma, rhabdomyoma, fibroma, lipoma, and teratoma.
- a cancerous sarcoma cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a sarcoma is treated with one or more compounds, combinations, or compositions of the present invention.
- a compound, a combination or a composition of the present invention is used to treat a subject having a gastrointestinal cancer.
- a gastrointestinal cancer includes, but is not limited to cancers of esophagus
- stomach cancer [squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma], stomach
- a cancerous gastrointestinal cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a gastrointestinal cancer is treated with one or more compounds, combinations or compositions of the present invention.
- a compound, a combination or a composition of the present invention is used to treat a subject having a cancer of the genitourinary tract.
- Cancers of the genitourinary tract include, but are not limited to cancers of kidney [adenocarcinoma, Wilms tumor (nephroblastoma), lymphoma, leukemia, renal cell carcinoma], bladder and urethra [squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma], prostate [adenocarcinoma, sarcoma], and testis [seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, Leydig cell tumor, fibroma, fibroadenoma, adenomatoid tumors, lipoma].
- a cancerous genitourinary tract cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a genitourinary cancer is treated with one or more compounds, combinations or compositions of the present invention.
- a compound, a combination or a composition of the present invention is used to treat a subject having a liver cancer.
- a liver cancer includes, but is not limited to, hepatocellular carcinoma, cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, and hemangioma.
- a cancerous liver cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a liver cancer is treated with one or more compounds, combinations or compositions of the present invention.
- a compound, a combination, or a composition of the present invention is used to treat a subject having a skin cancer.
- a skin cancer includes, but is not limited to, malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, nevi, dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, and psoriasis.
- a cancerous skin cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a skin cancer is treated with one or more compounds, combinations or compositions of the present invention.
- a compound, a combination or a composition of the present invention is used to treat a subject having a gynecological cancer.
- a gynecological cancer includes, but is not limited to, cancer of uterus [endometrial carcinoma], cervix [cervical carcinoma, pre-invasive cervical dysplasia], ovaries [ovarian carcinoma (serous cystadenocarcinoma, mucinous cystadenocarcinoma, endometrioid carcinoma, clear cell adenocarcinoma, malignant mixed mullerian tumors, unclassified carcinoma), granulosa-theca cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma and other germ cell tumors], vulva [squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma], vagina [clear cell carcinoma,
- a cancerous gynecological cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a gynecological cancer is treated with one or more compounds, combinations or compositions of the present invention.
- a compound, a combination or a composition of the present invention is used to treat a subject having a bone cancer.
- a bone cancer includes, but is not limited to, osteogenic sarcoma [osteosarcoma], fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma [reticulum cell sarcoma], multiple myeloma, malignant giant cell tumor, chordoma, osteochondroma [osteocartilaginous exostoses], benign chondroma, chondroblastoma, chondromyxoid fibroma, osteoid osteoma, and giant cell tumors.
- a cancerous bone cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a bone cancer is treated with one or more compounds, combinations or compositions of the present invention.
- a compound, a combination or a composition of the present invention is used to treat a subject having a cancer of the nervous system.
- a cancer of the nervous system includes, but is not limited to cancer of skull [osteoma, hemangioma, granuloma, xanthoma, Paget's disease of bone], meninges
- a cancerous cell of the nervous system expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a cancer of the nervous system is treated with one or more compounds, combinations or compositions of the present invention.
- a compound, a combination or a composition of the present invention is used to treat a subject having a cancer of adrenal glands.
- a cancer of adrenal glands includes, but is not limited to, neuroblastoma.
- a cancerous adrenal gland cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a cancer of adrenal glands is treated with one or more compounds, combinations or compositions of the present invention
- a compound, a combination, or a composition of the present invention is used to treat a subject having a hematologic cancer.
- a hematologic cancer includes, but is not limited to cancer of blood [B- and T-cell lymphomas, myeloid leukemia (acute and chronic), myelocytic leukemia (acute and chronic), mixed lineage leukemia, Waldenstrom's Macroglobulinemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic leukocytic leukemia, acute myelogenous leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, lymphoblastic leukemia, lymphocytic leukemia, monocytic leukemia,
- a cancerous hematologic cell expresses one or more antigens to which a therapeutic agent can bind.
- a subject having a hematologic cancer is treated with one or more compounds, combinations, or compositions of the present invention.
- hematologic cancers specifically, lymphomas and leukemia
- will often have different names e.g., due to changing systems of classification, and that patients having lymphomas and leukemia classified under different names may also benefit from the therapeutic regimens of the present invention.
- a subject having lymphoma is treated with one or more compounds, combinations or compositions of the present invention.
- a subject having leukemia is treated with one or more compounds, combinations or compositions of the present invention.
- a subject having acute lymphocytic leukemia (ALL) is treated with one or more compounds, combinations or compositions of the present invention.
- a subject having chronic lymphocytic leukemia (CLL) is treated with one or more compounds, combinations or compositions of the present invention.
- a subject having mast cell leukemia is treated with one or more compounds, combinations or compositions of the present invention.
- a subject having acute myeloid leukemia (AML) is treated with one or more compounds, combinations or compositions of the present invention.
- a subject having chronic myeloid leukemia (CML) is treated with one or more compounds, combinations or compositions of the present invention.
- a cancerous cell expresses one or more antigens, preferably the cancerous cell expresses one or more antigens of the cell surface of the cancerous cell. As described herein, those antigens can be targeted with a therapeutic compound.
- cancerous cells may express a variety of antigens.
- a cancerous cell expresses an antigen selected from the group consisting of CDlb, CDlc,
- CD49f CD50, CD51, CD54, CD55, CD56, CD58, CD59, CD61, CD62L, CD62P, CD63, CD64, CD66a, CD66b, CD66c, CD68, CD71, CD73, CD74, CD79a, CD79b, CD80, CD83, CD84, CD85f, CD85i, CD85j, CD86, CD87, CD89, CD90, CD91, CD92, CD95, CD96, CD97, CD98, CD100, CD101, CD102, CD103, CD104, CD105, CD106, CD107a, CD107b, CD108, CD 109, CD111, CD112, CD113, CD116, CD117, CD118, CD119, CD120a,
- an antigen is CD 19. In some embodiments of the present invention, an antigen is CD20. In some embodiments of the present invention, an antigen is CD22. In some embodiments of the present invention, an antigen is CD30. In some embodiments of the present invention, an antigen is CD38. In some embodiments of the present invention, an antigen is CD70. In some embodiments of the present invention, an antigen is CD269.
- the method comprises the step of administering to a subject having cancer and in need thereof an agonist of protein kinase C or a first pharmaceutical composition comprising an agonist of protein kinase C and, optionally, a first pharmaceutically acceptable carrier. Thereby, the subject is treated.
- the method comprises the step of administering to a subject having cancer and in need thereof a steroid or a second pharmaceutical composition comprising a steroid and, optionally, a second pharmaceutically acceptable carrier. Thereby, the subject is treated.
- the method comprises the step of administering to the subject the first pharmaceutical composition and the second pharmaceutical composition in an amount effective to increase expression of an antigen on the cell surface of a subject’s cancerous cell. Thereby expression of the antigen on the cell of the subject’s cancerous cell is increased. Thereby, the subject is treated.
- the method comprises the step of administering to the subject an effective amount of a combination of an agonist of protein kinase C with a steroid. Thereby expression of the antigen on the cell of the subject’s cancerous cell is increased. Thereby, the subject is treated.
- the method comprises the step of administering to a subject having cancer and in need thereof a retinoic acid or a third pharmaceutical composition comprising a retinoic acid and, optionally, a third pharmaceutically acceptable carrier. Thereby, the subject is treated.
- the method comprises the step of administering to the subject the first pharmaceutical composition and the third pharmaceutical composition in an amount effective to increase expression of an antigen on the cell surface of a subject’s cancerous cell. Thereby expression of the antigen on the cell of the subject’s cancerous cell is increased. Thereby, the subject is treated.
- the method comprises the step of administering to the subject an effective amount of a combination of an agonist of protein kinase C with a retinoic acid. Thereby expression of the antigen on the cell of the subject’s cancerous cell is increased. Thereby, the subject is treated.
- Various protein kinase C agonists can be used in the methods of the present invention.
