WO2007101235A2 - Improved antitumoral treatments - Google Patents

Improved antitumoral treatments Download PDF

Info

Publication number
WO2007101235A2
WO2007101235A2 PCT/US2007/062936 US2007062936W WO2007101235A2 WO 2007101235 A2 WO2007101235 A2 WO 2007101235A2 US 2007062936 W US2007062936 W US 2007062936W WO 2007101235 A2 WO2007101235 A2 WO 2007101235A2
Authority
WO
WIPO (PCT)
Prior art keywords
aplidine
cancer
treatment
drug
combination
Prior art date
Application number
PCT/US2007/062936
Other languages
French (fr)
Other versions
WO2007101235A3 (en
Inventor
Glynn Thomas Faircloth
Pablo Manuel Aviles Marin
Doreen Lepage
Jesus San Miguel Izquierdo
Atanasio Pandiella
Original Assignee
Pharma Mar, S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to RU2008138560/15A priority Critical patent/RU2481853C2/en
Priority to MX2008010999A priority patent/MX2008010999A/en
Priority to DK07757603.1T priority patent/DK2029155T3/en
Priority to CN2007800068661A priority patent/CN101389347B/en
Priority to AU2007220050A priority patent/AU2007220050B2/en
Priority to ES07757603.1T priority patent/ES2575518T3/en
Priority to KR1020087023662A priority patent/KR101512503B1/en
Priority to NZ571043A priority patent/NZ571043A/en
Priority to KR1020147021341A priority patent/KR20140101014A/en
Priority to MEP-2016-115A priority patent/ME02450B/en
Priority to EP07757603.1A priority patent/EP2029155B1/en
Priority to SI200731777A priority patent/SI2029155T1/en
Priority to JP2008557477A priority patent/JP5393159B2/en
Priority to CA2643238A priority patent/CA2643238C/en
Application filed by Pharma Mar, S.A. filed Critical Pharma Mar, S.A.
Priority to US12/278,559 priority patent/US8258098B2/en
Priority to RS20160545A priority patent/RS54928B1/en
Publication of WO2007101235A2 publication Critical patent/WO2007101235A2/en
Publication of WO2007101235A3 publication Critical patent/WO2007101235A3/en
Priority to IL193094A priority patent/IL193094A/en
Priority to NO20084047A priority patent/NO342012B1/en
Priority to HK09106467.2A priority patent/HK1127294A1/en
Priority to HRP20160646TT priority patent/HRP20160646T1/en
Priority to CY20161100620T priority patent/CY1117722T1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/15Depsipeptides; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Abstract

The present invention relates to combinations of aplidine or aplidine analogues with other antitumoral agents, and the use of these combinations in the treatment of cancer, in particular in the treatment of lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia and lymphoma.

