NZ788180A - Combination therapy with notch and pd-1 or pd-l1 inhibitors - Google Patents

Combination therapy with notch and pd-1 or pd-l1 inhibitors

Info

Publication number
NZ788180A
NZ788180A NZ788180A NZ78818017A NZ788180A NZ 788180 A NZ788180 A NZ 788180A NZ 788180 A NZ788180 A NZ 788180A NZ 78818017 A NZ78818017 A NZ 78818017A NZ 788180 A NZ788180 A NZ 788180A
Authority
NZ
New Zealand
Prior art keywords
cancer
compound
carcinoma
oxo
patient
Prior art date
Application number
NZ788180A
Inventor
Mark Harrath Bender
Hong Gao
Bharvin Kumar Patel
Original Assignee
Eli Lilly And Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eli Lilly And Company filed Critical Eli Lilly And Company
Publication of NZ788180A publication Critical patent/NZ788180A/en

Links

Abstract

The present invention provides medicaments for use in treating and methods of treating T- cell acute lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, erythroleukemia, triple negative breast cancer, breast cancer, ovarian cancer, melanoma, Sung cancer, non small-cell lung cancer, pancreatic cancer, glioblastoma, colorectal cancer, head and neck cancer, cervical cancer, prostate cancer, liver cancer, oral squamous cell carcinoma, skin cancer, medul!ob!astoma, hepatocellular carcinoma, intrahepatic and extrahepatic cholangiocarcinoma, desmoid tumor, soft tissue sarcoma, or adenoid cystic carcinoma in a patient comprising combination therapy with 4,4,4-trifluoro-N-[(1S)-2~[[(7S)-5-(2-hydroxyemyl)-6-oxo-7H-pyrido[23-d][3]benzazepin-7-yl]amino]-1-methyl-2-oxo- ethyljbutanamide, or a pharmaceutically acceptable salt or hydrate thereof, and a PD-1 or a PD-L1 inhibitor selected from pembrolizumab, nivolumab, atezolizumab, durvalumab, and avelumab. cancer, ovarian cancer, melanoma, Sung cancer, non small-cell lung cancer, pancreatic cancer, glioblastoma, colorectal cancer, head and neck cancer, cervical cancer, prostate cancer, liver cancer, oral squamous cell carcinoma, skin cancer, medul!ob!astoma, hepatocellular carcinoma, intrahepatic and extrahepatic cholangiocarcinoma, desmoid tumor, soft tissue sarcoma, or adenoid cystic carcinoma in a patient comprising combination therapy with 4,4,4-trifluoro-N-[(1S)-2~[[(7S)-5-(2-hydroxyemyl)-6-oxo-7H-pyrido[23-d][3]benzazepin-7-yl]amino]-1-methyl-2-oxo- ethyljbutanamide, or a pharmaceutically acceptable salt or hydrate thereof, and a PD-1 or a PD-L1 inhibitor selected from pembrolizumab, nivolumab, atezolizumab, durvalumab, and avelumab.

Description

COMBINATION THERAPY WITH NOTCH AND PD-1 OR PD-L1 INHIBITORS This ation is a divisional of New Zealand patent application 748631, which is the national phase entry in New Zealand of PCT international application 2017/032790 shed as which is incorporated herein by reference.
The present ion relates to cancer therapy with 4,4,4-trifluoro-N-[(1S)- 2-[[(7S)(2-hydroxyethyl)oxo-7H-pyrido[2,3-d][3]benzazepinyl]amino]- 1-methyloxo-ethyl]butanamide, or a pharmaceutically acceptable salt or hydrate thereof (Compound A) and a Programmed Death Receptor 1 (PD-1) inhibitor, or a mmed Death Receptor Ligand 1 (PD-L1) inhibitor and to methods of using combinations to treat cancer. 4,4,4-trifluoro-N-[(1S)[[(7S)(2-hydroxyethyl)oxo-7H-pyrido[2,3- d][3]benzazepinyl]amino]methyloxo-ethyl]butanamide, or a pharmaceutically acceptable salt or hydrate thereof, is a Notch pathway signaling inhibitor compound.
Notch signaling plays an important role during development and tissue homeostasis.
Dysregulation of Notch signaling due to mutation, ication, or pression of ligands and/or receptors, is implicated in a number of malignancies. Inhibition of Notch signaling is a potential target for the development of cancer therapeutics. nd A and methods of making and using this compound, including for the treatment of T-cell acute lymphoblastic leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, erythroleukemia, breast cancer, ovarian , melanoma, lung , pancreatic cancer, glioblastoma, colorectal cancer, head and neck cancer, cervical cancer, prostate cancer, liver cancer, squamous cell carcinoma (oral), skin cancer and medulloblastoma are disclosed in leiomyosarcoma in 2016/026119. Compound A is being investigated in a phase 1 al trial and expansion cohorts having a defined molecular pathway alteration, or a tissue based ant tumor, and in combination with other specifically identified anticancer agents against specified tumor types showing mutations, amplification, or gene expression alterations related to Notch pathway signaling, and in a clinical trial in ts with T-cell acute lymphoblastic leukemia or T-cell lymphoblastic lymphoma (TALL /T-LBL).