- the method comprises the step of administering to a subject having cancer and in need thereof an agonist of protein kinase C or a first pharmaceutical composition comprising an agonist of protein kinase C, and, optionally, a first pharmaceutically acceptable carrier.
- an agonist of protein kinase C is selected from the group consisting of ingenol- 3-angelate, prostratin, staurospine, valproate, tamoxifen chelerythrine, miyabenol C, verbascoside, BIM-1, indolocarbazole, isoquinolinesulfonamide, sphingosine, calphostin C (UCN-1028C), bisindolylmaleimide GF 109203X, a plant flavonoid, TNF-b, IL-Ib, a lipopolysaccharide, UV-light, a CD3 antibody, a CD3/CD28 antibody combination, etoposide, daunorubicin, hydrogen peroxide, nocodazole, LIGHT, bleomycin, camptothecin, cisplatin, celecoxib, ciprofibrate, cycloprodigiosin, dacarbazine, Dai
- the method comprises the step of administering to a subject having cancer and in need thereof a bryostatin compound or a first pharmaceutical composition comprising a bryostatin compound and, optionally, a first pharmaceutically acceptable carrier. Thereby, the subject is treated.
- a bryostatin compound or a first pharmaceutical composition comprising a bryostatin compound and, optionally, a first pharmaceutically acceptable carrier.
- a bryostatin compound is selected from the group consisting of Bryostatin- 1, Bryostatin-2, Bryostatin-3, Bryostatin-4, Bryostatin-5, Bryostatin-6, Bryostatin-7, Bryostatin-8, Bryostatin-9, Bryostatin- 10, Bryostatin-11, Bryostatin- 12, Bryostatin-13, Bryostatin- 14, Bryostatin- 15, Bryostatin- 16, Bryostatin- 17, Bryostatin- 18, Bryostatin- 19, Bryostatin-20, a bryostatin compound having Formula 1, a bryostatin compound having Formula 2, a bryostatin compound having Formula 3, a bryostatin compound having Formula 4, a bryostatin compound having Formula 5, a bryostatin compound having Formula 6, a bryostatin compound having Formula 7, single stereoisomers,
- the method comprises the step of administering to a subject having cancer and in need thereof Bryostatin-1 or a first pharmaceutical composition comprising Bryostatin-1 and, optionally, a first pharmaceutically acceptable carrier. Thereby, the subject is treated.
- Bryostatin-1 or a first pharmaceutical composition comprising Bryostatin-1 and, optionally, a first pharmaceutically acceptable carrier.
- a method for treating a subject having cancer comprises the step of administering to a subject having cancer and in need thereof a steroid or a second pharmaceutical composition comprising a steroid and, optionally, a second pharmaceutically acceptable carrier.
- a steroid or a second pharmaceutical composition comprising a steroid and, optionally, a second pharmaceutically acceptable carrier.
- the subject is treated.
- Useful steroids and carriers are described herein. Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of those steroids.
- a method for treating a subject having cancer comprises the step of administering to a subject having cancer and in need thereof a glucocorticoid or a second pharmaceutical composition comprising a glucocorticoid and, optionally, a second pharmaceutically acceptable carrier.
- a glucocorticoid or a second pharmaceutical composition comprising a glucocorticoid and, optionally, a second pharmaceutically acceptable carrier.
- glucocorticoids and carriers are described herein. Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of those glucocorticoids.
- the method comprises the step of administering to a subject having cancer and in need thereof a dexamethasone compound or a second pharmaceutical composition comprising a
- dexamethasone compound and, optionally, a second pharmaceutically acceptable carrier. Thereby, the subject is treated.
- Useful dexamethasone compounds and carriers are described herein. Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of a dexamethasone compound.
- a method for treating a subject having cancer comprises the step of administering to a subject having cancer and in need thereof dexamethasone or a second pharmaceutical composition comprising dexamethasone and, optionally, a second pharmaceutically acceptable carrier. Thereby, the subject is treated.
- dexamethasone or a second pharmaceutical composition comprising dexamethasone and, optionally, a second pharmaceutically acceptable carrier.
- the subject is treated.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of dexamethasone.
- the method comprises the step of administering to a subject having cancer and in need thereof prednisolone or a second pharmaceutical composition comprising prednisolone and, optionally, a second pharmaceutically acceptable carrier. Thereby, the subject is treated.
- prednisolone or a second pharmaceutical composition comprising prednisolone and, optionally, a second pharmaceutically acceptable carrier.
- the method comprises the step of administering to a subject having cancer and in need thereof a retinoic acid or a third pharmaceutical composition comprising a retinoic acid and, optionally, a third pharmaceutically acceptable carrier.
- a retinoic acid or a third pharmaceutical composition comprising a retinoic acid and, optionally, a third pharmaceutically acceptable carrier.
- the subject is treated.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of retinoic acids.
- the method comprises the step of administering to a subject having cancer and in need thereof tretinoin or a third pharmaceutical composition comprising tretinoin and, optionally, a third pharmaceutically acceptable carrier.
- tretinoin or a third pharmaceutical composition comprising tretinoin and, optionally, a third pharmaceutically acceptable carrier.
- the subject is treated.
- Useful are also single stereoisomers, mixtures of stereoisomers, pharmaceutically acceptable salts, esters, prodrugs and functional derivatives of tretinoin.
- the method comprises the step of synergistically increasing expression of an antigen. This is achieved by administering to a subject having cancer and in need thereof both the first pharmaceutical composition comprising an agonist of protein kinase C and the second pharmaceutical composition comprising a steroid or both the first pharmaceutical composition comprising an agonist of protein kinase C and the third pharmaceutical composition comprising a retinoic acid. Likewise, this is achieved by administering to a subject having cancer and in need thereof an effective amount of a combination of an agonist of protein kinase C with a steroid or an effective amount of a combination of an agonist of protein kinase C a retinoic acid. Thereby, the subject is treated.
- the method comprises the step of administering to a subject having cancer and in need thereof a therapeutic agent or a fourth pharmaceutical composition comprising a therapeutic agent and, optionally, a fourth pharmaceutically acceptable carrier. Thereby, the subject is treated.
- a therapeutic agent or a fourth pharmaceutical composition comprising a therapeutic agent and, optionally, a fourth pharmaceutically acceptable carrier.
- the method comprises the step of administering to the subject the therapeutic agent or the fourth pharmaceutical composition in an amount effective to bind to the antigen on the cell surface of the subject’s cancerous cell.
- the therapeutic agent binds to the antigen on the cell of the subject’s cancerous cell.
- the subject is treated.
- the method comprises the step of administering to a subject having cancer and in need thereof a therapeutic agent or a fourth pharmaceutical composition comprising a therapeutic agent selected from the group consisting of an antibody mediating antibody-dependent cellular cytotoxicity, a monoclonal antibody, a humanized antibody, a chimeric antibody, a bi- specific antibody, an ADC, an antibody fragment, a recombinant antibody, and an immunotoxin.
- a therapeutic agent selected from the group consisting of an antibody mediating antibody-dependent cellular cytotoxicity, a monoclonal antibody, a humanized antibody, a chimeric antibody, a bi- specific antibody, an ADC, an antibody fragment, a recombinant antibody, and an immunotoxin.
- the method comprises the step of administering to a subject having cancer and in need thereof a chimeric antibody or a fourth pharmaceutical composition comprising a chimeric antibody and, optionally, a fourth pharmaceutically acceptable carrier.
- a variety of chimeric antibodies may be used.
- the method comprises the step of administering to a subject having cancer and in need thereof Rituximab ® or a fourth pharmaceutical composition comprising Rituximab ® and, optionally, a fourth pharmaceutically acceptable carrier. Thereby, the subject is treated.
- the method comprises the step of administering to a subject having cancer and in need thereof a monoclonal antibody or a fourth pharmaceutical composition comprising a monoclonal antibody and, optionally, a fourth pharmaceutically acceptable carrier.
- the method comprises the step of administering to a subject having cancer and in need thereof Ofatumumab ® or a fourth pharmaceutical composition comprising Ofatumumab ® and, optionally, a fourth
- a subject having cancer and in need thereof is administered first an agonist of protein kinase or a first pharmaceutical composition comprising an agonist of protein kinase C. Thereafter, a subject having cancer and in need thereof is administered steroid or a second pharmaceutical composition comprising a steroid or a retinoic acid or a third pharmaceutical composition comprising a retinoic acid. After that step, a subject having cancer and in need thereof is administered a therapeutic agent or a fourth pharmaceutical composition comprising a therapeutic agent.