Description

IMPROVED ANTITUMORAL TREATMENTS
FIELD OF THE INVENTION
The present invention relates to combinations of Aplidine or aplidine analogues with other antitumoral agents, and the use of these combinations in the treatment of cancer, in particular in the treatment of lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia and lymphoma.
BACKGROUND OF THE INVENTION
Aplidine (Dehydrodidemnin B) is a cyclic depsipeptide that was isolated from a Mediterranean marine tunicate, Aplidium albicans, and it is the subject of WO 9104985. It is related to compounds known as didemnins, and has the following structure:
Figure imgf000002_0001
More information on Aplldine, aplidine analogues, its uses, formulations and synthesis can be found in patent applications WO 99 42125, WO Ol 35974, WO Ol 76616, WO 02 30441, WO 02 02596, WO 03 33013 and WO 2004 080477. We incorporate by specific reference the content of each of these PCT texts.
In both animal preclinical studies and human clinical Phase I studies Aplidine has been shown to have cytotoxic potential against a broad spectrum of tumor types including leukemia and lymphoma. See for example:
Faircloth, G. et al.: "Dehydrodidemnin B (DDB) a new marine derived anticancer agent with activity against experimental tumour models", 9th NCI-EORTC Symp. New Drugs Cancer Ther. (March 12-15, Amsterdam) 1996, Abst 111;
Faircloth, G. et al.: "Preclinical characterization of aplidine, a new marine anticancer depsipeptide", Proc. Amer. Assoc. Cancer Res. 1997, 38: Abst 692;
Depenbrock H, Peter R, Faircloth GT, Manzanares I, Jimeno J, Hanauske AR.: "In vitro activity of Aplidine, a new marine- derived anticancer compound, on freshly explanted clonogenic human tumour cells and haematopoietic precursor cells" Br. J. Cancer, 1998; 78: 739-744; Faircloth G, Grant W, Nam S, Jimeno J, Manzanares I, Rinehart K.: "Schedule-dependency of Aplidine, a marine depsipeptide with antitumor activity", Proc. Am. Assoc. Cancer Res. 1999; 40: 394; Broggini M, Marchini S, D'Incalci M, Taraboletti G, Giavazzi R, Faircloth G, Jimeno J.: "Aplidine blocks VEGF secretion and VEGF/VEGF-R1 autocrine loop in a human leukemic cell line", Clin. Cancer Res. 2000; 6 (suppl): 4509;
Erba E, Bassano L, Di Liberti G, Muradore I, Chiorino G, Ubezio P, Vignati S, Codegoni A, Desiderio MA, Faircloth G, Jimeno J and D'Incalci M.: "Cell cycle phase perturbations and apoptosis in tumour cells induced by aplidine", Br. J. Cancer 2002; 86: 1510-1517; Paz-Ares L, Anthony A, Pronk L, Twelves C, Alonso S, Cortes-Funes H, Celli N, Gomez C, Lopez-Lazaro L, Guzman C, Jimeno J, Kaye S.: "Phase I clinical and pharmacokinetic study of aplidine, a new marine didemnin, administered as 24-hour infusion weekly" Clin. Cancer Res. 2000; 6 (suppl): 4509;
Raymond E, Ady-Vago N, Baudin E, Ribrag V, Faivre S, Lecot F, Wright T, Lopez Lazaro L, Guzman C, Jimeno J, Ducreux M, Le Chevalier T, Armand JP. : "A phase I and pharmacokinetic study of aplidine given as a 24-hour continuous infusion every other week in patients with solid tumor and lymphoma", Clin. Cancer Res. 2000; 6 (suppl): 4510; Maroun J, Belanger K, Seymour L, Soulieres D, Charpentier D, Goel R, Stewart D, Tomiak E, Jimeno J, Matthews S. :"Phase I study of aplidine in a 5 day bolus q 3 weeks in patients with solid tumors and lymphomas", CUn. Cancer Res. 2000; 6 (suppl): 4509; Izquierdo MA, Bowman A, Martinez M, Cicchella B, Jimeno J, Guzman C, Germa J, Smyth J.: "Phase I trial of Aplidine given as a 1 hour intravenous weekly infusion in patients with advanced solid tumors and lymphoma", Clin. Cancer Res. 2000; 6 (suppl): 4509.
Mechanistic studies indicate that Aplidine can block VEGF secretion in ALL-MOLT4 cells and in vitro cytotoxic activity at low concentrations (5nM) has been observed in AML and ALL samples from pediatric patients with de novo or relapsed ALL and AML. Aplidine appears to induce both a Gl and a G2 arrest in drug treated leukemia cells in vitro. Apart from down regulation of the VEGF receptor, little else is known about the mode(s) of action of Aplidine.
In phase I clinical studies with Aplidine, L-carnitine was given as a 24 hour pretreatment or co-administered to prevent myelotoxicity, see for example WO 02 30441. Co -administration of L-carnitine was proven to be able to improve the recovery of the drug induced muscular toxicity and has allowed for dose escalation of Aplidine.
Previously, in vitro and in vivo assays conducted with Aplidine in combination with other anticancer agents shown that the assayed drug combinations were useful in combination therapy for the treatment of leukemia and lymphoma. In WO 2004 080421 , Aplidine was specifically evaluated in combination with methotrexate, cytosine arabinoside, mitoxantrone, vinblastine, methylprednisolone and doxorubicin for the treatment of leukemia and lymphoma.
Since cancer is a leading cause of death in animals and humans, several efforts have been and are still being undertaken in order to obtain an antitumor therapy active and safe to be administered to patients suffering from a cancer. The problem to be solved by the present invention is to provide antitumor therapies that are useful in the treatment of cancer.
SUMMARY OF THE INVENTION
We have established that Aplidine and aplidine analogues potentiates other anticancer agents and therefore can be successfully used in combination therapy for the treatment of cancer. This invention is directed to pharmaceutical compositions, pharmaceutical dosage forms, kits, methods for the treatment of cancer using these combination therapies and uses of Aplidine and aplidine analogues in the manufacture of a medicament for combination therapy.
In accordance with one aspect of this invention, we provide effective combination therapies based on Aplidine and aplidine analogues, using other drugs which are effective in the treatment of cancer. Preferably the other drug or other drugs are effective in the treatment of a cancer selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia and lymphoma. Most preferably the other drug or other drugs are selected from the group consisting of paclitaxel (TaxoF), doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil (5-FU), cytosine arabinoside (AraC), carboplatin, 7-ethyl-lO-hydroxycamptothecin (SN38), etoposide (VP 16), melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide (Revlimid®), interleukin-2 (IL-2), interferon-α 2 (INF-α), dacarbazine (DTIC), bevacizumab (Avastin®), idarubicin, thalidomide, and rituximab.
In another embodiment the invention encompasses a method of treating cancer comprising administering to a patient in need of such treatment a therapeutically effective amount of Aplidine or an aplidine analogue, or a pharmaceutically acceptable prodrug, salt, solvate or hydrate thereof, and a therapeutically effective amount of another drug which is effective in the treatment of cancer or a pharmaceutically acceptable prodrug, salt, solvate or hydrate thereof, administered prior, during, or after administering Aplidine or aplidine analogue. In an additional embodiment of the invention, a therapeutically effective amount of third drug is administered, and is administered prior, during, or after administering Aplidine or aplidine analogue and the second drug.
Preferably the other drug or other drugs are effective in the treatment of a cancer selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia and lymphoma. Most preferably the other drug or other drugs are selected from the group consisting of paclitaxel (Taxol®), doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil (5-FU), cytosine arabinoside (AraC), carboplatin, 7-ethyl-lO-hydroxycamptothecin (SN38), etoposide (VP16), melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide (Revlimid®), interleukin-2 (IL- 2), interferon-α 2 (INF-α), dacarbazine (DTIC), bevacizumab (Avastin®), idarubicin, thalidomide, and rituximab. The other drug or other drugs may form part of the same composition, or be provided as a separate composition for administration at the same time or at a different time.
In another aspect the invention encompasses a method of increasing the therapeutic efficacy of a drug effective in the treatment of cancer, preferably a drug effective in the treatment of a cancer selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia and lymphoma, most preferably a drug selected from the group consisting of paclitaxel (Taxol®), doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil (5-FU), cytosine arabinoside (AraC), carboplatin, 7-ethyl-10- hydroxycamptothecin (SN38), etoposide (VP 16), melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide (Revlimid®), interleukin-2 (IL-2), interferon-α 2 (INF-α), dacarbazine (DTIC), bevacizumab (Avastin®), idarubicin, thalidomide, and rituximab, or a pharmaceutically acceptable prodrug, salt, solvate or hydrate thereof, which comprises administering to a patient in need thereof an amount of Aplidine or an aplidine analogue, or a pharmaceutically acceptable prodrug, salt, solvate or hydrate thereof. Aplidine or the aplidine analogue is administered prior, during, or after administering the other drug. In an additional embodiment of the invention, a therapeutically effective amount of third drug is administered, and is administered prior, during, or after administering Aplidine or aplidine analogue and the second drug. Preferably the third drug is a drug effective in the treatment of a cancer selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia and lymphoma. Most preferably the third drug is selected from the group consisting of paclitaxel (Taxol®), doxorubicin, cisplatin, arsenic trioxide, 5- fluorouracil (5-FU), cytosine arabinoside (AraC), carboplatin, 7-ethyl-lO- hydroxycamptothecin (SN38), etoposide (VP 16), melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide (Revlimid®), interleukin-2 (IL-2), interferon-α 2 (INF-α), dacarbazine (DTIC), bevacizumab (Avastin®), idarubicin, thalidomide, and rituximab, or a pharmaceutically acceptable prodrug, salt, solvate or hydrate thereof.
In a further aspect the invention encompasses a pharmaceutical composition comprising Aplidine or an aplidine analogue, or a pharmaceutically acceptable prodrug, salt, solvate or hydrate thereof, and another drug effective in the treatment of cancer. In an additional embodiment of the invention, the pharmaceutical composition further comprises a third drug also effective in the treatment of cancer. Preferably the other drug or other drugs are effective in the treatment of a cancer selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia and lymphoma. Most preferably the other drug or other drugs are selected from the group consisting of paclitaxel (Taxol®), doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil (5-FU), cytosine arabinoside (AraC), carboplatin, 7-ethyl-lO-hydroxycamptothecin (SN38), etoposide (VP 16), melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide (Revlimid®), interleukin-2 (IL-2), interferon-α 2 (INF-α), dacarbazine (DTIC), bevacizumab (Avastin®), idarubicin, thalidomide, and rituximab. The invention also encompasses a kit for use in the treatment or prevention of cancer which comprises a dosage form of Aplidine or an aplidine analogue, or a pharmaceutically acceptable prodrug, salt, solvate or hydrate thereof, a dosage form of another drug effective in the treatment of cancer, or a pharmaceutically acceptable prodrug, salt, solvate or hydrate thereof, and instructions for the use of each actor in combination for the treatment or prevention of cancer. In an additional embodiment of the invention, the kit further comprises a dosage form of a third drug also effective in the treatment of cancer, or a pharmaceutically acceptable prodrug, salt, solvate or hydrate thereof. Preferably the other drug or other drugs are effective in the treatment of a cancer selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia and lymphoma. Most preferably the other drug or other drugs are selected from the group consisting of paclitaxel (Taxol®), doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil (5-FU), cytosine arabinoside (AraC), carboplatin, 7-ethyl-lO-hydroxycamptothecin (SN38), etoposide (VP16), melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide (Revlimid®), interleukin-2 (IL-2), interferon-α 2 (INF-α), dacarbazine (DTIC), bevacizumab (Avastin®), idarubicin, thalidomide, and rituximab.
Effective combination therapies based on the used of three drugs, Aplidine and aplidine analogues, plus two additional drugs (a second drug and a third drug) are also encompassed by the present invention.
In one preferred aspect, the present invention is concerned with synergistic combinations.
BRIEF DESCRIPTION OF THE FIGURES Fig 1. An example of linear regression analysis which is a method for revealing the presence of synergy.
Fig 2. In vitro activity data of Aplidine (Aplidin®) in combination with paclitaxel (Taxol®) against SKBR3 cells.
Fig 3. In vitro activity data of Aplidine (Aplidin®) in combination with paclitaxel (Taxol®) against MOLT3 cells.
Fig 4. In vitro activity data of Aplidine (Aplidin®) in combination with paclitaxel (Taxol®) against PC3 cells.
Fig 5. In vitro activity data of Aplidine (Aplidin®) in combination with paclitaxel (Taxol®) against HL60 cells.
Fig 6. In vitro activity data of Aplidine (Aplidin®) in combination with paclitaxel (Taxol®) against MXl cells.
Fig 7. In vitro activity data of Aplidine (Aplidin®) in combination with paclitaxel (Taxol®) against A549 cells.
Fig 8. In vitro activity data of Aplidine (Aplidin®) in combination with doxorubicin (DOX) against A549 cells.
Fig 9. In vitro activity data of Aplidine (Aplidin®) in combination with doxorubicin (DOX) against HT29 cells.
Fig 10. In vitro activity data of Aplidine (Aplidin®) in combination with doxorubicin (DOX) against PC3 cells.
Fig 11. In vitro activity data of Aplidine (Aplidin®) in combination with doxorubicin (DOX) against MOLT3 cells.
Fig 12. In vitro activity data of Aplidine (Aplidin®) in combination with doxorubicin (DOX) against MXl cells.
Fig 13. In vitro activity data of Aplidine (Aplidin®) in combination with doxorubicin (DOX) against SKBR3 cells.
Fig 14. In vitro activity data of Aplidine (AP, Aplidin®) in combination with cisplatin (DDP) against MXl cells.
Fig 15. In vitro activity data of Aplidine (Aplidin®) in combination with cisplatin (cis-DDP) against HT29 cells. Fig 16. In vitro activity data of Aplidine (APL, Aplidin®) in combination with cisplatin (cisDDP, DDP) against SKBR3 cells.
Fig 17. In vitro activity data of Aplidine (Aplidin®) in combination with cisplatin (cis-DDP, DDP) against MOLT3 cells.
Fig 18. In vitro activity data of Aplidine (Aplidin®) in combination with cisplatin (cis-DDP, DDP) against A549 cells.
Fig 19. In vitro activity data of Aplidine (Aplidin®) in combination with arsenic trioxide (TRI) against A549 cells.
Fig 20. In vitro activity data of Aplidine (Aplidin®) in combination with arsenic trioxide (TRI) against HT29 cells.
Fig 21. In vitro activity data of Aplidine (Aplidin®) in combination with arsenic trioxide (TRI) against PC3 cells.
Fig 22. In vitro activity data of Aplidine (Aplidin®) in combination with arsenic trioxide (TRI) against MOLT3 cells.
Fig 23. In vitro activity data of Aplidine (Aplidin®) in combination with
5-fhiorouracil (5FU) against HL60 cells.
Fig 24. In vitro activity data of Aplidine (Aplidin®) in combination with
5-fluorouracil (5FU) against SKBR3 cells.
Fig 25. In vitro activity data of Aplidine (Aplidin®) in combination with
5-fluorouracil (5FU) against A549 cells.
Fig 26. In vitro activity data of Aplidine (Aplidin®) in combination with
5-fluorouracil (5FU) against PC3 cells.
Fig 27. In vitro activity data of Aplidine (Aplidin®) in combination with
5-fluorouracil (5FU) against HT29 cells.
Fig 28. In vitro activity data of Aplidine (Aplidin®) in combination with cytosine arabinoside (AraC) against SKBR3 cells.
Fig 29. In vitro activity data of Aplidine (Aplidin®) in combination with cytosine arabinoside (AraC) against A549 cells.
Fig 30. In vitro activity data of Aplidine (Aplidin®) in combination with cytosine arabinoside (AraC) against PC3 cells. Fig 31. In vitro activity data of Aplidine (Aplidin®) in combination with cytosine arabinoside (AraC) against HL60 cells.
Fig 32. In vitro activity data of Aplidine (Aplidin®) in combination with cytosine arabinoside (AraC) against HT29 cells.
Fig 33. In vitro activity data of Aplidine (Aplidin®) in combination with carboplatin against PC3 cells.
Fig 34. In vitro activity data of Aplidine (Aplidin®) in combination with carboplatin against HT29 cells.
Fig 35. In vitro activity data of Aplidine (Aplidin®) in combination with carboplatin against A549 cells.
Fig 36. In vitro activity data of Aplidine (Aplidin®) in combination with carboplatin against MOLT3 cells.
Fig 37. In vitro activity data of Aplidine (Aplidin®) in combination with carboplatin against MXl cells.
Fig 38. In vitro activity data of Aplidine (Aplidin®) in combination with
SN-38 against A549 cells.
Fig 39. In vitro activity data of Aplidine (Aplidin®) in combination with
SN-38 against SKBR3 cells.
Fig 40. In vitro activity data of Aplidine (Aplidin®) in combination with
SN-38 against HL60 cells.
Fig 41. In vitro activity data of Aplidine (Aplidin®) in combination with
SN-38 against PC3 cells.
Fig. 42A and 42B. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP 16 (B) against HL-60 cells.
Fig. 43A and 43B. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP 16 (B) against K562 cells.
Fig. 44A and 44B. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP 16 (B) against MOLT-3 cells.
Fig. 45A and 45B. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP 16 (B) against MCl 16 cells. Fig. 46A and 46B. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP 16 (B) against RAMOS cells.
Fig. 47A and 47B. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP 16 (B) against U937 cells.
Fig. 48A and 48B. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP 16 (B) against NCI-H929 cells.
Fig. 49A and 49B. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP 16 (B) against HUNS-I cells.
Fig. 5OA and 5OB. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP16 (B) against U266 Bl cells.