Tumor cells escape detection and elimination by the immune system through various isms. Endogenously, immune oint pathways are used in nance of self-tolerance and control of T cell activation. Binding of the PD-1 ligands, PD-L1 and PD-L2, to the PD-1 receptor found on T cells, inhibits T cell proliferation and ne production. Upregulation of PD-1 ligands occurs in some tumors and signaling through this pathway contributes to inhibition of active T-cell immune surveillance of tumors. Inhibition of PD-1 or PD-L1, has been shown to restore immune mediated destruction of tumors. Clinical research has found that ing PD- 1or PD-L1 with antagonist antibodies releases the PD-1 pathway mediated inhibition of the immune response, including the anti-tumor response.
Notch pathway signaling is ed to be a regulator of PD-1 expression by activated CD8+ T cells, Mathieu et al, Immunology and Cell Biology, 2013, 91: 82-88.
Despite existing treatment s for patients with cancer, there continues to be a need for new and different therapies affording one or both of enhanced efficacy and lower toxicity. Current therapies have shown a t in a subset of cancer types and only in a subset of patients. Novel therapies or combination strategies are needed to improve the overall response against specific cancers or to facilitate extension of these treatments into cancers that may currently be less responsive to either agent alone.
It is believed the present invention provides cial therapeutic effects from the combined ty of Compound A and anti-PD-1 or PD-L1 monoclonal antibody inhibitor activity against T-cell acute lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, erythroleukemia, triple negative breast cancer, breast cancer, ovarian cancer, melanoma, lung cancer, non small-cell lung cancer, pancreatic cancer, astoma, colorectal cancer, head and neck cancer, cervical cancer, prostate , liver cancer, oral squamous cell carcinoma, skin cancer, medulloblastoma, hepatocellular carcinoma, intrahepatic and extrahepatic cholangiocarcinoma, desmoid tumors, soft tissue sarcoma, and adenoid cystic carcinoma as compared to the therapeutic effects provided by either agent alone.
One aspect of the present ion provides a method of treating T-cell acute lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, acute myelogenous leukemia, chronic enous leukemia, erythroleukemia, triple negative breast cancer, breast cancer, ovarian , ma, lung cancer, non small cell lung cancer, pancreatic , glioblastoma, colorectal cancer, head and neck cancer, cervical cancer, prostate cancer, liver cancer, oral squamous cell carcinoma, skin cancer, medulloblastoma, hepatocellular carcinoma, intrahepatic and extrahepatic cholangiocarcinoma, desmoid tumor, soft tissue a, or adenoid cystic oma in a patient comprising administering to a t in need of treatment an effective amount of 4,4,4-trifluoro-N-[(1S)[[(7S)(2-hydroxyethyl)oxo-7H-pyrido[2,3- d][3]benzazepinyl]amino]methyloxo-ethyl]butanamide, or a pharmaceutically able salt or hydrate thereof, and an ive amount of a PD-1 or PD-L1 inhibitor selected from pembrolizumab, nivolumab, atezolizumab, durvalumab, and avelumab.
A further aspect of the present invention provides a method of treating colorectal cancer in a patient, comprising administering to the patient in need of treatment an effective amount of 4,4,4-trifluoro-N-[(1S)[[(7S)(2-hydroxyethyl) oxo-7H-pyrido[2,3-d][3]benzazepinyl]amino]methyloxo-ethyl]butanamide, or a pharmaceutically acceptable salt or hydrate thereof, and an effective amount of a PD-1 or PD-L1 inhibitor selected from pembrolizumab, nivolumab, atezolizumab, durvalumab, and avelumab.
Another aspect of the present invention provides a method of treating T-cell acute lymphoblastic ia, acute lymphoblastic ia, chronic lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, erythroleukemia, triple negative breast cancer, breast cancer, ovarian cancer, melanoma, lung cancer, non small cell lung cancer, pancreatic cancer, glioblastoma, colorectal cancer, head and neck cancer, cervical cancer, prostate cancer, liver cancer, oral squamous cell carcinoma, skin cancer, medulloblastoma, hepatocellular carcinoma, epatic and extrahepatic cholangiocarcinoma, desmoid tumor, soft tissue sarcoma, or adenoid cystic carcinoma in a patient, comprising stering to a patient in need of treatment, simultaneously, tely, or sequentialy, an effective amount of 4,4,4- trifluoro-N-[(1S)[[(7S)(2-hydroxyethyl)oxo-7H-pyrido[2,3-d][3]benzazepin yl]amino]methyloxo-ethyl]butanamide, or a pharmaceutically acceptable salt or hydrate thereof, and an effective amount of a PD-1 or PD-L1 inhibitor selected from lizumab, nivolumab, atezolizumab, durvalumab, and avelumab.