- a subject having cancer and in need thereof is administered first a steroid or a second pharmaceutical composition comprising a steroid or a retinoic acid or a third pharmaceutical composition comprising a retinoic acid.
- a subject having cancer and in need thereof is administered an agonist of protein kinase C or a first pharmaceutical composition comprising an agonist of protein kinase C.
- a subject having cancer and in need thereof is administered a therapeutic agent or a fourth pharmaceutical composition comprising a therapeutic agent.
- a combination of an agonist of protein kinase C with a steroid (i) a combination of an agonist of protein kinase C with a retinoic acid, (iii) a combination of an agonist of protein kinase C with a steroid and with a therapeutic agent, (iv) a combination of an agonist of protein kinase C with a retinoic acid and with a therapeutic agent, and (v) a combination of an agonist of protein kinase C with a steroid, with a retinoic acid, and with a therapeutic agent, can be simultaneously or sequentially administered to the subject.
- a method for treating a subject having cancer (i) a combination of one or more agonists of protein kinase C with one or more steroids, (ii) a combination of one or more agonists of protein kinase C with one or more retinoic acids, (iii) a combination of one or more agonists of protein kinase C with one or more steroids and with one or more therapeutic agents, (iv) a combination of one or more agonists of protein kinase C with one or more retinoic acids and with one or more therapeutic agents, and (v) a combination of one or more agonists of protein kinase C with one or more steroids, with one or more retinoic acids, and with one or more therapeutic agents may be simultaneously or sequentially administered to the subject.
- the method comprises the step of combining a first pharmaceutical composition comprising an agonist of protein kinase C and a second pharmaceutical composition comprising a steroid to form a single pharmaceutical composition.
- a single pharmaceutical composition comprising an agonist of protein kinase C and a steroid will be administered to a subject having cancer and in need thereof.
- Such single pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers.
- the method comprises the step of combining an agonist of protein kinase C with a steroid to form a combination.
- a combination of an agonist of protein kinase C with a steroid will be administered to a subject having cancer and in need thereof. Thereby, the subject is treated.
- the method comprises the step of administering to a subject having cancer and in need thereof a first pharmaceutical composition comprising an agonist of protein kinase C and a second pharmaceutical composition comprising a steroid at about the same time. This also permits the first and the second pharmaceutical compositions to be administered in combination. Thereby, the subject is treated.
- the method comprises the step of combining a first pharmaceutical composition comprising an agonist of protein kinase C and a third pharmaceutical composition comprising a retinoic acid to form a single pharmaceutical composition. In such an embodiment, a single
- composition comprising an agonist of protein kinase C and a retinoic acid will be administered to a subject having cancer and in need thereof.
- composition may further comprise one or more pharmaceutically acceptable carriers.
- the method comprises the step of combining an agonist of protein kinase C with a retinoic acid to form a combination.
- a combination of an agonist of protein kinase C with a retinoic acid will be administered to a subject having cancer and in need thereof. Thereby, the subject is treated.
- the method comprises the step of administering to a subject having cancer and in need thereof a first pharmaceutical composition comprising an agonist of protein kinase C and a third pharmaceutical composition comprising a retinoic acid at about the same time.
- a first pharmaceutical composition comprising an agonist of protein kinase C
- a third pharmaceutical composition comprising a retinoic acid
- a method for treating a subject having cancer comprises the step of combining a first pharmaceutical composition comprising an agonist of protein kinase C, a second pharmaceutical composition comprising a steroid, and a fourth pharmaceutical composition comprising a therapeutic agent to form a single pharmaceutical composition.
- a single pharmaceutical composition comprising an agonist of protein kinase C, a steroid, and a therapeutic agent is administered to a subject having cancer and in need thereof.
- Such single pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers.
- the method comprises the step of combining an agonist of protein kinase C with a steroid and with a therapeutic agent to form a combination.
- a combination of an agonist of protein kinase C with a steroid and with a therapeutic agent will be administered to a subject having cancer and in need thereof. Thereby, the subject is treated.
- the method comprises the step of administering to a subject having cancer and need thereof a first pharmaceutical composition comprising an agonist of protein kinase C, a second pharmaceutical composition comprising a steroid and a fourth pharmaceutical composition comprising a therapeutic agent at about the same time. This permits the first, the second, and the fourth pharmaceutical composition to be administered in combination.
- a method for treating a subject having cancer comprises the step of combining a first pharmaceutical composition comprising an agonist of protein kinase C, a third pharmaceutical composition comprising a retinoic acid, and a fourth pharmaceutical composition comprising a therapeutic agent to form a single pharmaceutical composition.
- a single pharmaceutical composition comprising an agonist of protein kinase C, a retinoic acid, and a therapeutic agent is administered to a subject having cancer and in need thereof.
- Such single pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers.
- the method comprises the step of combining an agonist of protein kinase C with a retinoic acid and with a therapeutic agent to form a combination.
- a combination of an agonist of protein kinase C with a retinoic acid and with a therapeutic agent will be administered to a subject having cancer and in need thereof. Thereby, the subject is treated.
- the method comprises the step of administering to a subject having cancer and need thereof a first pharmaceutical composition comprising an agonist of protein kinase C, a third pharmaceutical composition comprising a retinoic acid and a fourth pharmaceutical composition comprising a therapeutic agent at about the same time.
- a first pharmaceutical composition comprising an agonist of protein kinase C
- a third pharmaceutical composition comprising a retinoic acid
- a fourth pharmaceutical composition comprising a therapeutic agent
- a method for treating a subject having cancer comprises the step of combining a first pharmaceutical composition comprising an agonist of protein kinase C, a second pharmaceutical composition comprising a steroid, a third pharmaceutical composition comprising a retinoic acid, and a fourth pharmaceutical composition comprising a therapeutic agent to form a single pharmaceutical composition.
- a single pharmaceutical composition comprising an agonist of protein kinase C, a steroid, a retinoic acid, and a therapeutic agent is administered to a subject having cancer and in need thereof.
- Such single pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers.
- the method comprises the step of combining an agonist of protein kinase C with a steroid, with a retinoic acid, and with a therapeutic agent to form a combination.
- a combination of an agonist of protein kinase C with a steroid, with a retinoic acid, and with a therapeutic agent will be administered to a subject having cancer and in need thereof. Thereby, the subject is treated.
- the method comprises the step of administering to a subject having cancer and need thereof a first pharmaceutical composition comprising an agonist of protein kinase C, a second pharmaceutical composition comprising a steroid, a third pharmaceutical composition comprising a retinoic acid, and a fourth pharmaceutical composition comprising a therapeutic agent at about the same time.
- a first pharmaceutical composition comprising an agonist of protein kinase C
- a second pharmaceutical composition comprising a steroid
- a third pharmaceutical composition comprising a retinoic acid
- a fourth pharmaceutical composition comprising a therapeutic agent at about the same time.
- Co-administration in the context of this invention includes the administration of more than one compound of the present invention in the course of a coordinated treatment to achieve an improved clinical outcome.
- Such co-administration may also be coextensive, that is, occurring during overlapping periods of time.
- Peripheral blood or bone marrow samples from patients with CLL, MCL, or B-ALL were assigned a sample ID upon receipt.
- the sample IDs consisted of (1) three digit patient identifier (2) a three letter indication identifier (ALL, CLL, or MCL) and (3) a sampling number for that patient (first sample received from that patient given sampling number of 1 and subsequent samples from same patient would receive the next increment).
- the exemplary sample with ID 674CLL1 corresponds to the 674th sample received at Applicant’s facility (“674”), the patient had been diagnosed with CLL (“CLL”) and that sample was the first sample received from that patient (“1”).
- 674CLL1 and 674CLL2 refer to distinct samples from the same patient, and may include different cells/ tissue or the same cells/ tissue serially sampled.
- Red blood cells were removed from remnant CLL MCL and ALL peripheral blood specimens using RBC Lysis Buffer (Thermo Fisher Scientific, Waltham, MA). The suspension of mononuclear cells was suspended in StemSpan SFEM II medium
- HS-5 cells ATCC, Manassas, VA
- FBS Thermo Fisher Scientific, Waltham, MA
- High Glucose DMEM ATCC, Manassas, VA
- HS-5 cells were maintained below passage 10 and in a 37°C, 95% humidity, 5% CO2 incubator.