Fig. 5 IA and 5 IB. In vitro activity data of Aplidine (A, Aplidin®) in combination with VP 16 (B) against RPMI 8226 cells.
Fig. 52. In vitro activity data of Aplidine (Aplidin®) in combination with carboplatin against LOX-I-MVI cells.
Fig. 53. In vitro activity data of Aplidine (Aplidin®) in combination with carboplatin against UACC-257 cells.
Fig. 54. Dose response to Aplidine after (48 h treatment) in MMlS,
MMlR, U266 and U266-LR7 cell lines.
Fig. 55. Comparison of dose efficacy of Aplidine (Aplidin®) and other drugs on the MMlS cell line (48 h of treatment).
Fig. 56. Combination Aplidine (Aplidin®) and Dexamethasone at 3 days.
A) Dose effect curve. B) Fa-CI plot.
Fig. 57. Combination Aplidine (Aplidin®) and Dexamethasone at 6 days.
A) Dose effect curve. B) Fa-CI plot.
Fig. 58. Combination Aplidine (Aplidin®) and Melphalan at 3 days. A)
Dose effect curve. B) Fa-CI plot.
Fig. 59. Combination Aplidine (Aplidin®) and Melphalan at 6 days. A)
Dose effect curve. B) Fa-CI plot.
Fig. 60. Combination Aplidine (Aplidin®) and Doxorubicin at 3 days. A)
Dose effect curve. B) Fa-CI plot. Fig. 61. Combination Aplidine (Aplidin®) and Doxorubicin at 6 days. A)
Dose effect curve. B) Fa-CI plot.
Fig. 62. Combination Aplidine (Aplidin®) and Lenalidomide (Revlimid®) at 3 days. A) Dose effect curve. B) Fa-CI plot.
Fig. 63. Combination Aplidine (Aplidin®) and Lenalidomide (Revlimid®) at 6 days. A) Dose effect curve. B) Fa-CI plot.
Fig. 64. Combination Aplidine (Aplidin®) and Bortezomib at 3 days. A)
Dose effect curve. B) Fa-CI plot.
Fig. 65. Combination of Aplidine (Aplidin®) and Bortezomib at 6 days.
A) Dose effect curve. B) Fa-CI plot.
Fig. 66. Kinetics of net tumor volume after initiation of treatment with
Aplidine (Aplidin®) as single agent or in combination with Dacarbazine in MRI-H- 187 melanoma tumor xenografts.
Fig. 67. Kinetics of net tumor volume after initiation of treatment with
Aplidine (Aplidin®) as single agent or in combination with Carboplatin in
MRI-H- 187 melanoma tumor xenografts.
Fig. 68. Kinetics of net tumor volume after initiation of treatment with
Aplidine (Aplidin®) as single agent or in combination with Interleukin-2
(IL- 2) in MRI-H- 187 melanoma tumor xenografts.
Fig. 69. Kinetics of net tumor volume after initiation of treatment with
Aplidine (Aplidin®) as single agent or in combination with Interferon-α
2a (INF-α) in MRI-H- 187 melanoma tumor xenografts.
Fig. 70. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL) as single agent or in combination with Dacarbazine
(DTIC) in LOX-IMVI melanoma tumor xenografts.
Fig. 71. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL) as single agent or in combination with Carboplatin in
LOX-IMVI melanoma tumor xenografts.
Fig. 72. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL, Aplidin®) as single agent or in combination with
Interleukin-2 (IL-2) in LOX-IMVI melanoma tumor xenografts. Fig. 73. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL, Aplidin®) as single agent or in combination with
Interferon-α 2a (INF-α) in LOX-IMVl melanoma tumor xenografts.
Fig. 74. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL) as single agent or in combination with Bevacizumab
(Avastin®) in CaKi- 1 renal tumor xenografts.
Fig. 75. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL) as single agent or in combination with Interleukin-2 (IL-
2) in CaKi-I renal tumor xenografts.
Fig. 76. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL) as single agent or in combination with Interferon-α 2a
(INF-α 2a) in CaKi-I renal tumor xenografts.
Fig. 77. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL) as single agent or in combination with Bevacizumab
(Avastin®) in MRI-H 121 renal tumor xenografts.
Fig. 78. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL) as single agent or in combination with Interleukin-2 (IL-
2) in MRI-H 121 renal tumor xenografts.
Fig. 79. Kinetics of net tumor volume after initiation of treatment with
Aplidine (APL) as single agent or in combination with Interferon-α 2a
(INF-α) in MRI-H121 renal tumor xenografts.
Fig. 80. Combination of Aplidine (A) + Lenalidomide (Revlimid®; R) +
Dexamethasone (D) in MMlS after 72 h of treatment. Aplidine doses are expressed in nM units, Lenalidomide doses are expressed in μM units and Dexamethasone doses are expressed in nM units.
Fig. 81. Combination of Aplidine (A) + Bortezomib (B) + Dexamethasone
(D) in MMlS after 72 h of treatment. Aplidine doses are expressed in nM units, Bortezomib doses are expressed in nM units and
Dexamethasone doses are expressed in nM units.
Fig. 82. Combination of Aplidine (A) + Bortezomib (B) + Lenalidomide
(Revlimid®; R) in MMlS after 72 h of treatment. Aplidine doses are expressed in nM units, Bortezomib doses are expressed in nM units and
Lenalidomide doses are expressed in μM units.
Fig. 83. Combination of Aplidine (A) + Thalidomide (T) +
Dexamethasone (D) in MMlS after 72 h of treatment. Aplidine doses are expressed in nM units, Thalidomide doses are expressed in μM units and Dexamethasone doses are expressed in nM units.
Fig. 84. Combination of Aplidine (A) + Melphalan (M) + Dexamethasone
(D) in MMlS after 72 h of treatment. Aplidine doses are expressed in nM units, Melphalan doses are expressed in μM units and
Dexamethasone doses are expressed in nM units.
Fig. 85. Combination of Aplidine (A) + Melphalan (M) + Bortezomib (B) in MMlS after 72 h of treatment. Aplidine doses are expressed in nM units, Melphalan doses are expressed in μM units and Bortezomib doses are expressed in nM units.
DETAILED DESCRIPTION OF THE INVENTION
By "cancer" it is meant to include tumors, neoplasias, and any other malignant tissue or cells. The present invention is directed to the use of Aplidine or an aplidine analogue in combination for the treatments of cancer in general, but more preferably for the treatment of lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia and lymphoma.
In order to study the possible potentiation of other anticancer agents with Aplidine we initiated a systematic study of drug combinations for possible use in the above mentioned cancer types. Drug combination studies were carried out on different types of cell lines. In vitro studies were performed using tumor cells lines such as NSCL A549, breast carcinoma MXl, promyelocytic leukemia HL60, colon adenocarcinoma HT29, prostate adenocarcinoma PC3, breast adenocarcinoma SKB R3 and acute lymphoblastic leukemia (MOLT3), which have different sensitivity to Aplidine (from low to high). Additional studies were also conducted with leukemias, lymphomas, multiple myeloma and melanoma cell lines. In addition, in vivo studies using melanoma, renal, myeloma, and lymphoma xenografts were used to establish the effect of Aplidine in combination with other standard agents. Finally, in vitro studies were conducted in multiple myeloma cell lines using triple combinations, that is combining Aplidine with two additional standard agents (a second and a third drug).
As a general conclusion we found that the cytotoxicity of Aplidine in tumor cells is greatly enhanced in combination with many of the standard agents used for this evaluation. Main synergistic effect was observed with the combination of Aplidine with paclitaxel (Taxol®), doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil (5-FU), cytosine arabinoside (AraC), carboplatin, 7-ethyl-lO-hydroxycamptothecin (SN38), etoposide (VP 16), melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide (Revlimid®), interleukin-2 (IL-2), interferon-α 2 (INF- α), dacarbazine (DTIC), bevacizumab (Avastin®), idarubicin, thalidomide, and rituxLmab. Additionally it was also found that an enhancement of the cytotoxicity was also obtained with triple combinations of Aplidine with the above mentioned agents.
Particularly preferred is the combination of Aplidine with paclitaxel in the treatment of cancer, and more particularly in the treatment of a cancer selected from breast cancer, leukemia, and prostate cancer. Particularly preferred is the combination of Aplidine with doxorubicin in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, colon cancer, prostate cancer, and multiple myeloma.
Particularly preferred is the combination of Aplidine with cisplatin in the treatment of cancer, and more particularly in the treatment of a cancer selected from breast cancer, and colon cancer.
Particularly preferred is the combination of Aplidine with arsenic trioxide in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, colon cancer, and prostate cancer.
Particularly preferred is the combination of Aplidine with 5- fluorouracil in the treatment of cancer, and more particularly in the treatment of a cancer selected from leukemia, lung cancer, breast cancer, and prostate cancer.
Particularly preferred is the combination of Aplidine with cytosine arabinoside in the treatment of cancer, and more particularly in the treatment of a cancer selected from lung cancer, breast cancer, and prostate cancer.
Particularly preferred is the combination of Aplidine with carboplatin in the treatment of cancer, and more particularly in the treatment of a cancer selected from colon cancer, prostate cancer, and melanoma. Particularly preferred is the combination of Aplidine with SN38 in the treatment of cancer, and more particularly in the treatment of lung cancer.
Particularly preferred is the combination of Aplidine with etoposide in the treatment of cancer, and more particularly in the treatment of a cancer selected from lymphoma, and multiple myeloma.
Particularly preferred is the combination of Aplidine with dexamethasone in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the combination of Aplidine with lenalidomide in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the combination of Aplidine with bortezomib in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the combination of Aplidine with dacarbazine in the treatment of cancer, and more particularly in the treatment of melanoma.
Particularly preferred is the combination of Aplidine with bevacizumab in the treatment of cancer, and more particularly in the treatment of renal cancer.
Particularly preferred is the combination of Aplidine with interleukin-2 in the treatment of cancer, and more particularly in the treatment of renal cancer. Particularly preferred is the combination of Aplidine with melphalan in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the combination of Aplidine with idarubicin in the treatment of cancer, and more particularly in the treatment of leukemia.
Particularly preferred is the combination of Aplidine with rituximab in the treatment of cancer, and more particularly in the treatment of lymphoma.
Particularly preferred is the combination of Aplidine with thalidomide in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the triple combination of Aplidine with lenalidomide and dexamethasone in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the triple combination of Aplidine with bortezomib and dexamethasone in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the triple combination of Aplidine with bortezomib and lenalidomide in the treatment of cancer, and more particularly in the treatment of multiple myeloma. Particularly preferred is the triple combination of Aplidine with bortezomib and thalidomide in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the triple combination of Aplidine with dexamethasone and thalidomide in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the triple combination of Aplidine with dexamethasone and melphalan in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
Particularly preferred is the triple combination of Aplidine with melphalan and bortezomib in the treatment of cancer, and more particularly in the treatment of multiple myeloma.
The compositions of the present invention may comprise all the components (drugs) in a single pharmaceutically acceptable formulation. Alternatively, the components may be formulated separately and administered in combination with one another. Various pharmaceutically acceptable formulations well known to those of skill in the art can be used in the present invention. Selection of an appropriate formulation for use in the present invention can be performed routinely by those skilled in the art based upon the mode of administration and the solubility characteristics of the components of the composition.
Examples of pharmaceutical compositions containing Aplidine or an aplidine analogue include liquid compositions (solutions, suspensions or emulsions) suitable for intravenous administration, and they may contain the pure compound or in combination with any carrier or other pharmacologically active compounds. Solubilised Aplidine shows substantial degradation under heat and light stress testing conditions, and a lyophilized dosage form was developed, see WO 99 42125 incorporated herein by reference.
Administration of Aplidine or compositions of the present invention is based on a Dosing Protocol preferably by intravenous infusion. We prefer that infusion times of up to 72 hours are used, more preferably 1 to 24 hours, with about 1 , about 3 or about 24 hours most preferred. Short infusion times which allow treatment to be carried out without an overnight stay in hospital are especially desirable. However, infusion may be around 24 hours or even longer if required. Infusion may be carried out at suitable intervals with varying patterns, illustratively once a week, twice a week, or more frequently per week, repeated each week optionally with gaps of typically one or several weeks.
The correct dosage of the compounds of the combination will vary according to the particular formulation, the mode of application, and the particular site, host and tumour being treated. Other factors like age, body weight, sex, diet, time of administration, rate of excretion, condition of the host, drug combinations, reaction sensitivities and severity of the disease shall be taken into account. Administration can be carried out continuously or periodically within the maximum tolerated dose. Further guidance for the administration of Aplidine is given in WO 01 35974 which is incorporated herein by reference in its entirety.
In one aspect, the present invention relates to synergistic combinations employing Aplidine or an aplidine analogue. An indication of synergy can easily be obtained by testing combinations and analyzing the results, for example by linear regression analysis. Reference is made to Figure 1 to illustrate this point. Alternative methods such as isobologram analysis are available for revealing synergism and can be employed for the present purposes.
Suitable aplidine analogues include the compounds defined by claim 1 of WO 02 02596, especially the compounds defined by any of the claims dependent on the claim 1. We incorporate by specific reference the disclosure in WO 02 02596 of compounds which are analogues of aplidine, including the claims.
EXAMPLES
EXAMPLE 1. In vitro studies to determine the effect of Aplidine in combination with another standard agent on tumor cell lines.
Aplidine as a single agent or in combination with selected standard chemotherapeutic agents was evaluated against several tumor cell lines to measure differences in cytotoxicity.
The following standard agents were selected as single agents and for combination with Aplidine: paclitaxel (Taxol®), doxorubicin, cisplatin, arsenic trioxide (Trisonex®), 5-fluorouracil (5-FU), cytosine arabinoside (AraC), carboplatin and 7-ethyl-lO-hydroxycamptothecin (SN38).
The single agent Aplidine is cytotoxic to several cancer types with varying potency. For this reason representative tumor cell lines were selected that have a low, medium or high sensitivity to Aplidine. The tumor cell lines that were used are listed in Table 1.
Table 1
Figure imgf000024_0001
The screening was performed in two parts:
a. In the first set of assays, IC50 values were determined for each compound after 72 hours of drug exposure in each of the tumor cell lines.
All cell lines were maintained in respective growth media at 370C, 5% CO2 and 98% humidity. All media formulations did not contain antibiotic. Day before plating cells all cultures were fed with fresh, complete growth media. On the harvest (plating) day cells were counted by Trypan Blue exclusion staining method (basic cell culture). Cells were harvested and seeded in 96 well microtiter plates at 10,000 cells per well in 190 μL of media and incubated for 24 hours to allow the cells to attach before drug addition. Cells were treated with the drugs and the cytotoxic effect was measured by the MTS Assay (Tetrazolium), which is a colorimetric method for determining the number of viable cells. After the 72 hours of incubation with drug, 25 μL of MTS+PMS solution was added to each microtiter well and incubated for 4 hours at 370C. Plates were then removed from incubator and placed on plate shaker for 5 minutes (covered with aluminium foil for protection from light). Optical densities were read at 490 nm on spectrophotometer plate reader. Data was analysed using SoftMax v 3.12 program.
IC50 was calculated, which is approximate equivalent of IG50 (concentration at which 50% growth inhibition is measured). A regression curve using SoftMax program was generated, and then 50% inhibition concentration was manually interpolated and converted that concentration to molar (M) by dividing by the molecular weight of the compound. The individual IC50 values (72 hours drug exposure) are shown in table 2. The IC50 values represent 100% of the drug concentration . Table 2
Figure imgf000025_0001
Table 2 (cont.)
Figure imgf000026_0001
Figure imgf000027_0001
b. In a second set of assays, each cell line was incubated with Aplidine in combination with each of the standard agents mentioned above in the following combinations of unique IC50 concentrations:
IC50 of Aplidine IC50 of Standard Agent
100% 0% 75% 25% 60% 40%
50% 50%
40% 60%
30% 70%
25% 75%
0% 100%
0% 0%
The microliter plates were incubated for 72 hrs, at 5% CO2 and 37°C. The cytotoxic effect was measured by MTS Assay. Optical densities read at 490 nm. Normalized data was plotted and interpreted as described. Data was analysed as:
1. Prism (Graphpad) software program was used to normalized the data to control values (100% = cell growth in the absence of agent (drug); 0% = blank control).
2. Data normalized were plotted as scatter plots. A line was drawn connecting the values of 100% IC50 for each agent (drug). Values significantly above the line indicated antagonism, below indicated synergy, and on the line indicated additivity.
Statistical treatment of data followed Laska E. et al. Biometrics (1994) 50:834-841 and Greco et al. Pharmacol Rev. (1995) 47: 331-385. Combinations at tested dose ratios were judged to be synergistic when inhibition of cell proliferation exceeded maximum inhibition values for each drug separately (at 100% IC50). Conversely, antagonism was concluded when inhibition was lower than both maxima. Additivity was concluded when the effects of combinations did not differ significantly from the maxima for both drugs. Statistical significance was assessed by performing a student's t-test on the inhibition at each dose ratio versus the inhibition at the maximum for each drug. Overall significance of drug combinations for each cell line was dependent on showing statistical significance for greater than 50% of dose ratios.
As a visual aid, response values were plotted on a scatter plot with dose ratios given on the x-axis and % response values on the y- axis. A horizontal line was drawn between the two endpoint response values (E.g. between the response values for 100% IC50 Aplidine and 100% IC50 standard chemo therapeutic agent). In cases where response values at the two endpoints were approximately equivalent, points lying above or below this predicted line of additivity could be interpreted as representing antagonistic or synergistic drug interaction, respectively.
The in vitro combinations of each drug with Aplidine have the potential to be synergistic, additive or antagonistic. Synergistic cytotoxicity to tumor cells is an optimal effect and implies that the combination of Aplidine with another drug is more effective than either drug alone.
According to this assay it was found that:
a. The combination Aplidine with paclitaxel showed synergism in breast adenocarcinoma SKB R3 cells (Figure 2), acute lymphoblastic leukaemia MOLT3 cells (Figure 3) and prostate adenocarcinoma PC3 cells (Figure 4). Trend to additivity was observed in promyelocytic leukemia HL60 cells (Figure 5), breast carcinoma MXl cells (Figure 6) and NSCL A549 cells (Figure 7).
b. The combination of Aplidine with doxorubicin showed synergism in NSCL A549 cells (Figure 8), colon adenocarcinoma HT29 cells (Figure 9) and prostate adenocarcinoma PC3 cells (Figure 10). Additivity was observed in acute lymphoblastic leukaemia MOLT3 cells (Figure 11), breast carcinoma MXl cells (Figure 12) and breast adenocarcinoma SKBR3 cells (Figure 13).
c. The combination of Aplidine with cisplatin showed synergism in breast carcinoma MXl cells (Figure 14) and colon adenocarcinoma HT29 cells (Figure 15). Additivity was observed in breast adenocarcinoma SKB R3 cells (Figure 16) and acute lymphoblastic leukaemia MOLT3 cells (Figure 17), and in NSCL A549 cells (Figure 18) trends to synergism were found.
d. The combination of Aplidine with arsenic trioxide showed synergism in NSCL A549 cells (Figure 19), colon adenocarcinoma HT29 cells (Figure 20) and prostate adenocarcinoma PC3 cells (Figure 21). Additivity was observed in acute lymphoblastic leukaemia MOLT3 cells (Figure 22).
e. The combination of Aplidine with 5-fluorouracil showed synergism in promyelocytic leukemia HL60 cells (Figure 23), breast adenocarcinoma SKBR3 cells (Figure 24), NSCL A549 cells (Figure 25) and prostate adenocarcinoma PC3 cells (Figure 26). Additivity was observed in colon adenocarcinoma HT29 cells (Figure 27).
f. The combination of Aplidine with cytosine arabinoside showed synergism in breast adenocarcinoma SKBR3 cells (Figure 28), NSCL breast A549 cells (Figure 29) and prostate adenocarcinoma PC3 cells (Figure 30). Additivity was found in promyelocytic leukemia HL60 cells (Figure 31) and colon adenocarcinoma HT29 cells (Figure 32).
g. The combination of Aplidine with carboplatin showed synergism in prostate adenocarcinoma PC3 cells (Figure 33) and colon adenocarcinoma HT29 cells (Figure 34). Additivity was observed in NSCL A549 cells (Figure 35), acute lymphoblastic leukaemia MOLT3 cells (Figure 36) and breast carcinoma MXl cells (Figure 37).
h. The combination of Aplidine with SN38 showed synergism in NSCL A549 cells (Figure 38). Additivity was observed in breast adenocarcinoma SKBR3 cells (Figure 39), promyelocytic leukemia HL60 cells (Figure 40) and prostate adenocarcinoma PC3 cells (Figure 41).
Example 2. In vitro studies to determine the effect of Aplidine in combination with another standard agent on leukemia, lymphoma, multiple myeloma and melanoma tumor cell lines.
Following the same procedure as disclosed in example 1 , Aplidine as a single agent or in combination with selected standard chemo therapeutic agents, was evaluated against several tumor cell lines to measure differences in cytotoxicity.
The following standard agents were selected as single agents and for combination with Aplidine: etoposide (VP 16) and carboplatin. The tumor cell lines selected for this assay are shown in Table 3.
Table 3
Figure imgf000031_0001
Figure imgf000032_0001
Cell culture method
All cell lines were maintained in respective growth media at 37°C, 5% CO2 and 98% humidity. All media formulations did not contain antibiotic. The day before plating cells all cultures were fed with fresh, complete growth media. On the harvest (plating) day cells were counted by Trypan Blue exclusion staining method.
Cell plating
Cells were harvested and seeded in 96 well microtiter plates at 15,000 cells per well in 190 μl of media and incubated for 24 hours to allow the cells to attach before drug addition.
Drug Treatment
Stock solution of Aplidine was prepared in 100% DMSO at 5 mg/ml. Stock solutions of chemo therapeutic agents VP 16 and carboplatin were prepared in 100% DMSO at the concentration 2 mg/ml for both drugs.
Cells were treated with Aplidine and the other standard agent at range as listed below, and individual drug concentration were made in triplicates per plate. The concentration of the tested agents used is expressed as a percent of the individual agent's IC50, which were determined as in Example 1.
IC50 of Aplidine IC50 of Standard Agent
100% 0% 75% 25% 60% 40% 50% 50% 40% 60% 30% 70% 25% 75% 0% 100%
The individual IC50 values of each agent for each cell line are shown in table 4.
Table 4
Figure imgf000033_0001
Figure imgf000034_0001
The cytotoxic effect was measured by the MTS Assay (Tetrazolium) , which is a colorimetric method for determining the number of viable cells.
After 72 hours of incubation with tested agents 25 μl of MTS+PMS solution was added to each microtiter well and incubated for 4 hours at 37°C. Plates were then removed from incubator and placed on plate shaker for 5 minutes (covered with aluminum foil for protection from light). Optical densities were read at 490nm on spectrophotometer plate reader. Data was analyzed as: 1. Prism (Graphpad) software program was used to normalized the data to control values (100% = cell growth in the absence of agent (drug); 0% = blank control).
2. Data normalized were plotted as scatter plots. A line was drawn connecting the values of 100% IC50 for each agent (drug). Values significantly above the line indicated antagonism, below indicated synergy, and on the line indicated additivity.
Statistical treatment of data followed Laska E. et al. Biometrics (1994) 50:834-841 and Greco et al. Pharmacol Rev. (1995) 47: 331-385. Combinations at tested dose ratios were judged to be synergistic when inhibition of cell proliferation exceeded maximum inhibition values for each drug separately (at 100% IC50). Conversely, antagonism was concluded when inhibition was lower than both maxima. Additivity was concluded when the effects of combinations did not differ significantly from the maxima for both drugs. Statistical significance was assessed by performing a student's t-test on the inhibition at each dose ratio versus the inhibition at the maximum for each drug. Overall significance of drug combinations for each cell line was dependent on showing statistical significance for greater than 50% of dose ratios.
As a visual aid, response values were plotted on a scatter plot with dose ratios given on the x-axis and % response values on the y- axis. A horizontal line was drawn between the two endpoint response values (E.g. Between the response values for 100% IC50 Aplidine and 100% IC50 standard chemotherapeutic agent). In cases where response values at the two endpoints were approximately equivalent, points lying above or below this predicted line of additivity could be interpreted as representing antagonistic or synergistic drug interaction respectively. In Figure 42A and 42B it is shown the in vitro activity data of Aplidine in combination with VP16 against HL-60 cells. According to this data additive effect is obtained.
In Figure 43A and 43B it is shown the in vitro activity data of Aplidine in combination with VP 16 against K562 cells. According to this data additivity is obtained.
In Figure 44A and 44B it is shown the in vitro activity data of Aplidine in combination with VP 16 against MOLT-3 cells. According to this data additivity is obtained.
In Figure 45A and 45B it is shown the in vitro activity data observed with Aplidine in combination with VP 16 against MCl 16 cells. According to this data additivity is obtained in this tumor cell line.
In Figure 46A and 46B it is shown the in vitro activity data observed with Aplidine in combination with VP 16 against RAMOS cells. According to this data synergism is obtained in this tumor cell line.
In Figure 47A and 47B it is shown the in vitro activity data observed with Aplidine in combination with VP 16 against U937 cells. According to this data additivity is obtained.
In Figure 48A and 48B it is shown the in vitro activity data observed with Aplidine in combination with VP 16 against NCI-H929 cells. According to this data synergism is obtained.
In Figure 49A and 49B it is shown the in vitro activity data observed with Aplidine in combination with VP 16 against HUNS-I cells. According to this data additivity is obtained in this tumor cell line. In Figure 50A and 5OB it is shown the in vitro activity data observed with Aplidine in combination with VP 16 against U266B-1 cells. According to this data additivity is obtained in this tumor cell line.
In Figure 5 IA and 5 IB it is shown the in vitro activity data observed with Aplidine in combination with VP 16 against RPMI 8226 cells. According to this data additivity is obtained in this tumor cell line.
In Figure 52 it is shown the in vitro activity data observed with Aplidine in combination with carboplatin against LOX-I-MVI cells. According to this data additivity is obtained in this tumor cell line.
In Figure 53 it is shown the in vitro activity data observed with Aplidine in combination with carboplatin against UACC-257 cells. According to this data synergism is obtained in this tumor cell line.
Example 3: In vitro studies to determine the effect of Aplidine in combination with other standard agents on multiple myeloma tumor cell lines.
Multiple myeloma (MM) is a malignant disease of plasma cells, generally originated from a clone of plasma cells which proliferate and accumulate in the bone marrow. Clinicopathological features of patients with myeloma include accumulation of monoclonal protein in the blood or urine, lytic bone lesions, anaemia and renal dysfunction (Sirohi B. et al. Lancet (2004) 363: 875-87).
Actual treatment for myeloma relies on high dose therapy supported by stem cell transplantation. Despite the advances in the last decade, nowadays MM remains an incurable disease with a median overall survival for patients of 2 to 5 years (Sirohi B. et al. Lancet (2004) 363: 875-87). New treatment approaches are needed to improve patients outcome following three major lines of investigation: a) enhancing the efficacy of chemotherapy through the use of high dose; b) enhancing the host immune response against myeloma cells; and c) developing novel drugs with more specific targets that may interfere not only with myeloma cells but also the bone marrow microenvironment (San Miguel JF et al. Curr. Treat. Options Oncol. (2003) 4: 247-58). Hopefully, the combination of two or more of these therapeutic agents will lead to an improved anti-MM efficacy and longer survival rates.
Herewith we report several studies on the effect of Aplidine as a new drug in the treatment of multiple myeloma. These studies include:
1) cytotoxic efficacy of Aplidine (alone or in combination) on several MM cell lines using cell viability (MTT assay) and apoptosis (annexin V staining) assays.
2) cytotoxic efficacy of combinations of Aplidine and other classical and recently developed drugs in the treatment of this disease.
Material and methods
Cell lines and cell culture reagents
Four MM-derived cell lines were used in this study: the dexamethasone-sensitive (MM. IS) and dexamethasone-resistant (MM. IR) variants of the human multiple myeloma cell line MM. l (Greenstein S. et al. Exp. Hematol (2003) 31:.271-82) which were kindly provided by Dr. S Rudikoff, Bethesda MD; and the U266 and its melphalan-resistant counterpart U266 LR7 cell lines were obtained from Dr. W. Dalton, Tampa, FL. All MM cell lines were grown in RPMI 1640 medium supplemented with 10% heat-inactivated fetal bovine serum, 100 U/ml penicillin, 100 μg/ml streptomycin and 2 mM L- glutamine. All cell culture media and reagents were purchased from Invitrogen Corporation (Carlsbad, CA).
Cell viability assays
The analysis of MM cell proliferation was assessed using the methylthiotetrazole (MTT; Sigma, St. Louis MO) colorimetric assay. MM cell lines were seeded at a density of 50000 cells/200μl medium per well in 48-well plates, and treated with a determined drug dose and time. Two hours before the end of the treatment, a MTT solution (5 mg/ml in PBS; usually a 10% of the volume in each well) was added and the tetrazolium salt was reduced by metabolically active cells to coloured formazan crystals. After solubilization of these crystals by overnight incubation with 10% SDS-HCl solution, absorbance was measured at 570 nm with correction at 630 nm. Four wells were analyzed for each condition, and the results are presented as the mean ± SD of quadruplicates of a representative experiment that was repeated at least three times.
Western blot
Cells were collected and washed with PBS, and total cell lysates were obtained after incubation in ice-cold lysis buffer (140 mM NaCl, 10 mM EDTA, 10% glycerol, 1% Nonidet P-40, 20 mM Tris (pH 7.0), 1 μM pepstatin, 1 μg/ml aprotinin, 1 μg/ml leupeptin, 1 mM sodium orthovanadate). Samples were then centrifuged at 10,00Og at 40C for 10 min and equal amounts of protein in supernatants were resolved by 6% - 12.5% SDS-PAGE. Proteins were then transferred onto nitrocellulose or PVDF membranes, blocked by incubation in 5% defatted dry milk in PBST buffer (0.05% -Tween 20 in PBS) and subsequently incubated with the specific primary antCruz Technologies, ibody [the anti-p-c-jun, anti-p-Erkl/2 and anti-Erk5 antibodies were purchased from Santa Cruz Biotechnologies (Santa Cruz, CA), whereas the anti-p-p38 was obtained from Cell Signaling (Danvers, MA) and the anti-PARP antibody from Becton Dickinson Biosciences (Bedford, MA)]. After a second incubation with the correspondent secondary antibody immunoblots were developed by enhanced chemilunescence (ECL; Amersham, Arlington Heights, IL). Identification of activated JNK and Erk5 required previous immunoprecipitation of protein lysates with the correspondent specific antibodies and protein A sepharose.
Isobologram analysis
The interaction between Aplidine and other anti-MM agents was analyzed using the Calcusyn software program (Biosoft, Ferguson, MO). Data from cell viability assay (MTT) were expressed as the fraction of cells affected by the dose (Fa) in drug treated cells as compared to untreated cells (control). This program is based upon the Chou-Talalay method (Chou TC et al. Adv. Enzyme Regul. (1984) 22: 27-55) according to the following equation CI = (D) 1/(Dx) I + (D) I (D) 2 /(Dx) I (Dx) 2 where (D)I and (D) 2 are the doses of drug 1 and 2 that have the same x effect when used alone. CI values less than 1.0 indicate synergism, CI values ≡ 1.0 indicate an additive effect, whereas values more than 1 correspond to antagonistic effect.
Results
Dose response to Aplidine in MM-deriυed cell lines
We first determined whether Aplidine affects cell viability using the MTT assay on MM-derived cell lines both sensitive and resistant to conventional therapy drugs. As seen in figure 54, Aplidine treatment for 48 h induces a similar decrease in cell viability in a dose-dependent manner in all cell lines tested. Fifty percent decrease in viable cells (IC50) after 48 h treatment was within the 1-10 nM range for the four cell lines tested.
Efficacy of Aplidine was also compared to other classic (Melphalan, Dexamethasone) and new drugs (Bortezomib) in the treatment of MM. Figure 55 shows superior potency of Aplidine as compared to the other drugs when tested at 0.1-1 nM doses for 48 h on the MM. IS cell line; its effect was similar to that of Bortexomib at maximal doses (10-100 nM).
Evaluation of synergism in double combinations of Aplidine
Since continuous exposure to MM chemotherapy is in many cases associated to increased toxicity and development of the noυo drug resistance, we tested whether the combination of minimally toxic concentrations of Aplidine and other drugs would affect MM cell viability. Specifically, Aplidine was combined with classic drugs in the treatment of MM (such as Dexamethasone or Melphalan), also with recently developed anti-myeloma agents (Bortezomib), and with drugs that would specifically target the bone-marrow microenvironment (lenalidomide (Revlimid®)).