Another aspect of the present invention provides a method of treating colorectal cancer in a patient, comprising administering to a patient in need of treatment, aneously, separately, or sequentialy, an effective amount of 4,4,4- trifluoro-N-[(1S)[[(7S)(2-hydroxyethyl)oxo-7H-pyrido[2,3-d][3]benzazepin yl]amino]methyloxo-ethyl]butanamide, or a pharmaceutically acceptable salt or hydrate thereof, and an effective amount of a PD-1 or PD-L1 inhibitor selected from pembrolizumab, nivolumab, atezolizumab, durvalumab, and avelumab.
A further aspect of the present invention provides a compound 4,4,4-trifluoro-N- [(1S)[[(7S)(2-hydroxyethyl)oxo-7H-pyrido[2,3-d][3]benzazepinyl]amino] methyloxo-ethyl]butanamide, or a pharmaceutically acceptable salt or hydrate thereof; and a PD-1 or PD-L1 inhibitor selected from pembrolizumab, nivolumab, izumab, umab, and avelumab; for simultaneous, separate, or sequential use in the treatment of T-cell acute lymphoblastic leukemia, acute lymphoblastic ia, chronic lymphoblastic leukemia, acute myelogenous leukemia, chronic enous leukemia, erythroleukemia, triple negative breast cancer, breast cancer, ovarian cancer, melanoma, lung cancer, non small cell lung cancer, pancreatic cancer, glioblastoma, colorectal cancer, head and neck cancer, cervical cancer, prostate cancer, liver cancer, oral squamous cell carcinoma, skin , medulloblastoma, cellular carcinoma, intrahepatic and extrahepatic cholangiocarcinoma, d tumor, soft tissue sarcoma, or adenoid cystic carcinoma.
Another aspect of the present invention es a nd 4,4,4-trifluoro-N- 2-[[(7S)(2-hydroxyethyl)oxo-7H-pyrido[2,3-d][3]benzazepinyl]amino] methyloxo-ethyl]butanamide, or a pharmaceutically acceptable salt or hydrate thereof; and a PD-1 or PD-L1 inhibitor selected from pembrolizumab, nivolumab, atezolizumab, durvalumab, and ab; for simultaneous, separate, or sequential use in the treatment of colorectal cancer.
A further aspect of the present invention provides: use of trifluoro-N- 2-[[(7S)(2-hydroxyethyl)oxo-7H-pyrido[2,3-d][3]benzazepinyl]amino] methyloxo-ethyl]butanamide, or a ceutically able salt or hydrate thereof for the manufacture of a ment; and use of a PD-1 or PD-L1 inhibitor selected from pembrolizumab, nivolumab, atezolizumab, durvalumab, and avelumab for the manufacture of a medicament; for the simultaneous, separate, or sequential treatment of T-cell acute lymphoblastic leukemia, acute blastic leukemia, chronic lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, oleukemia, triple negative breast cancer, breast , ovarian cancer, melanoma, lung cancer, non small cell lung cancer, pancreatic cancer, glioblastoma, colorectal cancer, head and neck cancer, cervical cancer, prostate cancer, liver cancer, oral squamous cell carcinoma, skin cancer, medulloblastoma, cellular carcinoma, intrahepatic and extrahepatic cholangiocarcinoma, desmoid tumor, soft tissue sarcoma, or adenoid cystic carcinoma.
A further aspect of the present invention provides: use of 4,4,4-trifluoro-N- [(1S)[[(7S)(2-hydroxyethyl)oxo-7H-pyrido[2,3-d][3]benzazepinyl]amino] methyloxo-ethyl]butanamide, or a pharmaceutically able salt or hydrate thereof for the manufacture of a medicament; and use of a PD-1 or PD-L1 inhibitor selected from pembrolizumab, mab, atezolizumab, durvalumab, and avelumab for the manufacture of a medicament; for the aneous, separate, or sequential treatment of colorectal cancer. r aspect of the present ion is a commercial package comprising a separate composition of each of the therapeutic agents together with instructions for simultaneous, separate or sequential administration for use in ng T-cell acute lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, erythroleukemia, triple negative breast cancer, breast cancer, ovarian cancer, melanoma, lung cancer, non small cell lung cancer, pancreatic cancer, glioblastoma, colorectal cancer, head and neck cancer, cervical cancer, prostate cancer, liver cancer, oral squamous cell carcinoma, skin cancer, medulloblastoma, hepatocellular carcinoma, intrahepatic and extrahepatic cholangiocarcinoma, desmoid tumor, soft tissue sarcoma, or d cystic carcinoma.
A still furth er aspect of the present invention is a commercial package comprising a separate composition of each of the therapeutic agents together with instructions for simultaneous, separate or sequential stration for use in treating colorectal cancer.