- some patient samples were plated in 384 well V-bottom plates (Greiner Bio-One, Kremsmiinster, Austria) at a density of 1 million cells/mL.
- StemSpan SFEM II medium supplemented as described above was further supplemented at varying concentrations with one or more cytokines: 0-200 ng/mL of IL6 (R&D Systems, Minneapolis, MN), 0-200 ng/mL of IL7 (Miltenyi, Bergisch Gladbach, Germany), 0-200 ng/mL of G-CSF (Miltenyi, Bergisch Gladbach, Germany), 0-200 ng/mL of GM-CSF (R&D Systems, Minneapolis, MN), 0-200 ng/mL of IL3 (R&D Systems, Minneapolis, MN), 0-200 ng/mL FLT3 Ligand (Miltenyi, Bergisch Gladbach, Germany), 0-200 ng/mL of SCF
- Bryostatin-1, Dexamethasone, Prednisolone, and Tretinoin were solubilized in DMSO and added at the concentrations indicated to cells in StemSpan SFEM IIC medium and plated into 384 well compound plates (Labcyte, San Jose, CA). Patient samples were compounded in triplicate with using a Labcyte Echo acoustic liquid handler. Compound plates were used only once and discarded after one freeze thaw. Samples were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours.
- the viable lymphocyte populations were gated by side scatter vs CD45 and B-cells were further gated from this population by CD 19 expression. T-cells were also gated from the lymphocyte population using CD3 as well as CD5 and were then used as an internal negative control for CD19, CD20, and CD22 expression, respectively.
- abnormal B-cells were distinguished from healthy B-cells by the presence of CD5 expression.
- abnormal B-cells are identified as CD19 + , CD45 dim (or negative).
- CD 19 expression status as determined by flow cytometric screening was compared against independent clinical assessment of CD19 status for the CLL patient samples.
- CD 19 negative or positive expression were defined using the geometric mean fluorescent intensity (gMFI) of CD 19 expression in abnormal B-cell population compared to the internal negative control T-cell population which are known to be CD3 positive and CD 19 negative.
- CD 19 positive populations were defined as having a gMFI .2 log greater than the negative control. 6. Assessment of CD20 Expression
- CD20 expression status as determined by flow cytometric screening was compared against independent clinical assessment of CD20 status for CLL and MCL patient samples.
- CD20 negative, dim, and positive expression were defined using the geometric mean fluorescent intensity (gMFI) of CD20 expression in abnormal B-cell population compared to the internal negative control T-cell population, which are CD20 negative.
- CD20 positive populations were defined as having a gMFI 1 log greater than the negative control, whereas CD20 dim population had between .1 to 1 log greater than the negative control.
- CD20 negative B-cell populations had gMFI within .1 logs of the negative control.
- CD22 expression status as determined by flow cytometric screening was compared against independent clinical assessment of CD22 status for the CLL/ALL/MCL patient samples.
- CD22 negative or positive expression were defined using the geometric mean fluorescent intensity (gMFI) of CD22 expression in abnormal B-cell population compared to the internal negative control T-cell population which are known to be CD3 positive and CD22 negative.
- CD22 positive populations were defined as having a gMFI .2 log greater than the negative control. .
- Red blood cells were removed from remnant CLL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously.
- Patient sample was plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1,
- Dexamethasone, and/or DMSO and the indicated concentrations. Samples were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- a CD20 positive CLL patient sample (Sample ID No. 674CLL2; CD20 status: CD20 + ) was prepared, primed and analyzed by flow cytometry as described in the methods above with Bryostatin-1 (1 nM), Dexamethasone (50 nM), and a combination of Bryostatin-1 (1 nM) and Dexamethasone (50 nM) to increase expression of CD20.
- This patient had previously received treatment involving anti-CD20 therapy, Rituximab ® , and had relapsed. It was hypothesized that CLL patients that had previously been treated with an anti-CD20 therapy may be less responsive to priming of CD20 with single agent Bryostatin-1 due to compensatory changes in cell intracellular signaling networks or clonal selection.
- FIG. 3A schematically depicting a histogram plot of CD20 staining intensity indicates that as single agents, Bryostatin-1 and dexamethasone have moderate effects on increasing CD20 antigen expression. However, when combination of Bryostatin-1 and dexamethasone is administered, a substantially and synergistically increase of expression of CD20 antigen is achieved. Similarly, the representation as depicted in FIG. 3B indicates that while the single agents show moderate increases of the CD20 antigen (mid cells), the combination of the two agents results in a substantial and synergistic increase in the CD20 high fraction of cells.
- Red blood cells were removed from remnant CLL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously.
- Patient sample was plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1,
- Dexamethasone, and/or DMSO at the indicated concentrations were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma- Aldrich, St. Louis, MO) and fluorescent-labelled primary antibodies: CD 19, CD20, CD5, CD45, CD22, CD3, and CD66B (Biolegend, San Diego, CA). Stains were directly dispensed into the patient samples plates using a Labcyte Echo acoustic liquid handler and then incubated at 4°C for 20 minutes.
- DAPI Sigma- Aldrich, St. Louis, MO
- CD20 negative CLL patient (Sample ID No. 764CLL1; CD20 status: CD20 ) was prepared, primed and analyzed by flow cytometry as described in the methods above with Bryostatin-1 (5 nM), Dexamethasone (50 nM), and a combination of Bryostatin-1 (5 nM) and
- Dexamethasone 50 nM to increase expression of CD20.
- FIGs. 4A-B An exemplary and representative result of this study is shown in FIGs. 4A-B.
- the addition of single agent Bryostatin-1 partially restored CD20 antigen expression to the abnormal B-cells and dexamethasone had an even more moderate effect.
- dexamethasone and Bryostatin-1 led to a substantial and synergistic increase of both the number of cells positive for CD20 (FIG. 4B) as well as in the intensity of CD20 on those primed cells (FIG. 4A). Without priming, this patient sample would have remained CD20 negative, however, upon administering a combination of Bryostatin-1 and dexamethasone the CD20 negative cells regained CD20 antigen expression and thus, become sensitive to anti-CD20 therapy.
- Red blood cells were removed from remnant CLL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously. Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1,
- Dexamethasone, and/or DMSO at the indicated concentrations were then incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- FIGs. 5A-B Sample ID No. 671CLL1
- FIGs. 5C-D Sample ID No. 725CLL1.
- both patient samples show little response to Bryostatin-1 as a single agent or to dexamethasone as a single agent.
- the aberrant B-cell populations of both patients show a substantial and synergistic increase in CD20 expression.
- Results described herein strongly indicate that increasing expression of CD20 on cells of CD20 dim patients renders those cells sensitive to anti-CD20 therapies.
- Red blood cells were removed from remnant CLL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously.
- Patient sample was plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1, Dexamethasone, and/or DMSO at the indicated concentrations.
- Samples were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- CD20 positive CLL patient (Sample ID No. 707CLL1, CD20 status: CD20 + ) were prepared, primed and analyzed by flow cytometry as described in the methods above with Bryostatin-1 (1 nM), Dexamethasone (200 nM), and a combination of Bryostatin-1 (1 nM) and Dexamethasone (200 nM) to increase expression of CD20.
- FIGs. 6A-B An exemplary and representative result of this study is shown in FIGs. 6A-B. As depicted in FIGs. 6A-6B, this patient sample shows a moderate increase in CD20 antigen expression in response to Bryostatin-1 alone or in response to dexamethasone alone.
- Example F CD20 Priininp Of Cells Obtained From CLL Patients
- Red blood cells were removed from remnant CLL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously. Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1,
- Dexamethasone, and/or DMSO at the indicated concentrations were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- FIGs. 7A-B Exemplary and representative results of this study is shown in FIGs. 7A-B.
- An increase in the log of the CD20 antigen expression (gMFI) compared to the vehicle treated control greater than .1 was taken as the threshold for a positive priming response.
- the response rate when administering Bryostatin-1 as a single agent was 25%; however, in combination with Dexamethasone the response rate increased to 75%.