Time course experiments at days 1, 2, 3 and 6 were also performed for the mentioned therapeutic agents to determine the appropriate suboptimal doses (10% to 30% growth inhibition) for combinatorial experiments as well as to explore for the duration of the treatments. Combination experiments of Aplidine and the rest of the drugs were performed after incubation for 3 days or 6 days. Cell growth of MM. IS cell line was measured by MTT assay as described above, and the percentages of inhibition were analyzed by the CalcuSyn Program. The computer- calculated combination index (CI) was used to judge the outcomes of a combination: CI>1, CI=I, and CI<1 indicating antagonism, additive, and synergistic effects, respectively. The conformity of data to the median-effect principle can be readily manifested by the linear correlation coefficient (r) of the median-effect plot : log (fa/fu) = m log (D)- m log (Dm), wherein D is the dose, Dm is the dose required for a 50% effect, fa is the fraction affected by dose, fu is the unaffected fraction, and m is a coefficient of the sigmoidicity of the dose-effect curve. For each double combination a non-constant ratio combination was utilized. Each experiment was repeated three times, and a minimum of three data points for each single drug and three combinations were performed.
Combination of Aplidine with Dexamethasone
At day 3, a synergism was observed for 0.5 nM Aplidine/ 1 nM and 10 nM Dexamethasone combinations (Figure 56).
As illustrated in figure 57, the combination was more synergistic at 6 days.
Combination of Aplidine with Melphalan
The following combinations showed nearly additive effects at 3 and 6 days (Figures 58 and 59 respectively).
Combination of Aplidine with Doxorubicin
At 3 days, only one combination was moderately synergic (Figure 60, combination 1) and two combinations were additive (Figure 60, combinations 3 and 4) .
At 6 days only two combinations were additive (Figure 61, combination 7 and 8).
Combination of Aplidine with lenalidomide (Reylimid®) In the present study, the combination of Aplidine and lenalidomide (Revlimid®) showed greater degrees of synergism than all other combinations examined (Figure 62).
Remarkably, the synergism was more important at 6 days (Figure 63).
Combination of Aplidine with Bortezomib
This combination showed antagonism at 3 days (Figure 64); at 6 days synergism was found for two combinations (Figure 65, combination 4 and 6).
Example 4: In vivo studies to determine the effect of Aplidine in combination with other standard agents in melanoma and renal xenografts.
The purpose of this study was to evaluate the antitumor activity of Aplidine when administered with an antitumoral standard agent, both administered using multiple dosing schedule in several types of tumor xenografts in female athymic mice.
Athymic nude female mice were received from Harlan Sprague Dawley, Madison, Wisconsin at 4-5 weeks of age. Mice were acclimated to the laboratory for at least one week prior to implantation of tumor. Animals were housed in static cages with food and water allowed ad libitum. Experimental animals were implanted with either tumor fragments derived from transplantation established human tumors or with cells obtain directly from in vitro culture. Tumors were implanted subcutaneously on the right flank on Day 0. Tumor size measurements were recorded twice weekly beginning on day 4 or 5 using vernier calipers. The formula to calculate volume for a prolate ellipsoid was used to estimate tumor volume from 2- dimentional tumor measurements: tumor volume (mm3) = (length x width2) ÷ 2. Assuming unit density, volume was converted to weight (i.e., 1 mm3 = 1 mg). When tumors reached an approximate volume range of 100 ± 15 mg, mice were randomized into treatment and control groups. Treatments were initiated and administered on an individual body weight basis. A dose range finding study was performed on each tumor model to determine the appropriate dose level of each compound used in the combination studies.
The following standard of care agents for the various cancer types (indications) were combined with Aplidine (APL) to determine if the combination therapy will provide a greater antitumor activity when compared to the combined activity of the two agents administered as monotherapies .
Figure imgf000044_0001
MRI-H 121 APL + Interleukin-2 (IL-2)
APL + Interferon-α 2a (INF-α)
APL + Bevacizumab (Avastln®)
Tables 5-10 show the kinetics of net tumor volume (mean ± S. E. M., mg) after initiation of treatment with Aplidine either alone or in combination with a standard of care agent in melanoma and renal cancer xenografts.
Table 5 and figure 66 and 67 show kinetics of net tumor volume after initiation of treatment with Aplidine (Aplidin®) as single agent or in combination with DTIC and carboplatin in MRI-H 187 melanoma tumor xenografts. Aplidine was administered every day for 9 consecutive days by intraperitoneal (i.p.) injection, saline control (sterile saline) every day for 9 consecutive days by i.p. injection, DTIC every day for 5 consecutive days by i.p. injection and Carboplatin one dose every 4 days for a total of 4 treatments by i.p. injection.
Table 5
Figure imgf000045_0001
Figure imgf000046_0001
S.E.M.=Standard error of the mean
Table 5 (cont.)
Figure imgf000046_0002
Figure imgf000047_0001
Table 5 (cont.)
Figure imgf000047_0002
From this xenograft study it was concluded that in melanoma MRI-H 187 cells the combination of Aplidine with DTIC and Aplidine with carboplatin show a clear statistically significant potentiation of antitumor activity, being more remarkable in the case of the combination with carboplatin. Table 6 and figure 68 and 69 show kinetics of net tumor volume after initiation of treatment with aplidine (Aplidin®) as single agent or in combination with Interleukin-2 (IL- 2) and Interferon-α 2a (INF-α) in MRI-H- 187 melanoma tumor xenografts. Aplidine was administered every day for 9 consecutive days by i.p injection, saline control (sterile saline) every day for 9 consecutive days by i.p injection, IL-2 every day during 5 weekdays (Monday-Friday) for 3 weeks by i.p. injection and INF-α every day during 5 weekdays (Monday-Friday) for 3 weeks by subcutaneous (s.c.) injection. Table 6
Figure imgf000048_0001
Figure imgf000049_0001
S.E.M.=Standard error of the mean
Table 6 (cont.)
Figure imgf000049_0002
From this xenograft study it was concluded that in melanoma MRI-H 187 cells the combination of Aplidine with IL-2 and Aplidine with INF-α showed additivity.
Table 7 and figure 70 and 71 show kinetics of net tumor volume after initiation of treatment with Aplidine (APL) as single agent or in combination with DTIC and carboplatin in LOX-IMVI melanoma tumor xenografts. Aplidine was administered every day for 9 consecutive days by i.p. injection, saline control (sterile saline) every day for 9 consecutive days by i.p. injection, DTIC every day for 5 consecutive days by i.p. injection and Carboplatin one dose every 4 days for a total of 4 treatments by i.p. injection.
Table 7
Figure imgf000050_0001
S.E.M.=Standard error of the mean From this xenograft study it can be concluded that in melanoma LOX-IMVI cells the combination of Aplidine with DTIC shows an additive pattern and the combination of Aplidine with carboplatin shows a trend to synergism.
Table 8 and figure 72 and 73 show kinetics of net tumor volume after initiation of treatment with Aplidine (APL) as single agent or in combination with Interleukin-2 (IL-2) and Interferon-α 2a (INF-α) in LOX-IMVI melanoma tumor xenografts. Aplidine was administered every day for 9 consecutive days by i.p injection, saline control (sterile saline) every day for 9 consecutive days by i.p injection, IL-2 every day during 5 weekdays (Monday- Friday) for 3 weeks by i.p. injection and INF-α every day during 5 weekdays (Monday-Friday) for 3 weeks by s.c. injection.
Table 8
Figure imgf000051_0001
Figure imgf000052_0001
S.E.M.=Standard error of the mean
From this xenograft study it was concluded that in melanoma LOX-IMVI cells the combination of Aplidine with IL-2 and Aplidine with INF-α show additivity.
Table 9 and figure 74, 75 and 76 show kinetics of net tumor volume after initiation of treatment with aplidine (APL) as single agent or in combination with Bevacizumab (Avastin®), Interleukin-2 (IL-2) and Interferon-α 2a (INF-α) in CaKi-I renal tumor xenografts. Aplidine was administered every day for 9 consecutive days by i.p injection, saline control (sterile saline) every day for 9 consecutive days by i.p injection, Bevacizumab (Avastin®) every 3 days for a total of 4 treatments by i.p. injection, IL-2 every day during 5 weekdays (Monday-Friday) for 3 weeks by i.p. injection and INF-α every day during 5 weekdays (Monday- Friday) for 3 weeks by s.c. injection.
Table 9
Figure imgf000052_0002
Figure imgf000053_0001
S.E.M.=Standard error of the mean
Table 9 I cont.)
Net' Tumor Volume ± S .E.M. (mg)
Drug
DAY 29 DAY 33 DAY 36 DAY 41 DAY 47 DAY 50
Saline 1939 ±
639± 64 741 i 69 897 → :75 1149: ±76 1595 ±74 Control 110
Figure imgf000054_0001
From this xenograft study it was concluded that in renal CaKi-I cells the combination of Aplidine with Avastin® and Aplidine with IL-2 show synergism, being more remarkable in the case of the combination with IL-2. On the other hand, the combination of Aplidine with INF-α shows an additive pattern.
Table 10 and figure 77, 78 and 79 show kinetics of net tumor volume after initiation of treatment with aplidine (APL) as single agent or in combination with Bevacizumab (Avastin®), Interleukin-2 (IL-2) and Interferon-α 2a (INF-α) in MRI-H 121 renal tumor xenografts. Aplidine was administered every day for 9 consecutive days by i.p injection, saline control (sterile saline) every day for 9 consecutive days by i.p injection, Bevacizumab (Avastin®) every 3 days for a total of 4 treatments by i.p. injection, IL-2 every day during 5 weekdays (Monday- Friday) for 3 weeks by i.p. injection and INF-α every day during 5 weekdays (Monday-Friday) for 3 weeks by s.c. injection.
Table 10
Figure imgf000055_0001
S.E.M.=Standard error of the mean
Figure imgf000056_0001
From this xenograft study it was concluded that in renal MRI- Hl 21 cells the three combinations show an additive pattern. Example 5. Additional in-vitro assays in multiple myeloma (RPMI-8226 and U266B1) cell lines were conducted to determine the effect of Aplidine in combination with two standard of care chemotherapeutic agents (Bortezomib and Melphalan).
Aplidine as single agent or in combination with either Bortezomib or Melphalan were evaluated against multiple myeloma cell lines, specifically RPMI-8226 and U266B1 cell lines.
These cell lines were cultured in RPMI 1640 medium with 10% FBS and 1% L-Glutamine. Each cell line was plated in a 96 well plates at 20,000 cell per well.
First Aplidine, Bortezomib and Melphalan were tested alone to determine the IC50 value for each of them individually. To determine IC50 value each drug was check at different range of drug concentration, following the procedure as disclosed in Example 1. Table 11 shows the individual IC50 obtained with each of the three drugs against the two multiple myeloma cell lines.
Table 11
Figure imgf000057_0001
In the next step either Bortezomib or Melphalan were combined with Aplidine. In these experiments, concentrations of Aplidine were used with a 1: 10 serial dilution. Each serial dilution was paired with 4 different concentrations of Bortezomib or Melphalan.
The plates were incubated for 3 days at 370C and 5% CO2. The plates were read using the Promega MTS assay system with MTS being metabolized by living cells turning into formazan which is fluorescent at 490nm wavelength. This is an indirect measure of cell viability. These were analyzed using the Softmax Pro program which determines cell viability based percent of control wells. This IC50 data was then transferred into the CalcuSyn program for combination index analysis. CalcuSyn compares IC50 the values of the drugs alone with that of the drugs in combination using an algorithm to determine a combination index. It is important to note that the combination index (CI) is a reflection of the combination effect of the two drugs: CI = 1 indicates an additive effect; CI <1 indicates a synergistic effect; and CI>1 indicates a antagonistic effect.
Table 12 summarises those doses wherein a synergistic effect was observed in the combination of Aplidine with Bortezomib against RMPI 8226 cell line:
Table 12
Figure imgf000058_0001
Table 13 summarises those dose wherein a synergistic effect was observed in the combination of Aplidine with Melphalan against RMPI 8226 cell line: Table 13
Figure imgf000059_0001
Table 14 summarises those doses wherein a synergistic effect was observed in the combination of Aplidine with Bortezomib against U266B1 cell line:
Table 14
Figure imgf000059_0002
Figure imgf000060_0001
Table 15 summarises those doses wherein a synergistic effect was observed in the combination of Aplidine with Melphalan against U266B1 cell line:
Table 15
Figure imgf000060_0002
Figure imgf000061_0001
Example 6. Additional in-vitro assays in leukemia (MOLT4 and K- 562) cell lines were conducted to determine the effect of Aplidine in combination with a standard of care chemotherapeutic agent such as Idarubicin.
Aplidine as single agent or in combination with Idarubicin were evaluated against leukemia cell lines, specifically MOLT4 and K562 cell lines.
These cell lines were cultured in RPMI 1640 medium with 10% FBS and 2mM L-Glutamine. Each cell line was plated in a 96 well plates at 20,000 cell per well.
First Aplidine and Idarubicin were tested alone to determine the IC50 value for each of them individually. To determine IC50 value each drug was check at different range of drug concentration, following the procedure as disclosed in Example 1. Table 16 shows the individual IC50 obtained with each of the two drugs against the two leukemia cell lines.
Table 16
Figure imgf000061_0002
In the next step Idarubicin was combined with Aplidine. In the experiment related to MOLT4 cell line, concentrations of Aplidine were used with a 1: 10 serial dilution. Each serial dilution was paired with 4 different concentrations of Idarubicin. In the experiment related to K562 cell line, concentrations of Aplidine were used with a 1:5 serial dilution, and Idarubicin concentration was added to each combination set based on a ratio. Thus, combination A was 1: 1, combination B was 0.008: 1, combination C was 6.4E-05: l and combination D was 5.12E-07: l.
The plates were incubated for 3 days at 370C and 5% CO2. The plates were read using the Promega MTS assay system with MTS being metabolized by living cells turning into formazan which is fluorescent at 490nm wavelength. This is an indirect measure of cell viability. These were analyzed using the Softmax Pro program which determines cell viability based percent of control wells. This IC50 data was then transferred into the CalcuSyn program for combination index analysis. CalcuSyn compares IC50 the values of the drugs alone with that of the drugs in combination using an algorithm to determine a combination index. It is important to note that the combination index (CI) is a reflection of the combination effect of the two drugs: CI = 1 indicates an additive effect; CI <1 indicates a synergistic effect; and CI>1 indicates a antagonistic effect.
Table 17 summarises those doses wherein a synergistic effect was observed in the combination of Aplidine with Idarubicin against MOLT4 cell line:
Table 17
Figure imgf000062_0001
Table 18 summarises those doses wherein a synergistic effect was observed in the combination of Aplidine with Idarubicin against K562 cell line:
Table 18
Figure imgf000063_0001
Example 7: In vivo studies to determine the effect of Aplidine in combination with another standard agent in melanoma xenografts. The purpose of this study was to evaluate the antitumor efficacy of Aplidine when administered in combination with Carboplatin against subcutaneous-implanted UACC-257 human melanoma cells in female, athymic NCr- nu/nu mice.
The animals were housed in microisolator cages, up to five per cage in a 12-hour light/ dark cycle. Six-weeks-old female, athymic NCr- nu/nu mice were acclimated in the laboratories for one week prior to the experiment.
Each mouse was inoculated subcutaneously near the right flank with UACC-257 human melanoma cells from an in vitro cell culture using a 23-gauge needle. Each mouse received 2 x 107 cells resuspended in 0.2 mL of Matrigel®. A vial with UACC-257 human melanoma frozen cells was thawed and cultured in RPMI 1640 medium containing low glucose (2,000 mg/L), sodium bicarbonate (1,500 mg/mL), 2 mM L-glutamine, and 10% fetal bovine serum (complete medium), and grown in a +37°C incubator in a humidified atmosphere with 5% CO2 until the necessary number of cells for inoculation of mice was obtained. Cells were harvested after four passages in culture. The cells were removed from the flasks, placed in 50-mL centrifuge tubes and centrifuged at 1,000 rpm for 10 minutes in a refrigerated centrifuge. The cell pellets were resuspended in fresh complete medium. The cell count and viability were determined with a Beckman Coulter VI CELL XR cell counter and viability analyzer. The cell suspension was centrifuged, and the cell pellet was resuspened in Matrigel® at a cell density of 1.0 x 108 cells/mL and placed on wet ice. The final concentration of Matrigel® in the cell suspension was 56.9%. On the day of cell harvest cell viability was 98.9%. The day of tumor cell inoculation was designated as Day 0. Individual tumors grew to 150-245 mg in weight (150-245 mm3 in size) on the day of treatment initiation, Day 13 after tumor cell inoculation. Forty animals with tumors in the proper size range were assigned to four treatment groups so that the median tumor weights on the first day of treatment were as close to each other as possible.