The nd trifluoro-N-[(1S)[[(7S)(2-hydroxyethyl)oxo-7H- pyrido[2,3-d][3]benzazepinyl]amino]methyloxo-ethyl]butanamide, or a pharmaceutically acceptable salt or hydrate thereof, (Compound A) has the CAS ry number 1421384. Alternativly, the compound may be named: N-[(1S)[[(7S)- 6,7-dihydro(2-hydroxyethyl)oxo-5H-pyrido[3,2-a][3]benzazepinyl]amino] methyloxoethyl]-4,4,4-trifluorobutanamide. Other names may be used to unambiguously identify Compound A.
The terms, as used herein, “PD-1 inhibitor” and PD-L1 inhibitor” mean a fully human, or humanized IgG, , optionally optimized, monoclonal antibody.
PD-1 inhibitors include nivolumab and pembrolizumab. mab, (Opdivo® ) is also known as iMDX- 1106, 06-04, ONO-4538, or BMS-936558 and has a CAS Registry Number: of 9464144. Nivolumab is a fully human IgG4 monoclonal antibody which specifically blocks PD-1. Nivolumab (clone 5C4) and other human monoclonal antibodies that specifically bind to PD-1 are disclosed in US 8,008,449 and WO2006/121168. Pembrolizumab, (Keytruda®) (formerly lambrolizumab), also known as Merck 3745, MK-3475 or SCH-900475, is a humanized IgG4 monoclonal antibody that binds to PD-1. Pembrolizumab is disclosed in Hamid, O. et al., New England Journal of Medicine, 2013, 369(2): 134-44; WO2009/114335; and US 8,354,509. Other anti- PD-1 antibodies are disclosed in US 089; US 2010028330; and/or US 14649.
PD-Ll inhibitors include YW243.55.S70, MPDL3280A, MEDI-4736, MSB- 8C, and MDX-1105. YW243.55.S70 is an anti-PD-Ll antibody described in WO2010/077634 and US20100203056. MDPL3280A (also known as RG7446, RO5541267, atezolizumab, Tecentriq™) is a fully zed Fc optimized IgG1 monoclonal antibody that binds to PD-L1. MPDL3280A and other human monoclonal antibodies to PD-L1 are disclosed in US 7,943,743 and US 20120039906. MEDI-4736 (also known as durvalumab) is an Fc optimized IgG1 monoclonal antibody to PD-L1 and is described in WO2011/066389. MSB-0010718C (also known as avelumab) is a fully human IgG1 monoclonal antibody to PD-L1 and is described in WO2013/079174. MDX- 1105, also known as BMS-936559, is a fully human IgG4 monoclonal anti-PD-Ll antibody described in WO2007/005874.
As used herein, the term “patient” refers to a mammal, preferably a human. peutically effective amount" or "effective amount" means the dosage of Compound A, or pharmaceutically acceptable salt or e thereof, or pharmaceutical composition containing Compound A, or pharmaceutically acceptable salt or e thereof, and the dosage of a PD-1 or PD-L1 inhibitor, or pharmaceutical composition containing a PD-1 or PD-L1 inhibitor necessary to inhibit tumor cell growth and eliminate or slow or arrest the ssion of the cancer in a t. Dosages of Compound A, or a pharmaceutically acceptable salt or hydrate thereof, are in the range of 2.5 mg/patient to 75 mg/patient once per day every other day over a five day period followed by two days without dosing (T.I.W.). Dosages of a PD-1 or PD-L1 inhibitor, unless otherwise specified on the label, are in the range of 1-3 mg/kg intravenous infusion over 30 to 60 minutes once every 14-21 days. Preferred dosages of Compound A, or a pharmaceutically acceptable salt or hydrate thereof, are in the range of 10 mg to 50 mg T.I.W. The exact dosage required to treat a patient and the length of treatment time will be determined by a physician in view of the stage and severity of the disease as well as the specific needs and response of the individual patient. The dosing stration may be adjusted to provide a more optimal therapeutic benefit to a t and to manage or ameliorate any drug related toxicities. Alternative dosing schedules such as once per day (QD), twice per day (B.I.D.), three times a day (T.I.D.); dosing once per day every other day (Q2D); or every third day (Q3D) may be appropriate for nd A. Dosing administration for PD-1 or PD-L1 inhibitors may be adjusted, including withholding a dose or ently discontinuing r dosing to manage or ameliorate drug related toxicities.
A combination therapy of the t invention is carried out by administering to a T-cell acute lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, erythroleukemia, triple negative breast cancer, breast , ovarian cancer, melanoma, lung cancer, non small cell lung cancer, pancreatic cancer, astoma, ctal cancer, head and neck cancer, cervical cancer, prostate cancer, liver cancer, oral squamous cell carcinoma, skin cancer, medulloblastoma, hepatocellular carcinoma, intrahepatic and extrahepatic giocarcinoma, desmoid tumor, soft ntissue sarcoma, and adenoid cystic carcinoma preferably a soft tissue sarcoma patient ing treatment, an effective amount of nd A, or a pharmaceutically acceptable salt or hydrate thereof, once per day every other day over five days and two days without dosing each week (7-days) over a 28 day cycle and a PD-1 or PD-L1 inhibitor at 1-3 mg/kg over 30-60 minutes once every 14 -21 days.