- Example G CD 19 Priming Of Cells Obtained From CLL Patients
- Red blood cells were removed from remnant CLL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously. Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1,
- Dexamethasone, and/or DMSO at the concentrations indicated. Samples were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- FIGs. 8A-B Exemplary and representative results of this study is shown in FIGs. 8A-B.
- An increase in the log of the CD 19 antigen expression (gMFI) compared to the vehicle treated control greater than .1 was taken as the threshold for a positive priming response.
- the response rate when administering Bryostatin-1 as a single agent was 66%; however, in combination with Dexamethasone the response rate increased to 72%. Results described strongly suggest that increasing expression of CD19 antigen on cancerous cells of patients will further sensitize those cells to anti-CD 19 therapies.
- Red blood cells were removed from the remnant CLL peripheral blood specimen using RBC Lysis Buffer and cultured as described previously.
- Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1, Dexamethasone, and/or DMSO at the concentration indicated. Samples were then incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- CD 19 positive CLL patient (CD 19 status: positive) were prepared, primed and analyzed by flow cytometry as described in the methods above with Bryostatin-1 (1 nM), Dexamethasone (200 nM), and a combination of Bryostatin-1 (1 nM) and Dexamethasone (200 nM) to increase expression of CD 19.
- FIGs. 9A-B An exemplary and representative result of this study is shown in FIGs. 9A-B. As depicted in FIGs. 9A-6B, this patient sample shows no increase of CD 19 antigen expression with dexamethasone alone and an increase in CD 19 antigen expression in response to Bryostatin-1 alone. However, this response is further increased upon
- Red blood cells were removed from remnant a B-ALL peripheral blood specimen using RBC Lysis Buffer and cultured as described previously.
- Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1, Dexamethasone, and/or DMSO at the concentration indicated.
- Samples were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- CD 19, CD20, CD5, CD45, CD22, CD3, and CD66B Biolegend, San Diego, CA. Stains were directly dispensed into the patient samples plates using a Labcyte Echo acoustic liquid handler and then incubated at 4°C for 20 minutes. Samples were then assayed using an iQue Screener PLUS flow cytometer (Intellicyt, Albuquerque, NM) and data was analyzed in FlowJo (V10). The viable lymphocyte populations were gated by side scatter vs CD45 and B-cells were further gated from this population by CD19 expression. T-cells were also gated from the lymphocyte population using CD3 as well as CD5 and were then used as an internal negative control for CD 19 expression. Abnormal B-cells are identified as CD19+, CD45 dim (or negative).
- Cells from an ALL patient (Sample ID No. 825ALL1, CD19 status: positive) were prepared, primed and analyzed by flow cytometry as described in the methods above with Bryostatin-1 (1 nM), Dexamethasone (200 nM), and a combination of Bryostatin-1 (1 nM) and Dexamethasone (200 nM) to increase expression of CD 19.
- FIGs. 10A-B An exemplary and representative result of this study is shown in FIGs. 10A-B. As depicted in FIGs. 10A-6B, this patient sample shows no only a moderate increase of CD 19 antigen expression upon administering dexamethasone alone and a higher increase in CD 19 antigen expression in response to Bryostatin-1 alone. However, this response is further increased upon administering a combination of Bryostatin-1 and dexamethasone. Increasing expression of CD 19 antigen on cells of patients with earlier B-cell malignancies (see, FIG. 1) might be expected to further sensitize responsive patient to anti-CD 19 therapies. Results described herein strongly suggest that increasing expression of CD 19 antigen on cancerous cells of ALL patients will further sensitize those cells to anti-CD 19 therapies.
- Red blood cells were removed from remnant CLL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously. Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1,
- Dexamethasone, and/or DMSO at the concentrations indicated. Samples were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- CD20 positive CLL patient (Sample ID No. 674CLL2; CD20 status: CD20 + ) were prepared, primed and analyzed by flow cytometry as described in the methods above with various combinations of Bryostatin-1 (1 nM, 2.5 nM, or 5 nM) and Dexamethasone (50 nM, 100 nM, or 200 nM) to increase expression of CD20.
- FIGs. 11 A-B Exemplary and representative results of this study is shown in FIGs. 11 A-B. While in this patient sample, increasing concentrations of Bryostatin-1 showed further increased priming of CD20, this trend reversed with the use of low dose Dexamethasone (FIG. 11B). As demonstrated by this study, lower doses of Bryostatin-1 in combination with dexamethasone were more effectively to prime CD20 expression.
- Red blood cells were removed from a remnant CLL peripheral blood specimen using RBC Lysis Buffer and cultured as described previously. Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1,
- Dexamethasone, and/or DMSO at the concentration indicated were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- CD20 dim CLL patient (Sample ID No.735CLLl, CD20 status: CD20 dim) were prepared, primed and analyzed by flow cytometry as described in the methods above with Bryostatin-1 (1 nM,), with Dexamethasone (200 nM) or with a combination of Bryostatin-1 and Dexamethasone to increase expression of CD20.
- Bryostatin-1 (1 nM,
- Dexamethasone 200 nM
- a combination of Bryostatin-1 and Dexamethasone to increase expression of CD20.
- this rare patient sample neither combination nor single agents increased CD20 expression.
- this rare patient sample provides a unique opportunity to identify underlying mechanism of non responsiveness to the compounds of the present invention.
- Red blood cells were removed from remnant CLL and MCL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously.
- Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin- 1, Dexamethasone, and/or DMSO at the concentrations indicated.
- Samples were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- CD22 status: positive were prepared, primed and analyzed by flow cytometry as described in the methods above with Bryostatin-1 (5 nM,), with Dexamethasone (200 nM) or with a combination of Bryostatin-1 (5 nM) and Dexamethasone (200 nM) to increase expression of CD22.
- FIG. 13 Exemplary and representative results of this study is shown in FIG. 13. While in the patient samples analyzed, dexamethasone showed at best a very moderate increase of CD22 antigen expression and Bryostatin-1 a moderate increase, Bryostatin-1 in combination with dexamethasone showed a substantial and synergistic expression of CD22. Increasing expression of CD22 antigen on cells of patients with lymphoid malignancies (see, FIG. 1) might be expected to further sensitize responsive patient to anti-CD22 therapies. Results described herein strongly suggest that increasing expression of CD22 antigen on cancerous cells of patients will further sensitize those cells to anti-CD22 therapies.
- Example M CD22 Priming With Bryostatin-1, Tretinoin,
- Red blood cells were removed from remnant CLL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously. Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1,
- Cells from a two CD22 patient samples (Sample ID No. 414CLL1, CD22 status: negative; Sample ID No. 423CLL1; CD22 status: negative) were prepared, primed and analyzed by flow cytometry as described in the methods above with dimethyl sulfoxide (control), with Bryostatin-1 (5 nM,), with Dexamethasone (200 nM), with Tretinoin (50 nM) or with combinations comprising Bryostatin-1 (5 nM) and Dexamethasone (200 nM) or Bryostatin-1 (5 nM) and Tretinoin (50 nM) to increase expression of CD22.
- control dimethyl sulfoxide
- Bryostatin-1 5 nM
- Dexamethasone 200 nM
- Tretinoin 50 nM
- combinations comprising Bryostatin-1 (5 nM) and Dexamethasone (200 nM) or Bryostatin-1 (5
- FIGs. 14A and 14B Exemplary and representative results of this study are shown in FIGs. 14A and 14B. While in patient sample 414CLL1, dexamethasone showed a very moderate increase of CD22 antigen expression, somewhat higher expression levels were observed upon administering tretinoin or Bryostatin-1 alone. However, both combinations, i.e., Bryostatin-1 in combination with dexamethasone and Bryostatin-1 in combination with tretinoin showed a substantial and synergistic expression of CD22 (FIG. 14A). A similar observation was made in patient sample 423CLL1 (FIG. 14B). Increasing expression of CD22 antigen on cells of patients with lymphoid malignancies (see, FIG. 1) might be expected to further sensitize responsive patient to anti-CD22 therapies. Results described herein strongly suggest that increasing expression of CD22 antigen on cancerous cells of patients will further sensitize those cells to anti-CD22 therapies.
- Red blood cells were removed from remnant a B-ALL peripheral blood specimen using RBC Lysis Buffer and cultured as described previously.
- Patient samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1, Dexamethasone, and/or DMSO at the concentrations indicated.