The experiment consisted of a vehicle-treated control group of 10 mice and three drug- treated groups of 10 mice per group for a total of 40 mice on the first day of treatment, Day 13 after tumor cell inoculation. Animals in Group 1 were treated i.p. every day for 9 consecutive days (Days 13-21) with a vehicle of Aplidine diluted with saline. Aplidine was administered i.p. every day during 9 days (Days 13- 21) at a dosage of 60 μg/kg/dose alone (Group 2) or in combination with Carboplatin (Group 4). Carboplatin was administered i.v. every 4 days for a total of three treatments (Days 13, 17, and 2 1) at a dosage of 50 mg/kg/dose alone (Group 3) or in combination with Aplidine (Group 4). On days when both compounds were administered in Group 4, Aplidine was injected first to all ten animals in the group, followed immediately by the administration of Carboplatin (Group 4).
Group 1 was treated i.p. with 0.18% cremophor EL/0.18% ethanol/0.84% WFI/98.8% saline (injection volume: 0.1 mL/10 g body weight). Aplidine was reconstituted with a vehicle containing 15% cremophor EL/ 15% ethanol/70% WFI and diluted with saline (injection volume: 0.1 mL/ 10 g body weight). Carboplatin was prepared in WFI (injection volume: 0.1 mL/10 g body weight).
Animals were observed daily and clinical signed were noted. The s. c. tumors were measured and the animals were weighed twice weekly starting with the first day of treatment, Day 13. Tumor volume was determined by caliper measurements (mm) and using the formula for an ellipsoid sphere:
Lx W2/2=mm3, where L and W refer to the larger and smaller perpendicular dimensions collected at each measurement. This formula was also used to calculate tumor weight, assuming unit density (1 mm3= 1 mg).
Comparison of the median tumor weight in the treatment groups (T) to the median tumor weight in the control group (T/C x 100%) on Day 23 (two days after the end of the treatment) and Day 70 (the day of study termination) were used for evaluation of the antitumor efficacy. %T/C for each treatment is reported in Table 19.
Table 19
Schedule vehicle: qld x 9 day 13
Schedule Aplidine: qld x 9 day 13
Schedule Carboplatin: q4d x 3 day 13
Schedule Aplidine/Carboplatin: qld x 9 day 13/ q4d x 3 day 13
The combination treatment of Aplidine plus Carboplatin was tolerated without deaths. The combination treatment resulted in T/C values of 95% and 53% on Days 23 and 70, respectively. Example 8: In vivo studies to determine the effect of Aplidine in combination with another standard agent in myeloma xenografts.
The purpose of this study was to evaluate the antitumor efficacy of Aplidine when administered in combination with bortezomib against subcutaneous-implanted RPMI 8226 human myeloma cells in male, SCID mice.
The animals were housed in microisolator cages, up to five per cage in a 12-hour light/dark cycle. Six- weeks-old male, SCID mice were acclimated in the laboratories for one week prior to the experiment.
Each mouse was inoculated s.c. near the right flank with RPMI 8226 human myeloma cells from an in vitro cell culture using a 23- gauge needle. Each mouse received 2 x 107 cells resuspended in 0.2 mL of Matrigel®. The RPMI 8226 human myeloma cells were originally purchased from ATCC (ATCC number: CCL- 155). A vial with frozen cells was thawed and cultured in RPMI 1640 medium containing high glucose (4,500 mg/L), sodium bicarbonate (1,500 mg/mL), 2 mM L- glutamine, 10 mM Hepes, 1 mM sodium pyruvate, and 10% fetal bovine serum (complete medium), and grown in a +370C incubator in a humidified atmosphere with 5% CO2 until the necessary number of cells for inoculation of mice was obtained. Cells were harvested after four passages in culture. The cells were removed from the flasks, placed in 50-mL centrifuge tubes and centrifuged at 1,000 rpm for 10 minutes in a refrigerated centrifuge. The cell pellets were resuspended in fresh complete medium. The cell count and viability were determined with a Beckman Coulter VI CELL XR cell counter and viability analyzer. The cell suspension was recentrifuged, and the cell pellet was resuspended in Matrigel® at a cell density of 1.0 x 108 cells/mL and placed on wet ice. The final concentration of Matrigel® in the cell suspension was 78.3%. On the day of cell harvest cell viability was 89.3%.
The day of tumor cell inoculation was designated as Day 0. Individual tumors grew to 75-188 mg in weight (75-188 mm3 in size) on the day of treatment initiation, Day 18 after tumor cell inoculation. Those animals selected with tumors in the proper size range were assigned to four treatment groups so that the median tumor weights on the first day of treatment were as close to each other as possible.
The experiment consisted of a vehicle-treated control group of 10 mice and three drug-treated groups of 10 mice per group for a total of 40 mice on the first day of treatment, Day 18 after tumor cell inoculation. Animals in Group 1 were treated i.p. for two rounds every day during 9 consecutive days (Days 18-26 and Days 38-46) with a vehicle of Aplidine diluted with saline. Aplidine was administered i.p. for two rounds every day during 9 consecutive days (Days 18-26 and Days 38-46) at a dosage of 60 μg/kg/dose alone (Group 2) or in combination with bortezomib (Group 4). Bortezomib was administered i.v. at a dosage of 0.35 mg/kg/dose for four weeks every 3 days for a total of 2 treatments starting on Day 18 followed by one more i.v. injection administered on Day 46 alone (Group 3) or in combination with Aplidine (Group 4). On days when both compounds were administered in Group 4, Aplidine was injected first to all ten animals in the group, followed immediately by the administration of bortezomib (Group 4).
Group 1 was treated i.p. with 0.18% cremophor EL/0.18% ethanol/0.84% WFI/98.8% saline (injection volume: 0.1 mL/ 10 g body weight). Aplidine was reconstituted with a vehicle containing 15% cremophor EL/ 15% ethanol/70% WFI and diluted with saline (injection volume: 0.1 mL/ 10 g body weight). Velcade® (Bortezomib) was prepared in saline (injection volume: 0.1 mL/10 g body weight).
Animals were observed daily and clinical signed were noted. The s. c. tumors were measured and the animals were weighed twice weekly starting with the first day of treatment, Day 18 after tumor cell inoculation. Tumor volume was determined by caliper measurements (mm) and using the formula for an ellipsoid sphere as described in Example 7.
Comparison of the median tumor weight in the treatment groups (T) to the median tumor weight in the control group (T/ C x 100%) on Day 27 (one day after the end of the first round of Aplidine treatment) and Day 48 (two days after the end of the treatment with Aplidine and Bortezomib) were used for evaluation of the antitumor efficacy. %T/C for each treatment is reported in Table 20.
Table 20
Figure imgf000069_0001
Schedule vehicle: qld x 9 day 18, 38
Schedule Aplidine: qld x 9 day 18, 38
Schedule Bortezomib: q3d x 2 day 18, 25, 32, 39; qld x 1 day 46
Schedule Aplidine/Bortezomib: qld x 9 day 18, 38/ q3d x 2 day 18, 25, 32,
39; qld x 1 day 46 The combination treatment of Aplidine plus Bortezomib was tolerated without deaths. The combination treatment was effective in the inhibition of the growth of the RPMI 8226 myeloma cells, resulting in T/C values of 49% and 31% on Days 27 and 48, respectively. The antitumor activity of the combination treatment was greater than additive compared to the antitumor activity produced by administration of each compound alone.
Example 9: In vivo studies to determine the effect of Aplidine in combination with another standard agent in lymphoma xenografts.
The purpose of this study was to evaluate the antitumor efficacy of Aplidine when administered in combination with rituximab against subcutaneous-implanted RL human lymphoma cells in female, SCID mice.
The animals were housed in microisolator cages, up to five per cage in a 12-hour light/ dark cycle. Six-weeks-old male, SCID mice were acclimated in the laboratories for one week prior to the experiment.
Each mouse was inoculated s.c. near the right flank with RL human lymphoma cells from an in vitro cell culture using a 23 -gauge needle. Each mouse received 1.0 x 107 cells resuspended in 0.2 mL of Matrigel®. A vial with frozen cells was thawed and cultured in RPMI 1640 medium containing high glucose (4,500 mg/L), sodium bicarbonate (1,500 mg/mL), 2 mM L-glutamine, 10 mM Hepes, 1 mM sodium pyruvate, and 10% fetal bovine serum (complete medium), and grown in a +37°C incubator in a humidified atmosphere with 5% CO2 until the necessary number of cells for inoculation of mice was obtained. Cells were harvested after six passages in culture. The cells were removed from the flasks, placed in 50-mL centrifuge tubes and centrifuged at 1,000 rpm for 10 minutes in a refrigerated centrifuge. The cell pellets were resuspended in fresh complete medium. The cell count was determined with a Coulter Model Zl cell counter and viability was measured following propidium iodide staining and analyzed using a Beckman Coulter EPICS XL flow cytometer. The cell suspension was recentrifuged, and the cell pellet was resuspended in Matrigel® at a cell density of 5.0 x 107 cells/mL and placed on wet ice. The final concentration of Matrigel® in the cell suspension was 73.0%. On the day of cell harvest cell viability was 98.9%.
The day of tumor cell inoculation was designated as Day 0. Individual tumors grew to 100-196 mg in weight (100-196 mm3 in size) on the day of treatment initiation, Day 15 after tumor cell inoculation. Forty animals with tumors in the proper size range were assigned to four treatment groups so that the median tumor weights in all groups on the first day of treatment were as close to each other as possible.
The experiment consisted of a vehicle-treated control group of 10 mice and three drug-treated groups of 10 mice per group for a total of 40 mice on the first day of treatment, Day 15 after tumor cell inoculation. Animals in Group 1 were treated i.p. for two rounds every day during 9 consecutive days (Days 15-23 and Days 28-36) with a vehicle of Aplidine diluted with saline. Aplidine was administered i.p. for two rounds every day during 9 consecutive days (Days 15-23 and Days 28-36) at a dosage of 60 μg/kg/dose alone (Group 2) or in combination with Rituximab (Group 4). Rituximab was administered i.p. at a dosage of 20 mg/kg/dose for four weekly rounds of treatment every 3 days for a total of 2 treatments (q3d x 2 schedule) starting on Day 15 alone (Group 3) or in combination with Aplidine (Group 4). On days when both compounds were administered in Group 4, Aplidine was injected first to all ten animals in the group, followed immediately by the administration of Rituximab (Group 4).
Group 1 was treated i.p. with 0.18% cremophor EL/0.18% ethanol/0.84% WFI/98.8% saline (injection volume: 0.1 mL/ 10 g body weight). Aplidine was reconstituted with a vehicle containing 15% cremophor EL/ 15% ethanol/70% WFI and diluted with saline (injection volume: 0.1 mL/ 10 g body weight). Rituxan® (Rituximab) was prepared in saline (injection volume: 0.1 mL/ 10 g body weight).
Animals were observed daily and clinical signed were noted. The s. c. tumors were measured and the animals were weighed twice weekly starting with the first day of treatment, Day 15. Tumor volume was determined by caliper measurements (mm) and using the formula for an ellipsoid sphere as described in Example 7.
Comparison of the median tumor weight in the treatment groups (T) to the median tumor weight in the control group (T/C x 100%) on Day 24 (one day after the end of the first 9-day round of Aplidine treatment) and Day 38 (two days after the end of the second 9 -day round of Aplidine treatment) were used for evaluation of the antitumor efficacy. %T/C for each treatment is reported in Table 21.
Table 21
Figure imgf000072_0001
Figure imgf000073_0001
Schedule vehicle: qld x 9 day 15, 28
Schedule Aplidine: qld x 9 day 15, 28
Schedule Rituximab: q3d x 2 day 15, 22, 29, 36
Schedule Aplidine/Rituximab: qld x 9 day 15, 28/ q3d x 2 day 15, 22, 29, 36
The combination treatment of Aplidine plus Rituximab was tolerated without deaths. The combination treatment was effective in the inhibition of the growth of the RL lymphoma cells, resulting in T/C values of 69% and 74% on Days 24 and 38, respectively. Therefore, when Aplidine is combined with Rituximab a potentiation of the antitumor activity was observed.
Example 10: In vivo studies to determine the effect of Aplidine in combination with other standard agents (triple combinations) on multiple myeloma tumor cell lines.
In the present study triple combinations of antitumor agents were analyzed. All the combinations were tested using cell viability assay (MMT) in MM. IS cell line, a very sensitive MM cell line. Results were analyzed using the Calcusyn software.
Cell lines and cell culture reagents
The dexamethasone-sensitive MM cell line MM. IS was kindly provided by Dr. S Rudikoff, Bethesda MD). The cell line was grown in RPMI 1640 medium supplemented with 10% heat-inactivated fetal bovine serum, 100 U /ml penicillin, 100 μg/ml streptomycin and 2 mM L-glutamine. All cell culture media and reagents were purchased from Invitrogen Corporation (Carlsbad, CA).
Cell viability assays
The analysis of MM cell proliferation was assessed using the methylthiotetrazole (MTT; Sigma, St. Louis MO) colorimetric assay. MM cell lines were seeded at a density of 50000 cells/200μl medium per well in 48-well plates, and treated with a determined drug dose and time. Two hours before the end of the treatment, a MTT solution (5 mg/ml in PBS; usually a 10% of the volume in each well) was added and the tetrazolium salt was reduced by metabolically active cells to coloured formazan crystals. After solubilization of these crystals by overnight incubation with 10% SDS-HCl solution, absorbance was measured at 570nm with correction at 630nm. Four wells were analyzed for each condition, and the results are presented as the mean ± SD of quadruplicates of a representative experiment that was repeated at least three times.
Isobologram analysis
The interaction between Aplidine and other anti-MM agents was analyzed using the Calcusyn software program (Biosoft, Ferguson, MO). Data from cell viability assay (MTT) were expressed as the fraction of cells affected by the dose (Fa) in drug treated cells as compared to untreated cells (control). This program is based upon the Chou-Talalay method according to the following equation CI = (D) 1/(Dx)I + (D) l(D)2/(Dx)l(Dx)2 where (D) I and (D)2 are the doses of drug 1 and 2 that have the same x effect when used alone.
The computer-calculated combination index (CI) was used to judge the outcomes of a combination: CI>1, CI=I, and CI<1 indicating antagonism, additive, and synergistic effects, respectively. The conformity of data to the median-effect principle can be readily manifested by the linear correlation coefficient (r) of the median- effect plot: log (fa/fu) = m log (D)- m log (Dm), where D is the dose, Dm is the dose required for a 50% effect, fa is the fraction affected by dose, fu is the unaffected fraction , and m is a coefficient of the sigmoidicity of the dose-effect curve. For each combination a non-constant ratio combination was utilized.
Results
Combination of Aplidine + Lenalidomide (Reylimid®) + Dexamethasone The addition of Dexamethasone to the combination of Aplidine + Lenalidomide showed an important synergy in all the combinations tested in the sensitive cell line (MMlS) (Figure 80). In figure 80 Aplidine doses are expressed in nM units, Lenalidomide doses are expressed in μM units and Dexamethasone doses are expressed in nM units.
Combination of Aplidine + Bortezomib + Dexamethasone The addition of Dexamethasone to the combination of Aplidine with Bortezomib resulted in several doses with a clear trend to synergy (Figure 81). In Figure 81 Aplidine doses are expressed in nM units, Bortezomib doses are expressed in nM units and Dexamethasone doses are expressed in nM units.
Combination of Aplidine + Bortezomib + Lenalidomide (Reylimid®) The addition of Bortezomib to the combination of Aplidine with an immunomodulatory agent such as Lenalidomide clearly increased its antitumoral effect with CI in the synergistic range in MMlS (Figure 82). In Figure 82 Aplidine doses are expressed in nM units, Bortezomib doses are expressed in nM units and Lenalidomide doses are expressed in μM units.
Combination of Aplidine + Thalidomide + Dexamethasone This combination showed clear synergy in the triple combinations as can be seen in Figure 83. In Figure 83 Aplidine doses are expressed in nM units, Thalidomide doses are expressed in μM units and Dexamethasone doses are expressed in nM units.
Combination of Aplidin + Melphalan + Dexamethasone This combination showed also a clear synergistic range, mainly with high doses of the drugs (Figure 84). In Figure 84 Aplidine doses are expressed in nM units, Melphalan doses are expressed in μM units and Dexamethasone doses are expressed in nM units.
Combination of Aplidin + Melphalan + Bortezomib
This combination resulted in CI in the synergistic range when using high doses (Figure 85). In Figure 85 Aplidine doses are expressed in nM units, Melphalan doses are expressed in μM units and Bortezomib doses are expressed in nM units.
These findings in respect of Aplidine can be extended to aplidine analogues, derivatives and related compounds. For example, the present invention provides a combination of a compound such as those of WO 02 02596 with an anticancer drug, preferably paclitaxel (Taxol®), doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil (5-FU), cytosine arabinoside (AraC), carboplatin, 7-ethyl-lO-hydroxycamptothecin (SN38), etoposide (VP 16), melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide (Revlimid®), interleukin-2 (IL-2), interferon-α 2 (INF- α), dacarbazine (DTIC), bevacizumab (Avastin®), idarubicin, thalidomide, and rituximab. Examples of analogues of aplidine which can be used in place of Aplidine itself include the preferred compounds given in WO 02 02596, and in particular we import into this patent specification the discussion of preferred compounds and related aspects given in WO 02 02596. More preferably, the analogues are structurally close to Aplidine, and usually differ from Aplidine in respect of one amino acid or the terminal sidechain. The different amino acid can be in the cyclic part of the molecule or in the sidechain. Many examples of such compounds are given in WO 02 02596, and they are candidates for use in the present invention.