The terms "treatment," "treat," and "treating," are meant to include the full spectrum of intervention for the cancer from which the patient is ing, such as administration of Compound A and A PD-1 or PD-L1 tor to alleviate, slow, stop, or reverse one or more of the symptoms and to delay, stop, or reverse progression of the cancer even if the cancer is not actually eliminated.
Compound A or a pharmaceutically acceptable salt or hydrate thereof, is preferably formulated as a pharmaceutical composition using a pharmaceutically acceptable carrier and administered by a variety of routes. Preferably, such compositions are for oral administration. A PD-1 or PD-L1 inhibitor is preferably formulated as a pharmaceutical composition using a pharmaceutically able carrier and administered by a parenteral route, preferably intravenous infusion. ably , such compositions may be a lyophilized powder or a liquid composition. Reconstitution or dilution to ready for administration dosages are according to label or by routine skill in the art. Such pharmaceutical compositions and processes for preparing them are well known in the art.
See, for example, HANDBOOK OF PHARMACEUTICAL EXCIPIENTS, 5th edition, Rowe et al., Eds., Pharmaceutical Press ; and REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY (Troy, et al., Eds., 21st n, cott Williams & Wilkins (2006).
Compound A is capable of reaction with a number of inorganic and organic rions to form pharmaceutically acceptable salts. Such pharmaceutically acceptable salts and common methodology for preparing them are well known in the art.
See, for example, P. Stahl, et al., HANDBOOK OF PHARMACEUTICAL SALTS: PROPERTIES, SELECTION AND USE, Wiley-VCH, 2002); S.M. Berge, et al., “Pharmaceutical Salts, “ Journal of Pharmaceutical es, Vol. 66, No. 1, January 1977.
The efficacy of the ation treatment of the invention can be measured by various endpoints commonly used in evaluating cancer treatments, ing but not limited to, tumor regression, tumor weight or size shrinkage, time to progression, overall survival, progression free survival, l response rate, duration of response, tion of metatstatic spread without tumor regression, and PET/CT imaging.
The terms “combination,” “therapeutic combination” and “pharmaceutical combination” refer to a non-fixed dose combination, optionally packaged together with instructions for combined stration where the individual therapeutic agents, nd A, or a pharmaceutically acceptable salt or hydrate thereof, and a PD-1 or PD-L1 inhibitor may be administered ndently at the same time or separately within time intervals that allow the therapeutic agents to exert a cooperative effect.
The term “simultaneous” administration means the administration of each of Compound A and a PD-1 or PD-L1 inhibitor to a patient in a single action such as where each of Compound A and a PD-1 or PD-L1 inhibitor are administered independently at ntially the same time or separately within time intervals that allow Compounds A and a PD-1 or PD-L1 inhibitor to show a cooperative therapeutic effect.
The term “separate” administration means the administration of each of Compound A and a PD-1 or PD-L1 inhibitor to a patient from xed dose dosage forms simultaneously, substantially concurrently, or sequentially in any order. There may, or may not, be a specified time interval for administration of each Compound A and a PD-1 or PD-L1 inhibitor.
The term “sequential” administration means the administration of each of Compound A and a PD-1 or PD-L1 inhibitor to a patient from non-fixed (separate) dosage forms in separate actions. The two administration actions may, or may not, be linked by a specified time interval. For example, administering Compound A T.I.W. and administering a PD-1 or PD-L1 inhibitor over a specified time such as once every 14 to 21 days..
The phrase “in combination with” includes the simultaneous, te, and sequential administration of each of nd A and a PD-1 or PD-L1 tor to a cancer patient in need of treatment, particularly a colorectal cancer patient.
The term “co-administration” or “combined administration” encompasses the stration of the therapeutic agents to a single patient, and include treatment regimens in which the agents may be administered by different routes of administration or at different times.
The beneficial action of two therapeutic agents producing an effect in a single patient which is greater than the simple additive effects of each agent administered alone may be calculated, for example, using le s known in the art such as the Sigmoid-Emax equation (Holford and Scheiner, Clin. Pharmacokinet., 1981, 6: 429-453), the equation of Loewe additivity (Loewe and Muischenk, Arch. Exp. . Pharmacol., 1926, 114: 313-326), the median-effect equation (Chou and Talalay, Adv. Enzyme Regul., 1984, 22: 27-55), and the Bliss Independence method, or known equivalents. Each equation may be applied to experimental data to generate a corresponding graph to aid in assessing the effects of a drug combination as additive, within a biologically relevant range of additive, less than ve, or greater than additive.
The oncogenic role of Notch was first reported in human T-cell leukemia involving a translocation of the Notch1 intracellular domain to the T-cell receptor-β promoter region, resulting in the over expression of Notch1 intracellular domain (Grabher et al. Nature Review , ):347-359; Weng et al. Science, 06):269-271).