- Samples were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- Cells from an ALL patient (Sample ID No. 825ALL1, CD22 status: positive) were prepared, primed and analyzed by flow cytometry as described in the methods above with dimethyl sulfoxide (control), with Bryostatin-1 (5 nM,), with Dexamethasone (200 nM), with Tretinoin (50 nM) or with combinations comprising Bryostatin-1 (5 nM) and
- Dexamethasone 200 nM or Bryostatin-1 (5 nM) and Tretinoin (50 nM) to increase expression of CD22.
- FIG. 15 An exemplary and representative result of this study is shown in FIG. 15. While in this patient sample the combinations of Bryostatin-1 and dexamethasone did not result in an increase of CD22 antigen expression, but rather in a reduced expression when compared to Bryostatin-1 alone, a composition comprising Bryostatin-1 and tretinoin showed a higher expression of CD22 than what was obtained with either compound alone (FIG. 15).
- dexamethasone e.g., FIGs. 14A, B.
- FIGs. 14A, B the combination of Bryostatin-1 and Dexamethasone actually suppressed the single agent priming of CD22 by Bryostatin-1
- FIG. 15 the combination of Bryostatin-1 and Tretinoin significantly increased priming of CD22 antigen when compared to either Bryostatin-1 alone or Tretinoin alone
- Increasing expression of CD22 antigen on cells of patients with earlier B-cell malignancies see, FIG. 1 might be expected to further sensitize responsive patient to anti-CD22 therapies. Results described herein strongly suggest that increasing expression of CD22 antigen on cancerous cells of patients will further sensitize those cells to anti-CD22 therapies.
- Red blood cells were removed from remnant CLL peripheral blood specimens using RBC Lysis Buffer and cultured as described previously. Patients samples were plated in 384 well V-bottom at a density of 1 million cells/mL, treated with Bryostatin-1,
- Dexamethasone, and/or DMSO at the concentration indicated were incubated in a robotic 37°C, 95% humidity, 5% CO2 incubator for 96 hours. Following incubation, cell numbers, viability, and drug-induced changes in expression of antigen cell surface markers were determined by flow cytometry. Cells were stained with DAPI (Sigma-Aldrich, St.
- Cells from a CLL patient (Sample ID No. 803CLL1; CD20 status: CD20 dim) were prepared, primed and analyzed by flow cytometry as described in the methods above with with Bryostatin-1 (5 nM,), with Dexamethasone (200 nM), with Prednisolone (220 nM) or with combinations comprising Bryostatin-1 (5 nM) and Dexamethasone (200 nM) or Bryostatin-1 (5 nM) and Prednisolone (50) to increase expression of CD20.
- Bryostatin-1 5 nM
- Dexamethasone 200 nM
- Prednisolone 220 nM
- combinations comprising Bryostatin-1 (5 nM) and Dexamethasone (200 nM) or Bryostatin-1 (5 nM) and Prednisolone (50) to increase expression of CD20.
- FIG. 16 An exemplary and representative result of this study is shown in FIG. 16. While in this patient sample neither dexamethasone nor prednisolone resulted in an increase of CD20 antigen expression, but rather in a decrease of CD20 antigen expression, compositions comprising Bryostatin-1 and dexamethasone or Bryostatin-1 and prednisolone showed a significantly higher expression of CD20 than what was obtained with Bryostatin-1 alone (FIG. 16). As demonstrated herein, Bryostatin-1 shows increased priming of CD20 not only in combination with Dexamethasone but also with other glucocorticoids, such as prednisolone.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- Epidemiology (AREA)
- Molecular Biology (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Engineering & Computer Science (AREA)
- General Chemical & Material Sciences (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Hematology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
La présente invention concerne un agoniste pour la protéine kinase C seule, un stéroïde seul ou un acide rétinoïque seul qui n'augmente sensiblement pas l'expression d'antigènes sur des cellules cancéreuses. Cependant, l'expression de ces antigènes est sensiblement et synergiquement augmentée lors de l'administration d'une combinaison d'un agoniste de la protéine kinase C avec un stéroïde ou lors de l'administration d'une combinaison d'un agoniste de la protéine kinase C avec un acide rétinoïque. Des combinaisons et des compositions utiles particulières de l'invention comprennent un composé de bryostatine et un stéroïde ou un composé de bryostatine et un acide rétinoïque. Des combinaisons et des compositions encore plus utiles de la présente invention comprennent de la bryostatine-1 et de la dexaméthasone, de la bryostatine-1 et de la prednisolone, ou de la bryostatine-1 et de la trétinoïne. Des combinaisons et des compositions de la présente invention sont particulièrement utiles pour traiter le cancer, pour inhiber la croissance de cellules cancéreuses, pour augmenter l'expression d'antigènes sur des cellules cancéreuses et pour sensibiliser ces cellules pour des thérapies.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201962805064P | 2019-02-13 | 2019-02-13 | |
US62/805,064 | 2019-02-13 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2020168106A1 true WO2020168106A1 (fr) | 2020-08-20 |
Family
ID=69811915
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2020/018154 WO2020168106A1 (fr) | 2019-02-13 | 2020-02-13 | Combinaisons d'agonistes de protéine kinase c avec des stéroïdes ou des acides rétinoïques pour le traitement du cancer |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2020168106A1 (fr) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11976122B2 (en) | 2020-07-31 | 2024-05-07 | Adc Therapeutics Sa | Anti-IL13Rα2 antibodies |
Citations (19)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5821254A (en) * | 1995-02-17 | 1998-10-13 | The United States Of America As Represented By The Department Of Health And Human Services | Uses of 9-cis-retinoic acids and derivatives thereof alone or in combination with antineoplastic agents in the prevention or treatment of cancer |
US20050014701A1 (en) | 2001-10-26 | 2005-01-20 | Margareta Elg | Combination product comprising melagatran and dexamethasone |
US20060034915A1 (en) | 2003-02-20 | 2006-02-16 | Rice Paul W | Dexamethasone-containing formulations for oral administration as well the process for manufacturing required therefor |
US20080004694A1 (en) | 2006-05-23 | 2008-01-03 | Abbott Laboratories | Compositions and methods for administering dexamethasone which promote human coronary artery endothelial cell migration |
US20080241231A1 (en) | 2007-02-21 | 2008-10-02 | Victory Pharma, Incorporated | Transdermal delivery of dexamethasone and promethazine |
US20090270492A1 (en) | 2007-08-31 | 2009-10-29 | Wender Paul A | Bryostatin analogues, synthetic methods and uses |
US20100082095A1 (en) | 2008-09-29 | 2010-04-01 | Abbott Cardiovascular Systems Inc. | Coatings Including Dexamethasone Derivatives And Analogs And Olimus Drugs |