Claims

Claims
1. Use of Aplidine or an aplidine analogue in the manufacture of a medicament for the treatment of cancer by combination therapy employing Aplidine or an aplidine analogue with another drug.
2. The use of Aplidine or an aplidine analogue according to claim 1, wherein the other drug is effective in the treatment of a cancer selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia, and lymphoma.
3. The use of Aplidine or an aplidine analogue according to any of the preceding claims, wherein the Aplidine or the aplidine analogue and the other drug form part of the same composition.
4. The use of Aplidine or an aplidine analogue according to claim 1 or 2, wherein the Aplidine or the aplidine analogue and the other drug are provided as separate compositions for administration at the same time or at different times.
5. The use of Aplidine or an aplidine analogue according to any of the preceding claims, wherein the other drug is selected from the group consisting of paclitaxel, doxorubicin, cisplatin, arsenic trioxide, 5- fluorouracil, cytosine arabinoside, carboplatin, 7-ethyl-10- hydroxycamptothecin, etoposide, melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide, interleukin-2, interferon-α 2, dacarbazine, bevacizumab, idarubicin, thalidomide, and rituximab.
6. The use of Aplidine or an aplidine analogue according to any of the preceding claims, wherein the Aplidine or the aplidine analogue is Aplidine.
7. The use of Aplidine or an aplidine analogue according to any of the preceding claims, wherein a third drug is administered.
8. The use of Aplidine or an aplidine analogue according to claim 7, wherein the second and the third drug are selected from the group consisting of paclitaxel, doxorubicin, cisplatin, arsenic trioxide, 5- fluorouracil, cytosine arabinoside, carboplatin, 7-ethyl-10- hydroxycamptothecin, etoposide, melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide, interleukin-2, interferon-α 2, dacarbazine, bevacizumab, idarubicin, thalidomide, and rituximab.
9. A method of treating cancer comprising administering to a patient in need of such treatment a therapeutically effective amount of Aplidine or an aplidine analogue and a therapeutically effective amount of another drug which is effective in the treatment of cancer.
10. The method of treating cancer according to claim 9, wherein the cancer to be treated is selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia, and lymphoma.
11. The method of treating cancer according to claim 9 or 10, wherein the other drug is selected from the group consisting of paclitaxel, doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil, cytosine arabinoside, carboplatin, 7-ethyl-lO-hydroxycamptothecin, etoposide, melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide, interleukin-2, interferon-α 2, dacarbazlne, bevacizumab, idarubicin, thalidomide, and ritnximab.
12. A method of increasing the therapeutic efficacy of a drug effective in the treatment of cancer, which comprises administering to a patient in need thereof an amount of Aplidine or an aplidine analogue.
13. The method of increasing the therapeutic efficacy of a drug effective in the treatment of cancer according to claim 12, wherein the cancer to be treated is selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia, and lymphoma.
14. The method of increasing the therapeutic efficacy of a drug effective in the treatment of cancer according to claim 12 or 13, wherein the drug is selected from the group consisting of paclitaxel, doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil, cytosine arabinoside, carboplatin, 7-ethyl-lO-hydroxycamptothecin, etoposide, melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide, interleukin-2, interferon-α 2, dacarbazine, bevacizumab, idarubicin, thalidomide, and rituximab.
15. The method according to any of claims 9 to 14, wherein the Aplidine or the aplidine analogue and the other drug form part of the same composition.
16. The method according to any of claims 9 to 14, wherein the Aplidine or the aplidine analogue and the other drug are provided as separate compositions for administration at the same time or at different times.
17. The method according to any of claims 9 to 16, wherein the Aplidine or the aplidine analogue is Aplidine.
18. The method according to any of claims 9 or 17, wherein a therapeutically effective amount of a third drug is administered.
19. The method according to claim 18, wherein the second and third drug are selected from the group consisting of paclitaxel, doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil, cytosine arabinoside, carboplatin, 7-ethyl-lO-hydroxycamptothecin, etoposide, melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide, interleukin-2, interferon-α 2, dacarbazine, bevacizumab, idarubicin, thalidomide, and rituximab.
20. A kit for use in the treatment or prevention of cancer which comprises a dosage form of Aplidine or an aplidine analogue, a dosage form of another drug effective in the treatment of cancer and instructions for the use of each actor in combination for the treatment or prevention of cancer.
21. The kit according to claim 20, wherein the kit is for use in the treatment or prevention of a cancer selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia, and lymphoma.
22. The kit according to claim 20 or 21, wherein the other drug is selected from the group consisting of paclitaxel, doxorubicin, cisplatin, arsenic trioxide, 5-fiuorouracil, cytosine arabinoside, carboplatin, 7- ethyl-10-hydroxycamptothecin, etoposide, melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide, interleukin-2, interferon-α 2, dacaxbazine, bevacizumab, idarubicin, thalidomide, and rituximab.
23. The kit according to claim 20 or 21, which additionally comprises a dosage form of a third drug effective in the treatment of cancer.
24. The kit according to claim 23, wherein the second and third drug are selected from the group consisting of paclitaxel, doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil, cytosine arabinoside, carboplatin, 7-ethyl-lO-hydroxycamptothecin, etoposide, melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide, interleukin-2, interferon-α 2, dacarbazine, bevacizumab, idarubicin, thalidomide, and rituximab.
25. A pharmaceutical composition comprising Aplidine or an aplidine analogue and another drug effective in the treatment of cancer.
26. The pharmaceutical composition according to claim 25, wherein the other drug is effective in the treatment of a cancer selected from lung cancer, breast cancer, colon cancer, prostate cancer, renal cancer, melanoma, multiple myeloma, leukemia, and lymphoma.
27. The pharmaceutical composition according to claim 25 or 26, wherein the other drug is selected from the group consisting of paclitaxel, doxorubicin, cisplatin, arsenic trioxide, 5-fluorouracil, cytosine arabinoside, carboplatin, 7-ethyl-10-hydroxycamptothecin, etoposide, melphalan, dexamethasone, cyclophosphamide, bortezomib, erlotinib, trastuzumab, lenalidomide, interleukin-2, interferon-α 2, dacarbazine, bevacizumab, idarubicin, thalidomide, and rituximab.
PCT/US2007/062936 2006-02-28 2007-02-28 Improved antitumoral treatments WO2007101235A2 (en)

Priority Applications (21)

Application Number Priority Date Filing Date Title
RS20160545A RS54928B1 (en) 2006-02-28 2007-02-28 Improved treatment of multiple myeloma
DK07757603.1T DK2029155T3 (en) 2006-02-28 2007-02-28 IMPROVED treatment of multiple myeloma
CN2007800068661A CN101389347B (en) 2006-02-28 2007-02-28 Improved antitumoral treatments
AU2007220050A AU2007220050B2 (en) 2006-02-28 2007-02-28 Improved antitumoral treatments
ES07757603.1T ES2575518T3 (en) 2006-02-28 2007-02-28 Improved treatment of multiple myeloma
KR1020087023662A KR101512503B1 (en) 2006-02-28 2007-02-28 Improved antitumoral treatments
NZ571043A NZ571043A (en) 2006-02-28 2007-02-28 Anticancer combination of aplidine and dexamethasone
KR1020147021341A KR20140101014A (en) 2006-02-28 2007-02-28 Improved antitumoral treatments
MEP-2016-115A ME02450B (en) 2006-02-28 2007-02-28 Improved antitumoral treatments
EP07757603.1A EP2029155B1 (en) 2006-02-28 2007-02-28 Improved treatment of multiple myeloma
SI200731777A SI2029155T1 (en) 2006-02-28 2007-02-28 Improved treatment of multiple myeloma
RU2008138560/15A RU2481853C2 (en) 2006-02-28 2007-02-28 Improved methods for treating tumours
CA2643238A CA2643238C (en) 2006-02-28 2007-02-28 Improved antitumoral treatments
JP2008557477A JP5393159B2 (en) 2006-02-28 2007-02-28 Improved anti-tumor treatment
US12/278,559 US8258098B2 (en) 2006-02-28 2007-02-28 Antitumoral treatments
MX2008010999A MX2008010999A (en) 2006-02-28 2007-02-28 Improved antitumoral treatments.
IL193094A IL193094A (en) 2006-02-28 2008-07-28 Use of aplidine and another drug in the manufacture of a medicament for the treatment of cancer wherein the combination of both drugs provides a synergistic effect
NO20084047A NO342012B1 (en) 2006-02-28 2008-09-23 Improved antitumoral treatments
HK09106467.2A HK1127294A1 (en) 2006-02-28 2009-07-16 Improved treatment of multiple myeloma
HRP20160646TT HRP20160646T1 (en) 2006-02-28 2016-06-10 Improved treatment of multiple myeloma
CY20161100620T CY1117722T1 (en) 2006-02-28 2016-07-05 IMPROVED EDUCATION OF MULTIPLE MYELOMA

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US81360606P 2006-02-28 2006-02-28
US60/813,606 2006-02-28

Publications (2)

Publication Number Publication Date
WO2007101235A2 true WO2007101235A2 (en) 2007-09-07
WO2007101235A3 WO2007101235A3 (en) 2008-04-03

Family

ID=38459806

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/062936 WO2007101235A2 (en) 2006-02-28 2007-02-28 Improved antitumoral treatments

Country Status (24)

Country Link
US (1) US8258098B2 (en)
EP (1) EP2029155B1 (en)
JP (2) JP5393159B2 (en)
KR (2) KR101512503B1 (en)
CN (1) CN101389347B (en)
AU (1) AU2007220050B2 (en)
CA (1) CA2643238C (en)
CY (1) CY1117722T1 (en)
DK (1) DK2029155T3 (en)
ES (1) ES2575518T3 (en)
HK (1) HK1127294A1 (en)
HR (1) HRP20160646T1 (en)
HU (1) HUE028055T2 (en)
IL (1) IL193094A (en)
ME (1) ME02450B (en)
MX (1) MX2008010999A (en)
NO (1) NO342012B1 (en)
NZ (1) NZ571043A (en)
PL (1) PL2029155T3 (en)
PT (1) PT2029155E (en)
RS (1) RS54928B1 (en)
RU (1) RU2481853C2 (en)
SI (1) SI2029155T1 (en)
WO (1) WO2007101235A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008135793A1 (en) * 2007-05-04 2008-11-13 Pharma Mar S.A. Combination of aplidine and carboplatin in anticancer treatments
WO2009111698A1 (en) * 2008-03-07 2009-09-11 Pharma Mar, S.A. Improved anticancer treatments
US20110015135A1 (en) * 2008-03-07 2011-01-20 Pharma Mar S.A. Antitumoral Treatments
US8258098B2 (en) 2006-02-28 2012-09-04 Pharma Mar, S.A. Antitumoral treatments
US20140296178A1 (en) * 2010-12-01 2014-10-02 Niiki Pharma Acquisition Corp. 2 Combination Therapy with a Gallium Complex
EP2851070A1 (en) * 2010-01-05 2015-03-25 Celgene Corporation A combination of lenalidomide and artesunate/artemisone for treating cancer
US10428151B2 (en) 2011-11-09 2019-10-01 Bristol-Myers Squibb Company Treatment of hematologic malignancies with an anti-CXCR4 antibody
US10538535B2 (en) 2017-04-27 2020-01-21 Pharma Mar, S.A. Antitumoral compounds
EP3895733A4 (en) * 2018-12-14 2022-03-02 Konica Minolta, Inc. Method for forecasting arrival of drug inside diseased tissue

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE363910T1 (en) * 1999-11-15 2007-06-15 Pharma Mar Sa TREATMENT OF CANCER DISEASES BY APLIDINE
AU2008313627A1 (en) * 2007-10-19 2009-04-23 Pharma Mar, S.A. Improved antitumoral treatments
BR122017028570B1 (en) * 2010-11-12 2022-03-03 Pharma Mar, S.A USE OF PM01183, OR A PHARMACEUTICALLY ACCEPTABLE SALT THEREOF, IN SYNERGIC COMBINATION WITH A TOPOISOMERASE I AND/OR II INHIBITOR AND KIT
KR101369936B1 (en) * 2011-12-28 2014-03-06 연세대학교 산학협력단 Pharmaceutical composition for treating lupus nephritis comprising thalidomide
RU2547999C1 (en) * 2013-10-28 2015-04-10 Государственное бюджетное учреждение здравоохранения Московской области "Московский областной научно-исследовательский клинический институт им. М.Ф. Владимирского" (ГБУЗ МО МОНИКИ им. М.Ф. Владимирского) Method of treating adenogenous locally advanced cancer of lower ampullary part of rectum
CN113018415B (en) * 2021-03-17 2022-07-01 遵义医科大学 Medicine composition and application thereof

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991004985A1 (en) 1989-09-29 1991-04-18 Pharma Mar S.A. Dehydrodidemnin b
WO1999042125A1 (en) 1998-02-18 1999-08-26 Pharma Mar, S.A. Pharmaceutical formulation of a didemnin compound
WO2001035974A2 (en) 1999-11-15 2001-05-25 Pharma Mar S.A. Aplidine treatment of cancers
WO2001076616A1 (en) 2000-04-07 2001-10-18 The Trustees Of The University Of Pennsylvania Tamandarin and didemnin analogs and methods of making and using them
WO2002002596A2 (en) 2000-06-30 2002-01-10 Pharma Mar, S.A. Synthetic methods for aplidine and new antitumoral derivatives, methods of making and using them
WO2002030441A2 (en) 2000-10-12 2002-04-18 Pharma Mar, S.A. Treatment of cancers by aplidine in conjunction with a myoprotector
WO2003033013A1 (en) 2001-10-19 2003-04-24 Pharma Mar, S.A. Use of aplidine for the treatment of pancreatic cancer
WO2004080477A1 (en) 2003-03-12 2004-09-23 Dana-Farber Cancer Institute, Inc. Aplidine for multiple myeloma treatment
WO2004080421A2 (en) 2003-03-12 2004-09-23 Pharma Mar, S.A. Improved antitumoral treatments