Over expression of Notch1 intracellular domain in hematopoietic progenitor cells of mice caused the mice to exhibit T-cell acute lymphoblastic leukemia similar to humans. In addition to T-cell acute lymphoblastic leukemia, there is increasing evidence that Notch signals are oncogenic in other cancers through multiple isms including receptor amplification and over expression of ligands and/or receptors including acute lymphoblastic leukemia, chronic lymphoblastic leukemia (Rosati et al, Blood, 2009(113): 856-865), acute myelogenous leukemia (Sliwa et al. Int J Clin Exp Pathol, 2014(7(3)): 882-889), chronic enous leukemia (Nakahara et al. Blood, 2010(115(14)): 2872- 2881), and erythroleukemia (Robert-Moreno et al, Leukemia, 2007(21): 503).
Aberrant constitutive Notch signaling due to mutation or over expression of ligands and/or receptors is also implicated in a number of solid tumor malignancies ing triple negative breast cancer (Stoeck et al, Cancer Discovery, 2014(4): 1154-1167), breast , ovarian cancer (Park et al. Cancer Research, 2006(66):6312-6318), melanoma (Gast et al. Genes, somes & Cancer, 2010(49):733-745), lung cancer, non small cell lung cancer (Westhoff et al. PNAS, 2009(106):22293-22298), pancreatic cancer, glioblastoma, colorectal cancer, head and neck cancer, cervical , prostate cancer, liver cancer, squamous cell carcinoma (oral), skin cancer and medulloblastoma (Rangathan et al., Nature Review Cancer, 2011(11):338-351 and Supplementary ation S1 (table)). nt constitutive Notch signaling due to mutation or over expression of ligands and/or receptors is also ated in angiosarcoma (Ravi et al, J Clin Oncol, 2007, S, June 20 Supplement)): Abstract 10030), rhabdomyosarcoma (Belyea et al, Clin Cancer Res, 2011(17(23)): 336; Roma et al, Clin Cancer Res, 2011(17(3)): 505-513), rcoma (J Clin Oncol, 2009, (27(15S, Supplement)): Abstract 10526), malignant fibrous histiocytoma (Wang et al, Cancer Res, 2012, (72): 1013-1022), hepatocellular carcinoma (Villanueva et al, Gastroenterology, 2012, (143): 1660-1669), intrahepatic and extrahepatic cholangiocarcinoma (Wu et al, Int J Exp Pathol, 2014, : 3272-3279; Sekiya et al, J Clin Invest, 2012, (122(11)): 3914-3918; Yoon et al, World J Gastroenterol, 2011, )): 4023-4030), and adenoid cystic carcinoma (Bell et al, Annals of Diagnostic Pathology, 2014, (18): 10-13; Stoeck et al, Cancer Discov, 2014, (4): 1154-1167).
The nature of cancer is multifactorial. Under appropriate circumstances, therapeutic agents with different mechanisms of action may be combined. However, only considering a combination of therapeutic agents having different modes of action does not necessarily lead to combinations with advantageous effects. Specific therapeutic agents affording demonstrated beneficial effects (therapeutic effect such as enhanced efficacy and/or lower toxicity) compared with monotherapy of only one of the therapeutic agents is preferred.
The combination of the present invention is believed suitable for the treatment of T-cell acute lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphoblastic leukemia, acute myelogenous ia, chronic enous leukemia, erythroleukemia, triple negative breast cancer, breast cancer, ovarian cancer, melanoma, lung cancer, non small cell lung cancer, pancreatic cancer, glioblastoma, colorectal cancer, head and neck cancer, cervical , prostate cancer, liver cancer, oral squamous cell carcinoma, skin cancer, medulloblastoma, hepatocellular carcinoma, intrahepatic and epatic cholangiocarcinoma, desmoid tumor, soft ntissue sarcoma, and adenoid cystic carcinoma, and particularly le for the treatment of soft tissue sarcoma patients, who have failed standard therapy. This includes patients having cancer showing resistance to monotherapy or showing resistance to combinations different from those of the present invention.
The terms “Complete Response” (CR), al Response” (PR), “Progressive Disease” (PD), “Stable Disease” (SD), “Objective Response” (OR) are used with definitions ing to RECIST v1.1, auer et al., European Journal of Cancer, 2009, 45, 228-247.
The term “time to disease progression” (TTP) refers to the time, generally measured in weeks or months, from the time of initial treatment, until the cancer progresses (see RECIST v1.1 tion for progressive di sease) which is at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on . In addition to the relative increase of 20%, the sum must also demonstrate an te increase of at least 5 mm. The appearance of one or more new lesions is also considered progression. Such progression is evaluated by a skilled clinician.
The term “extending TTP” refers to increasing the time to disease progression in a d patient relative to i) an untreated patient, or ii) a patient treated with less than both of Compound A and a PD-1 or PD-L1 inhibitor.