US20100280262A1 (en) | 2007-10-19 | 2010-11-04 | Wender Paul A | Bryostatin analogues, synthetic methods and uses |
US20110014699A1 (en) | 2009-07-20 | 2011-01-20 | Wender Paul A | Prostratin analogs, bryostatin analogs, prodrugs, synthetic methods, and methods of use |
US20110269713A1 (en) | 2008-04-16 | 2011-11-03 | University Of Utah Research Foundation | Bryostatin analogues and methods of making and using thereof |
US20130310352A1 (en) | 2010-11-12 | 2013-11-21 | Board Of Regents, The University Of Texas System | Dexamethasone combination therapy |
US20140315867A1 (en) | 2013-04-22 | 2014-10-23 | Macau University Of Science And Technology | Development of a new formulation overcoming thymus atrophy with concomitantly enhancement of anti-inflammatory effect of dexamethasone |
US20150024034A1 (en) | 2014-07-21 | 2015-01-22 | Kimia Zist Parsian Co | Method and system for synthesizing nanocarrier based long acting drug delivery system for dexamethasone |
US20150065474A1 (en) | 2008-04-18 | 2015-03-05 | Warsaw Orthopedic, Inc. | Dexamethasone formulations in a biodegradable material |
US20150342964A1 (en) | 2014-05-30 | 2015-12-03 | Warsaw Orthopedic, Inc. | Dexamethasone polymer implant for pain |
US20170239212A1 (en) | 2014-08-11 | 2017-08-24 | University Of Utah Research Foundation | Bryostatin analogs and use thereof as antiviral agents |
US20170304319A1 (en) | 2014-11-04 | 2017-10-26 | Acucort Ab | Dexamethasone oral film |
US20180221390A1 (en) | 2008-02-26 | 2018-08-09 | Emory University | Steroid Analogues for Neuroprotection |
US20180303851A1 (en) | 2013-05-24 | 2018-10-25 | Icon Bioscience, Inc. | Use of sustained release dexamethasone in post-cataract surgery inflammation |
-
2020
- 2020-02-13 WO PCT/US2020/018154 patent/WO2020168106A1/fr active Application Filing
Patent Citations (20)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5821254A (en) * | 1995-02-17 | 1998-10-13 | The United States Of America As Represented By The Department Of Health And Human Services | Uses of 9-cis-retinoic acids and derivatives thereof alone or in combination with antineoplastic agents in the prevention or treatment of cancer |
US20050014701A1 (en) | 2001-10-26 | 2005-01-20 | Margareta Elg | Combination product comprising melagatran and dexamethasone |
US20060034915A1 (en) | 2003-02-20 | 2006-02-16 | Rice Paul W | Dexamethasone-containing formulations for oral administration as well the process for manufacturing required therefor |
US20080004694A1 (en) | 2006-05-23 | 2008-01-03 | Abbott Laboratories | Compositions and methods for administering dexamethasone which promote human coronary artery endothelial cell migration |
US20080241231A1 (en) | 2007-02-21 | 2008-10-02 | Victory Pharma, Incorporated | Transdermal delivery of dexamethasone and promethazine |
US20090270492A1 (en) | 2007-08-31 | 2009-10-29 | Wender Paul A | Bryostatin analogues, synthetic methods and uses |
US20130123518A1 (en) | 2007-10-19 | 2013-05-16 | The Board Of Trustees Of The Leland Stanford Junior University | Bryostatin Analogues, Synthetic Methods and Uses |
US20100280262A1 (en) | 2007-10-19 | 2010-11-04 | Wender Paul A | Bryostatin analogues, synthetic methods and uses |
US20180221390A1 (en) | 2008-02-26 | 2018-08-09 | Emory University | Steroid Analogues for Neuroprotection |
US20110269713A1 (en) | 2008-04-16 | 2011-11-03 | University Of Utah Research Foundation | Bryostatin analogues and methods of making and using thereof |
US20150065474A1 (en) | 2008-04-18 | 2015-03-05 | Warsaw Orthopedic, Inc. | Dexamethasone formulations in a biodegradable material |
US20100082095A1 (en) | 2008-09-29 | 2010-04-01 | Abbott Cardiovascular Systems Inc. | Coatings Including Dexamethasone Derivatives And Analogs And Olimus Drugs |
US20110014699A1 (en) | 2009-07-20 | 2011-01-20 | Wender Paul A | Prostratin analogs, bryostatin analogs, prodrugs, synthetic methods, and methods of use |
US20130310352A1 (en) | 2010-11-12 | 2013-11-21 | Board Of Regents, The University Of Texas System | Dexamethasone combination therapy |
US20140315867A1 (en) | 2013-04-22 | 2014-10-23 | Macau University Of Science And Technology | Development of a new formulation overcoming thymus atrophy with concomitantly enhancement of anti-inflammatory effect of dexamethasone |
US20180303851A1 (en) | 2013-05-24 | 2018-10-25 | Icon Bioscience, Inc. | Use of sustained release dexamethasone in post-cataract surgery inflammation |
US20150342964A1 (en) | 2014-05-30 | 2015-12-03 | Warsaw Orthopedic, Inc. | Dexamethasone polymer implant for pain |
US20150024034A1 (en) | 2014-07-21 | 2015-01-22 | Kimia Zist Parsian Co | Method and system for synthesizing nanocarrier based long acting drug delivery system for dexamethasone |
US20170239212A1 (en) | 2014-08-11 | 2017-08-24 | University Of Utah Research Foundation | Bryostatin analogs and use thereof as antiviral agents |
US20170304319A1 (en) | 2014-11-04 | 2017-10-26 | Acucort Ab | Dexamethasone oral film |
Non-Patent Citations (64)
Title |
---|
ALI M ARDEKANI ET AL: "Bryostatin-1, Fenretinide and 1[alpha],25 (OH) 2 D 3 Induce Growth Inhibition, Apoptosis and Differentiation in T and B Cell-Derived Acute Lymphoblastic Leukemia Cell Lines (CCRF-CEM and Nalm-6)", AVICENNA JOURNAL OF MEDICAL BIOTECHNOLOGY, vol. 3, 1 January 2012 (2012-01-01), XP055697991 * |
AL-ZOOBI ET AL., INT IMMUNOL, vol. 26, no. 8, 2014, pages 451 - 65 |
ANDO ET AL., EXP EYE RES, vol. 80, no. 4, 2005, pages 501 - 7 |
BERGE ET AL.: "Pharmaceutical Salts", JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 1 - 19, XP002675560, DOI: 10.1002/jps.2600660104 |
BESSHO ET AL., ANTICANCER RES, vol. 19, no. 1B, 1999, pages 693 - 8 |
BIBERACHER ET AL., HAEMATOLOGICA, vol. 97, no. 5, 2012, pages 771 - 9 |
BLANC ET AL., CLIN CANCER RES, vol. 17, no. 20, 2011, pages 6448 - 58 |
BOLAND ET AL., JBIOL CHEM, vol. 275, no. 33, 2000, pages 25231 - 8 |
CHING ET AL., BIOCHEM PHARMACOL, vol. 58, no. 7, 1999, pages 1173 - 81 |
CHO ET AL., CAN J PHYSIOL PHARMACAL, vol. 82, no. 6, 2004, pages 380 - 6 |
CHO ET AL., FEBS LETT, vol. 579, no. 20, 2005, pages 4213 - 8 |
CHOUTALALAY, ADV ENZYME REGUL, vol. 22, 1984, pages 27 - 55 |
CHOUTALALAY: "New Avenues in Developmental Cancer Chemotherapy", 1987, ACADEMIC PRESS |
DAS ET AL., J BIOL CHEM, vol. 272, no. 23, 1997, pages 14914 - 20 |
ELISEEV ET AL., JBIOL CHEM, vol. 279, no. 25, 2004, pages 26019 - 27 |
FERLINE ET AL., BR J CANCER, vol. 79, no. 2, 1999, pages 257 - 63 |
GRDINA ET AL., MIL MED, vol. 167, no. 2, 2002, pages 51 - 3 |
GUN-YOUNG HONG ET AL: "Combination of paclitaxel- and retinoic acid-incorporated nanoparticles for the treatment of CT-26 colon carcinoma", ARCHIVES OF PHARMACAL RESEARCH, PHARMACEUTICAL SOCIETY OF KOREA, HEIDELBERG, vol. 34, no. 3, 6 May 2011 (2011-05-06), pages 407 - 417, XP019899893, ISSN: 1976-3786, DOI: 10.1007/S12272-011-0308-8 * |