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4493796A (en) * 1980-09-12 1985-01-15 Board Of Trustees, Univ. Of Ill. Didemnins A, B, C, and derivatives thereof, as antiviral agents
US4950649A (en) * 1980-09-12 1990-08-21 University Of Illinois Didemnins and nordidemnins
IT1153974B (en) * 1982-09-23 1987-01-21 Erba Farmitalia PHARMACOLOGICAL COMPOSITIONS BASED ON CISPLATIN AND METHOD FOR THEIR OBTAINMENT
ATE74761T1 (en) * 1985-09-20 1992-05-15 Cernitin Sa USE OF PLANT POLLEN EXTRACTS FOR THE MANUFACTURE OF TUMOR CELL GROWTH INHIBITING PHARMACEUTICAL PREPARATIONS AND PROCESS FOR THEIR MANUFACTURE.
US4948791A (en) 1989-04-10 1990-08-14 The Board Of Trustees Of The University Of Illinois Novel Cytotoxic cyclic depsipeptides from the tunicate trididemnum solidum
US20030148933A1 (en) * 1990-10-01 2003-08-07 Pharma Mar S.A. Derivatives of dehydrodidemnin B
DE4120327A1 (en) 1991-06-20 1992-12-24 Basf Ag NEW PEPTIDES, THEIR PREPARATION AND USE
US5580871A (en) * 1992-11-20 1996-12-03 The Dupont Merck Pharmaceutical Company 4-Heteroaryl- 1,4-dihydropyridine compounds with calcium agonist and alpha1 -antagonist activity
US5462726A (en) * 1993-12-17 1995-10-31 Bristol-Myers Squibb Company Method of inhibiting side effects of solvents containing ricinoleic acid or castor oil or derivatives thereof employing a thromboxane A2 receptor antagonist and pharmaceutical compositions containing such solvents
US6156724A (en) * 1996-06-07 2000-12-05 Rinehart; Kenneth L. Uses of didemnins as immunomodulating agents
JP2001503746A (en) 1996-10-24 2001-03-21 ザ・ボード・オブ・トラステイーズ・オブ・ザ・ユニバーシテイ・オブ・イリノイ Semisynthetic method for didemnin analogues
DE69723728T2 (en) 1996-10-24 2004-06-03 The Board Of Trustees For The University Of Illinois, Urbana TOTAL SYNTHESIS OF THE AMINO HIP ANALOG OF DIDEMNIN A
US6034058A (en) * 1997-04-15 2000-03-07 Rinehart; Kenneth L. Semi-synthetic alanyl dilemnin analogs
DE69840497D1 (en) * 1997-05-07 2009-03-12 William J Crumb Use of aplidine for the treatment of cardiovascular diseases
GB9821975D0 (en) 1998-10-08 1998-12-02 Pharma Mar Sa New cytotoxic alkaloids
US6245759B1 (en) * 1999-03-11 2001-06-12 Merck & Co., Inc. Tyrosine kinase inhibitors
RU2266734C2 (en) * 1999-05-13 2005-12-27 Фарма Мар, С.А. Composition and utilization of et743 for malignant tumor treatment
US6635677B2 (en) * 1999-08-13 2003-10-21 Case Western Reserve University Methoxyamine combinations in the treatment of cancer
US6509315B1 (en) * 2000-04-07 2003-01-21 The Trustees Of The University Of Pennsylvania Didemnin analogs and fragments and methods of making and using them
BR0114604A (en) * 2000-10-12 2003-10-14 Pharma Mar Sa Cancer treatment
CA2435418A1 (en) 2001-01-24 2002-08-01 Mestex Ag Use of neurotoxic substances in producing a medicament for treating joint pains
WO2004084812A2 (en) 2003-03-21 2004-10-07 Joullie Madeleine M Tamandarin analogs and fragments thereof and methods of making and using
JP2007516693A (en) * 2003-06-09 2007-06-28 ザ・リージェンツ・オブ・ザ・ユニバーシティ・オブ・ミシガン Compositions and methods for the treatment and diagnosis of cancer
ES2575518T3 (en) 2006-02-28 2016-06-29 Pharma Mar S.A. Improved treatment of multiple myeloma
DE102006061344A1 (en) * 2006-12-22 2008-06-26 Robert Bosch Gmbh Clutch hydraulic circuit
WO2008080956A1 (en) 2006-12-29 2008-07-10 Pharma Mar, S.A. Prognostic molecular markers for the cancer therapy with aplidine
WO2008135793A1 (en) 2007-05-04 2008-11-13 Pharma Mar S.A. Combination of aplidine and carboplatin in anticancer treatments
AU2008313627A1 (en) * 2007-10-19 2009-04-23 Pharma Mar, S.A. Improved antitumoral treatments
WO2010029158A1 (en) 2008-09-12 2010-03-18 Pharma Mar, S.A. Aplidine in the treatment of chronic myeloproliferative disorders

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991004985A1 (en) 1989-09-29 1991-04-18 Pharma Mar S.A. Dehydrodidemnin b
WO1999042125A1 (en) 1998-02-18 1999-08-26 Pharma Mar, S.A. Pharmaceutical formulation of a didemnin compound
WO2001035974A2 (en) 1999-11-15 2001-05-25 Pharma Mar S.A. Aplidine treatment of cancers
WO2001076616A1 (en) 2000-04-07 2001-10-18 The Trustees Of The University Of Pennsylvania Tamandarin and didemnin analogs and methods of making and using them
WO2002002596A2 (en) 2000-06-30 2002-01-10 Pharma Mar, S.A. Synthetic methods for aplidine and new antitumoral derivatives, methods of making and using them
WO2002030441A2 (en) 2000-10-12 2002-04-18 Pharma Mar, S.A. Treatment of cancers by aplidine in conjunction with a myoprotector
WO2003033013A1 (en) 2001-10-19 2003-04-24 Pharma Mar, S.A. Use of aplidine for the treatment of pancreatic cancer
WO2004080477A1 (en) 2003-03-12 2004-09-23 Dana-Farber Cancer Institute, Inc. Aplidine for multiple myeloma treatment
WO2004080421A2 (en) 2003-03-12 2004-09-23 Pharma Mar, S.A. Improved antitumoral treatments

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
BROGGINI M; MARCHINI S; D'INCALCI M; TARABOLETTI G; GIAVAZZI R; FAIRCLOTH G; JIMENO J.: "Aplidine blocks VEGF secretion and VEGF/VEGF-R1 autocrine loop in a human leukemic cell line", CLIN. CANCER RES., vol. 6, 2000, pages 4509
CHOU TC ET AL., ADV. ENZYME REGUL., vol. 22, 1984, pages 27 - 55
DEPENBROCK H; PETER R; FAIRCLOTH GT; MANZANARES I; JIMENO J; HANAUSKE AR: "In vitro activity of Aplidine, a new marine-derived anticancer compound, on freshly explanted clonogenic human tumour cells and haematopoietic precursor cells", BR. J. CANCER, vol. 78, 1998, pages 739 - 744, XP001002515
ERBA E; BASSANO L; DI LIBERTI G; MURADORE I; CHIORINO G; UBEZIO P; VIGNATI S; CODEGONI A; DESIDERIO MA; FAIRCLOTH G: "Cell cycle phase perturbations and apoptosis in tumour cells induced by aplidine", BR. J. CANCER, vol. 86, 2002, pages 1510 - 1517
FAIRCLOTH G; GRANT W; NAM S; JIMENO J; MANZANARES I; RINEHART K.: "Schedule-dependency of Aplidine, a marine depsipeptide with antitumor activity", PROC. AM. ASSOC. CANCER RES., vol. 40, 1999, pages 394, XP001002421
FAIRCLOTH, G. ET AL.: "Dehydrodidemnin B (DDB) a new marine derived anticancer agent with activity against experimental tumour models", 9TH NCI-EORTC SYMP. NEW DRUGS CANCER THER., 12 March 1996 (1996-03-12)
FAIRCLOTH, G. ET AL.: "Preclinical characterization of aplidine, a new marine anticancer depsipeptide", PROC. AMER. ASSOC. CANCER RES., 1997, pages 38
GRECO ET AL., PHARMACOL REV., vol. 47, 1995, pages 331 - 385
GREENSTEIN S. ET AL., EXP. HEMATOL, vol. 31, 2003, pages 271 - 82
IZQUIERDO MA; BOWMAN A; MARTINEZ M; CICCHELLA B; JIMENO J; GUZMAN C; GERMA J; SMYTH J.: "Phase I trial of Aplidine given as a 1 hour intravenous weekly infusion in patients with advanced solid tumors and lymphoma", CLIN. CANCER RES., vol. 6, 2000, pages 4509
LASKA E. ET AL., BIOMETRICS, vol. 50, 1994, pages 834 - 841
MAROUN J; BELANGER K; SEYMOUR L; SOULIERES D; CHARPENTIER D; GOEL R; STEWART D; TOMIAK E; JIMENO J; MATTHEWS S.: "Phase I study of aplidine in a 5 day bolus q 3 weeks in patients with solid tumors and lymphomas", CLIN. CANCER RES., vol. 6, 2000, pages 4509
PAZ-ARES L; ANTHONY A; PRONK L; TWELVES C; ALONSO S; CORTES-FUNES H; CELLI N; GOMEZ C; LOPEZ-LAZARO L; GUZMAN C: "Phase I clinical and pharmacokinetic study of aplidine, a new marine didemnin, administered as 24-hour infusion weekly", CLIN. CANCER RES., vol. 6, 2000, pages 4509
RAYMOND E; ADY-VAGO N; BAUDIN E; RIBRAG V; FAIVRE S; LECOT F; WRIGHT T; LOPEZ LAZARO L; GUZMAN C; JIMENO J: "A phase I and pharmacokinetic study of aplidine given as a 24-hour continuous infusion every other week in patients with solid tumor and lymphoma", CLIN. CANCER RES., vol. 6, 2000, pages 4510
SAN MIGUEL JF ET AL., CURR. TREAT. OPTIONS ONCOL., vol. 4, 2003, pages 247 - 58
See also references of EP2029155A4
SIROHI B. ET AL., LANCET, vol. 363, 2004, pages 875 - 87

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8258098B2 (en) 2006-02-28 2012-09-04 Pharma Mar, S.A. Antitumoral treatments
WO2008135793A1 (en) * 2007-05-04 2008-11-13 Pharma Mar S.A. Combination of aplidine and carboplatin in anticancer treatments
WO2009111698A1 (en) * 2008-03-07 2009-09-11 Pharma Mar, S.A. Improved anticancer treatments
US20110015135A1 (en) * 2008-03-07 2011-01-20 Pharma Mar S.A. Antitumoral Treatments
US9226913B2 (en) 2010-01-05 2016-01-05 Celgene Corporation Methods of treating cancer using a combination of an immunomodulatory compound and an artemisinin or a derivative thereof
EP2851070A1 (en) * 2010-01-05 2015-03-25 Celgene Corporation A combination of lenalidomide and artesunate/artemisone for treating cancer
EP2521543B1 (en) * 2010-01-05 2016-04-13 Celgene Corporation A combination of an immunomodulatory compound and an artemisinin derivative for treating cancer
US20140296178A1 (en) * 2010-12-01 2014-10-02 Niiki Pharma Acquisition Corp. 2 Combination Therapy with a Gallium Complex
US10428151B2 (en) 2011-11-09 2019-10-01 Bristol-Myers Squibb Company Treatment of hematologic malignancies with an anti-CXCR4 antibody
US10538535B2 (en) 2017-04-27 2020-01-21 Pharma Mar, S.A. Antitumoral compounds
US11332480B2 (en) 2017-04-27 2022-05-17 Pharma Mar, S.A. Antitumoral compounds
US11339180B2 (en) 2017-04-27 2022-05-24 Pharma Mar, S.A. Antitumoral compounds
US11713325B2 (en) 2017-04-27 2023-08-01 Pharma Mar, S.A. Antitumoral compounds
EP3895733A4 (en) * 2018-12-14 2022-03-02 Konica Minolta, Inc. Method for forecasting arrival of drug inside diseased tissue
JP7396578B2 (en) 2018-12-14 2023-12-12 コニカミノルタ株式会社 How to predict drug arrival into diseased tissue

Also Published As

Publication number Publication date
US20090246168A1 (en) 2009-10-01
KR20080105123A (en) 2008-12-03
NO20084047L (en) 2008-11-25
HK1127294A1 (en) 2009-09-25
CN101389347A (en) 2009-03-18
KR101512503B1 (en) 2015-04-15
RS54928B1 (en) 2016-10-31
JP5393159B2 (en) 2014-01-22
US8258098B2 (en) 2012-09-04
RU2008138560A (en) 2010-04-10
CA2643238A1 (en) 2007-09-07
EP2029155A2 (en) 2009-03-04
RU2481853C2 (en) 2013-05-20
CN101389347B (en) 2013-03-27
JP2009528379A (en) 2009-08-06
PT2029155E (en) 2016-06-17
JP2013199500A (en) 2013-10-03
EP2029155B1 (en) 2016-04-13
KR20140101014A (en) 2014-08-18
HUE028055T2 (en) 2016-11-28
DK2029155T3 (en) 2016-08-01
PL2029155T3 (en) 2016-09-30
ES2575518T3 (en) 2016-06-29
SI2029155T1 (en) 2016-06-30
CA2643238C (en) 2016-05-03
WO2007101235A3 (en) 2008-04-03
CY1117722T1 (en) 2017-05-17
NO342012B1 (en) 2018-03-12
IL193094A0 (en) 2011-08-01
MX2008010999A (en) 2008-09-08
ME02450B (en) 2016-09-20
HRP20160646T1 (en) 2016-08-12
AU2007220050A1 (en) 2007-09-07
EP2029155A4 (en) 2009-10-14
AU2007220050B2 (en) 2013-08-29
IL193094A (en) 2015-06-30
NZ571043A (en) 2012-06-29

Similar Documents

Publication Publication Date Title
DK2029155T3 (en) IMPROVED treatment of multiple myeloma
US20090227490A1 (en) Antitumoral Treatments
US20110015135A1 (en) Antitumoral Treatments
KR101978108B1 (en) New anti-malignant tumor agents based on the metabolic specificity of cancer cells
US20100240595A1 (en) Improved Antitumoral Treatments
KR101847252B1 (en) Antitumor agent including irinotecan hydrochloride hydrate
WO2008135793A1 (en) Combination of aplidine and carboplatin in anticancer treatments
Faircloth et al. Improved antitumoral treatments

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 193094

Country of ref document: IL

Ref document number: 2007220050

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2007220050

Country of ref document: AU

Date of ref document: 20070228

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2643238

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/010999

Country of ref document: MX

Ref document number: 2008557477

Country of ref document: JP

Ref document number: 200780006866.1

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 571043

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 3650/KOLNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2007757603

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020087023662

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2008138560

Country of ref document: RU

Kind code of ref document: A

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07757603

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 12278559

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: P-2016/0545

Country of ref document: RS