The term “survival” refers to the patient remaining alive, and includes overall survival as well as progression free survival.
The term, “overall survival” refers to the patient remaining alive for a defined period of time, such as 1 year, 5 years, etc. from the time of sis or treatment.
The term, ession free survival” refers to the patient remaining alive, without the cancer progressing.
As used herein, the term “extending survival” is meant increasing overall or progression free survival in a treated patient ve to i) an untreated t, ii) a patient treated with less than both of Compound A and a PD-1 or PD-L1 inhibitor, or iii) a control treatment protocol. Survival is monitored for a defined period of time, such as one month, six months, 1 year, 5 years, or 10 years, etc., ing the initiation of treatment or following the initial diagnosis of cancer.
The term “primary tumor” or “primary lesion” is meant the original cancer and not a metastatic tumor or lesion located in r tissue, organ, or location in the patient’s body.
In one embodiment, the dose of Compound A is escalated until the Maximum Tolerated Dosage is reached, and a PD-1 or PD-L1 inhibitor of the present invention is administered with a fixed dose. Alternatively, Compound A may be administered in a fixed dose and the dose of a PD-1 or PD-L1 inhibitor may be escalated. Each t may receive doses of nd A and/or a PD-1 or PD-L1 inhibitor either daily or intermittently. The efficacy of the treatment may be determined in such studies, e.g., after 12, 18 or 24 weeks by evaluation of symptom scores every 6 weeks.
Compound A may be prepared by the procedures described in WO 2013/016081.
A PD-1 or PD-L1 tor may be prepared by the procedures described in US 8,008,449 and WO2006/121168; Hamid, O. et al., New England l of Medicine, 2013, 369 (2): 134-44; WO2009/114335, and US 8,354,509; US 8,609,089, US 2010028330, and/or US 20120114649; WO2010/077634; 203056; US 7,943,743; US 20120039906; WO2011/066389; WO2013/079174; and WO2007/005874; or by ures well known and routinely used by one skilled in the art.
The following Examples illustrate the activity of each of Compound A alone, a PD-1 inhibitor alone, or a PD-L1 inhibitor alone, and the combination of Compound A and a PD-1 or PD-L1 inhibitor.
Biological Example 1 In-vivo study: For in-vivo studies 1 x 106 CT26 cells (ATCC® CRL2639TM) a ctal cancer cell line, in 0.2 mL Hank’s Balanced Salt Solution (HBSS) is implanted by subcutaneous injection in the hind leg of 6-8 weeks of age BALB/C female mice (Harlan Laboratories).
Mice are fed ad libitum on normal chow. Treatment is initiated on day 6 of tumor implantation with oral administration (gavage) of Compound A in 1% Sodium carboxymethyl cellulose (Na-CMC) in 0.25% Tween® 80, or intraperitoneal injection of mouse anti PD-L1 antibody (10F.9G2, BioXcell gue #: BE0101 ) in phosphate buffered saline (PBS) or intraperitoneal injection of mouse anti PD-1 (CD279) dy (Clone: RMP1-14, BioXCell#: BP0146-R) in PBS or their respective vehicle in 0.2 mL volume. Compound A is administered at 8 mg/kg on a Monday, Wednesday and Friday schedule for 2 weeks and 10F.9G2 and RMP1-14 are administered at 250 e/animal on Monday and Thursday schedule for 2 weeks.
Tumor growth and body weight are monitored over time to evaluate efficacy and signs of toxicity. Bidimensional measurements of tumors are performed twice a week and tumor s are calculated based on the ing formula: (Tumor Volume) = [(L) x (W2) x (Π/6)] where L is mid-axis length and W is mid-axis width. Tumor volume data are transformed to a log scale to equalize variance across time and treatment groups.
The log volume data are analyzed with a y repeated measures analysis of variance by time and treatment using the MIXED™ procedures in SAS™ software (version 8.2).
The correlation model for the repeated measures is spatial power. Least squares means from the repeated measures is, anti-logged to the tumor volume scale, are shown in Table 1. P-values for ing each pair of groups on study day 20 are shown in Table 2. Test Groups are: 1: 1% CMC/0.25% Tween® 80/0.05% Antifoam, -Wednesday-Friday x2, PO / PBS Monday-Thursday x2, IP 2: Compound A , 8 mg/kg, Monday-Wednesday-Friday x2, PO 3: Compound B (PD-L1), 250 e, Monday-Thursday x2, IP 4: Compound C (PD-1), 250 µg/dose, Monday-Thursday x2, IP : Compound A, 8 mg/kg, Monday-Wednesday-Friday x2, PO / nd B (PD-L1), 250 µg/dose, Monday-Thursday x2, IP 6: Compound A, 8 mg/kg, Monday-Wednesday-Friday x2, PO / Compound C (PD-1), 250 µg/dose, Monday-Thursday x2, IP Tumor growth and body weight are monitored over time to evaluate efficacy and signs of toxicity. Bidimensional ements of tumors are performed twice a week and tumor s are calculated based on the following a: (Tumor Volume) = [(L) x (W2) x (Π/6)] where L is mid-axis length and W is mid-axis width. Tumor volume data are transformed to a log scale to equalize variance across time and treatment groups.