HALEMARHAM, THE HARPER COLLINS DICTIONARY OF BIOLOGY, 1991 |
HARLOWLANE: "The Cambridge Dictionary of Science and Technology", 1988, COLD SPRING HARBOR PUBLICATIONS |
HIRAGA ET AL., BLOOD, vol. 113, no. 20, 2009, pages 4885 - 93 |
HWANG ET AL., CANCER BIOCHEM BIOPHYS, vol. 14, no. 4, 1995, pages 265 - 72 |
ISE ET AL., CLIN LYMPHOMA MYELOMA LEUK, vol. 16, no. 5, 2016, pages e59 - 64 |
ISHII ET AL., TOXICOL APPL PHARMICOL, vol. 184, no. 2, 2002, pages 88 - 97 |
J. MARCH: "Advanced Organic Chemistry", 1992, JOHN WILEY AND SONS |
KIM ET AL., J CANCER RES CLIN ONCOL, vol. 130, no. 9, 2004, pages 551 - 60 |
KISHIDA ET AL., CANCER CHEMOTHER PHARMACOL, vol. 55, no. 4, 2005, pages 393 - 403 |
KOO ET AL., IMMUNOPHARMACOL IMMUNOTOXICOL, vol. 23, no. 2, 2001, pages 175 - 86 |
LEV ET AL., MOL CANCER THER, vol. 2, no. 8, 2003, pages 753 - 63 |
LI ET AL., CARCINOGENESIS, vol. 17, no. 11, 1996, pages 2305 - 9 |
LI ET AL., ONCO TARGETS THER, vol. 10, 2017, pages 3645 - 3665 |
LOPANIK ET AL., OECOLOGIA, vol. 139, 2004, pages 131 - 139 |
MESSER ET AL., CYTOKINE, vol. 2, no. 6, 1990, pages 389 - 97 |
MINNEMAN ET AL., J NEUROCHEM, vol. 74, no. 6, 2000, pages 2392 - 400 |
MOHAMMAD R M ET AL: "The addition of bryostatin 1 to cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) chemotherapy improves response in a CHOP-resistant human diffuse large cell lymphoma xenograft model", CLINICAL CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 6, no. 12, 1 December 2000 (2000-12-01), pages 4950 - 4956, XP002516178, ISSN: 1078-0432 * |
NEMETH ET AL., JBIOL CHEM, vol. 277, no. 10, 2002, pages 7713 - 9 |
NIE ET AL., MOL PHARMACOL, vol. 53, no. 4, 1998, pages 663 - 9 |
NIJHOF ET AL., LEUKEMIA, vol. 29, no. 10, 2015, pages 2039 - 49 |
OGAWA ET AL., J INVEST DERMATOL, vol. 126, no. 2, 2006, pages 349 - 53 |
OSBORN ET AL., PROC NATL ACAD SCI USA, vol. 86, no. 7, 1989, pages 2336 - 40 |
PIRET ET AL., NUCLEIC ACIDS RES, vol. 24, no. 20, 1996, pages 4242 - 8 |
POST ET AL., JNEUROSCI, vol. 18, no. 20, 1998, pages 8236 - 46 |
ROBBIE G. MAJZNER ET AL: "Tumor Antigen Escape from CAR T-cell Therapy", CANCER DISCOVERY, vol. 8, no. 10, 1 October 2018 (2018-10-01), US, pages 1219 - 1226, XP055664575, ISSN: 2159-8274, DOI: 10.1158/2159-8290.CD-18-0442 * |
ROSETTE ET AL., J CELL BIOL, vol. 128, no. 6, 1995, pages 1111 - 9 |
SCHON ET AL., EXPERT OPIN THER TARGETS, vol. 10, no. 1, 2006, pages 69 - 76 |
SEN ET AL., CELL, vol. 47, no. 6, 1986, pages 921 - 8 |
SHRECK, EMBO J, vol. 10, no. 8, 1991, pages 2247 - 58 |
SINGLETON ET AL.: "Culture of Animal Cells a Manual of Basic Technique", 1994, pages: 231 - 241 |
SMALL ET AL., PEER J, vol. 1, 2013, pages e31 |
STEIN ET AL., MOL CELL BIOL, vol. 9, no. 11, 1989, pages 5169 - 81 |
STELLA ET AL.: "Directed Drug Delivery", 1985, HUMANA PRESS, article "Prodrugs: A Chemical Approach to Targeted Drug Delivery", pages: 247 - 267 |
TESHIMA ET AL., NITRIC OXIDE, vol. 11, no. 1, 2004, pages 9 - 16 |
TIAN ET AL., MOL PHARMACOL, vol. 68, no. 6, 2005, pages 1534 - 42 |
TING-CHAO CHOU: "Pharmacol. Rev.", vol. 58, 2006, article "Theoretical Basis, Experimental Design, and Computerized Simulation of Synergism and Antagonism in Drug Combination Studies", pages: 621 - 681 |
TONG-STARKESEN ET AL., J IMMUNOL, vol. 142, no. 2, 1989, pages 702 - 7 |
WANG ET AL., SCIENCE, vol. 274, no. 5288, 1996, pages 784 - 7 |
WENDER ET AL., ISR J CHEM, vol. 51, no. 3-4, 2011, pages 453 - 472 |
WIHNAN: "Prodrugs in Cancer Chemotherapy", BIOCHEMICAL SOCIETY TRANSACTIONS, vol. 14, 1986, pages 375 - 382 |
WOJCIECHOESKI ET AL., J IMMUNOL, vol. 174, 2005, pages 7859 - 7868 |
WOJCIECHOWSKI W ET AL: "Enhanced expression of CD20 in human tumor B cells is controlled through ERK-dependent mechanisms", THE JOURNAL OF IMMUNOLOGY, AMERICAN ASSOCIATION OF IMMUNOLOGISTS, US, vol. 174, no. 12, 15 June 2005 (2005-06-15), pages 7859 - 7868, XP002707715, ISSN: 0022-1767, DOI: 10.4049/JIMMUNOL.174.12.7859 * |
YIN ET AL., J NEUROIMMUNOL, 7 March 2006 (2006-03-07) |
ZHANG, MABS, vol. 1, no. 4, 2009, pages 326 - 331 |
ZHOU-STACHE ET AL., MED BIOL ENG COMPUT, vol. 40, no. 6, 2002, pages 698 - 703 |
ZOU ET AL., J CELL PHYSIOL, vol. 205, no. 3, 2005, pages 437 - 43 |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11976122B2 (en) | 2020-07-31 | 2024-05-07 | Adc Therapeutics Sa | Anti-IL13Rα2 antibodies |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP6833885B2 (ja) | がんを処置するためのクローディン18.2に対する抗体を伴う併用療法 | |
TWI797091B (zh) | 利用嵌合抗原受體之癌症治療 | |
EP3209769B1 (fr) | Compositions et procédés pour induire la phagocytose de cellules positives de classe i du cmh et pour contrer la résistance aux agents anti-cd47/sirpa | |
KR101912957B1 (ko) | 다발성 골수종 및 nhl 치료를 위한 항-cd38 항체 및 레날리도마이드 또는 보르테조밉 | |
JP6840127B2 (ja) | がんの治療における抗pd−1抗体および抗m−csf抗体の併用 | |
CN111465612A (zh) | B细胞成熟抗原结合蛋白 | |
WO2017149515A1 (fr) | Cellules exprimant de multiples molécules de récepteur d'antigène chimère (car) et leurs utilisations | |
EP2694553A2 (fr) | Anticorps contre des peptides cytosoliques | |
WO2014127785A1 (fr) | Polythérapie impliquant des anticorps dirigés contre la claudine 18,2 pour le traitement du cancer | |
JP2020502259A (ja) | アルギナーゼ阻害剤併用療法 | |
US20220324965A1 (en) | Combination therapy involving antibodies against claudin 18.2 and immune checkpoint inhibitors for treatment of cancer | |
US10179816B2 (en) | Pharmaceutical compositions comprising an antibody which binds the human anti-Müllerian hormone receptor type II | |
JP2022553410A (ja) | 固形がんおよび血液がんを処置するための併用療法 | |
WO2020011972A9 (fr) | Fragments de liaison fc comprenant un côté de liaison à l'antigène cd137 | |
WO2020168106A1 (fr) | Combinaisons d'agonistes de protéine kinase c avec des stéroïdes ou des acides rétinoïques pour le traitement du cancer | |
JP2020528061A (ja) | BET阻害剤、Bcl−2阻害剤及び抗CD20抗体を用いた併用療法 | |
EP2958945A1 (fr) | Polythérapie impliquant des anticorps dirigés contre la claudine 18.2 pour le traitement du cancer | |
WO2023155852A1 (fr) | Cellules effectrices immunitaires modifiées et leurs utilisations | |
EP3153178A1 (fr) | Procédé de traitement de patients atteints de cancer au moyen d'un médicament ciblé vers folr1 et d'antifolate, et médicament | |
US10478497B2 (en) | Combinations of inecalcitol with an anti-CD38 agent and their uses for treating cancer | |
WO2014143835A1 (fr) | Thérapie de combinaison/adjuvant pour une maladie positive pour wt-1 | |
AU2016203429B2 (en) | Binding Molecules to the Human OX40 Receptor | |
US20200046737A1 (en) | Methods for treating cancer, and compositions therefor | |
WO2023073645A1 (fr) | Thérapie avec un anticorps anti-cd19 et un inhibiteur enzymatique activateur de sumo | |
RU2789478C2 (ru) | Комбинированная терапия с использованием антител к клаудину 18.2 для лечения рака |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 20711412 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 20711412 Country of ref document: EP Kind code of ref document: A1 |