The log volume data are analyzed with a two-way repeated measures analysis of variance by time and treatment using the MIXED™ procedures in SAS™ software (version 8.2).
The correlation model for the repeated measures is spatial power. Treated groups are compared to the control group at each time point. The MIXED™ procedure is also used tely for each treatment group to calculate adjusted means and standard errors at each time point. Both analyses account for the autocorrelation within each animal and the loss of data that occurs when animals with large tumors are d from the study early.
The adjusted means and standard errors are plotted for each treatment group versus time. mor activity is expressed as a tumor volume percentage for ent versus control (% T/C) and is calculated by comparing tumor volume in the treatment group with vehicle treatment group. Percentage T/C and statistical significance value (p value) for the treatment groups is measured essentially as described above and summarized in Table Table 1. Tumor volume (mm3): Geometric Mean Study Days Group 8 10 14 17 20 Vehicle 52.92 99.25 486.99 1045.42 1643.20 Compound A 47.96 88.80 376.50 751.13 1227.40 Compound B 49.82 96.48 378.49 790.67 1313.36 nd C 45.30 87.73 241.50 491.72 622.56 Compound A+B 42.79 88.58 221.73 321.19 472.83 Compound A+C 47.88 102.62 245.30 501.45 569.20 Table 2.
Tumor volume all pairs comparison p value 2 3 4 5 6 Group Compound Compound Compound Compound Compound A B C A+B A+C Vehicle 0.26 0.362 <0.001 <0.001 <0.001 Compound A 0.828 0.009 <0.001 0.001 Compound B 0.005 <0.001 <0.001 Compound C 0.036 0.455 Compound A+B 0.17 Table 2 shows the combination of Compound A and PD-L1 (Group 5), in this test, demonstrated statistically significant tumor growth inhibition s over each of nd A (Group 2) and PD-L1 alone (Group 3). The combination of Compound A and PD-1 (Group 6) demonstrated statistically significant growth tion results over Compound A (Group 2) alone, but not over PD-1 (Group 4) alone. ation analysis Using the repeated measures analysis previously described, a contrast statement is used to test for an interaction effect on study day 20, using the two specific treatments (Compound A and PD-L1) that were combined. This test is statistically significant with p = 0.008, trating better than ve, or synergistic activity, since the estimated mean tumor volume in the combination group (298 mm3) is less than the expected additive tumor volume per the Bliss Independence method (1134 x
NZ788180A 2016-05-20 2017-05-16 Combination therapy with notch and pd-1 or pd-l1 inhibitors NZ788180A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US62/339,363 2016-05-20

Publications (1)

Publication Number Publication Date
NZ788180A true NZ788180A (en) 2022-05-27

Family

ID=

Similar Documents

Publication Publication Date Title
RU2757373C2 (en) Combination therapy with antitumor alkaloid
ES2904880T3 (en) Combination therapy with Notch and PD-1 or PD-L1 inhibitors
US11298362B2 (en) Combination therapy with Notch and CDK4/6 inhibitors for the treatment of cancer
JP2012500180A5 (en)
JP7278331B2 (en) Combination therapy with a Notch inhibitor and a PI3K/mTOR inhibitor for use in treating cancer
JP2014132009A5 (en)
CN104997808A (en) Method and compositions for treatment of cancer
JP2023542093A (en) Use of thiauranib in combination with immune checkpoint inhibitors in antitumor therapy
TW201922282A (en) Combination use of PD-1 antibody and epigenetic modulating agent in the preparation of a medicament for the treatment of tumor
TW201127384A (en) Therapeutic combination comprising a Cdc7 inhibitor and an antineoplastic agent
NZ788180A (en) Combination therapy with notch and pd-1 or pd-l1 inhibitors
RU2806635C2 (en) Combination therapy including an acylthiourea compound and abiraterone
RU2778887C2 (en) Combination of trifluridine/tipiracil hydrochloride, antitumor platinum complex, and modulator of control points of immune response
KR20240046527A (en) Novel oral pharmaceutical composition for cancer treatment
JP2006527753A (en) Composition comprising ZD6126 in combination with 5-FU, CPT-11 or 5-FU and CPT-11 having vascular damage effects for the treatment of colorectal cancer and the like
NZ786609A (en) Combination therapy with notch and pi3k/mtor inhibitors for use in treating cancer
NZ786604A (en) Combination therapy with notch and cdk4/6 inhibitors for the treatment of
EP1599210A1 (en) A combined therapy comprising an indolopyrrolocarbazole derivative and another antitumor agent
TW201315461A (en) Antitumour combination comprising ombrabulin and cetuximab, associated with radiotherapy