NZ734946B2 - Combination treatment protocol - Google Patents

Combination treatment protocol Download PDF

Info

Publication number
NZ734946B2
NZ734946B2 NZ734946A NZ73494616A NZ734946B2 NZ 734946 B2 NZ734946 B2 NZ 734946B2 NZ 734946 A NZ734946 A NZ 734946A NZ 73494616 A NZ73494616 A NZ 73494616A NZ 734946 B2 NZ734946 B2 NZ 734946B2
Authority
NZ
New Zealand
Prior art keywords
cll
ibrutinib
compound
dose
cells
Prior art date
Application number
NZ734946A
Other versions
NZ734946A (en
Inventor
Darcy Bates
Alan R Eastman
Gabriel Kremmidiotis
Original Assignee
Bionomics Limited
Filing date
Publication date
Application filed by Bionomics Limited filed Critical Bionomics Limited
Priority claimed from PCT/AU2016/050135 external-priority patent/WO2016138559A1/en
Publication of NZ734946A publication Critical patent/NZ734946A/en
Publication of NZ734946B2 publication Critical patent/NZ734946B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/665Phosphorus compounds having oxygen as a ring hetero atom, e.g. fosfomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Abstract

The present disclosure provides a combination of a compound of formula (I) or its disodium phosphate ester prodrug with ibrutinib and a specific mode of administration and a method for treating chronic lymphocytic leukemia (CLL).

Description

_ 1 _ COMBINATION TREATMENT PROTOCOL BACKGROUND FIELD The present disclosure teaches a combination therapy for chronic lymphocytic leukemia (CLL).
DESCRIPTION OF RELATED ART Bibliographic details ofthe publications referred to by author in this speci?cation are collected alphabetically at the end of the description.
Reference to any prior art in this speci?cation is not, and should not be take as, acknowledgement or any form of suggestion that this prior art forms part of the common general knowledge in any country.
The reference in this cation to any prior ation (or ation derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived'from it) or known matter forms part ofthe common general knowledge in the ?eld of endeavour to which this speci?cation relates.
Cancer is typically treated with surgiCal, chemical and/or radiation ablation therapy. Whilst chemical and radiation ablation therapy is often ive to destroy a signi?cant amount oftumor cells, such therapies often leave behind a number oftumor cells that are ant to the treatment. These ant cells can proliferate and/or asize to form new tumors that are or have the potential to become recalcitrant to treatment.
Furthermore, the continuous use of chemotherapeutic drugs has given rise to drug resistant tumor cells. Even when combinations of drugs are employed, multidrug resistant (MDR) tumor cells can arise.
The American Cancer Society estimates there will be more" than 15,000 new cases and more than 4,500 deaths from chronic lymphocytic leukemia (CLL) in 2013 alone. sful use of purine analogue-containing immunotherapy regimes extended survival ofyounger patients with CLL. However, eventual progression to ?udarabine-resistant disease and lack of low—risk curative strategies warrant ation of novel treatment strategies.
CLL is characterized by the lation of mature CD5+CD19+CD23+ B lymphocytes in peripheral blood, bone marrow, lymph nodes and spleen, which is thought to be caused by a defect in the y to regulated cell death rather than an uncontrolled ism of cell proliferation. Such a defect can lead to chemoresistance and thus strategies are needed to lead to more potent therapeutics. The B-cell lymphoma/leukemia 2 ) protein is over-expressed in CLL and, therefore, represents a target in attempts to overcome the resistance oftumors to anti-cancer ents. CLL is a debilitating leukemia and, hence, there is an urgent need for selective treatments for this disease.
Introduction ofthe inhibitors ofBCR-associated kinases has provided a great deal promise'in targeted therapies in CLL. Ibrutinib, an inhibitor of BTK, resulted in an overall response rate of~71% in a Phase Ib/II multicenter study in patients with relapsed/refractory CLL, a able single drug activity. Complete remission was, however, rare (2.4%), and daily administration of the drug is typically required to maintain treatment ef?cacy.
Monotherapy with BCR-targeting agents (including ibrutinib) led to the development of peripheral CLL cell lymphocytosis, which persisted for >12 months in 20% ofpatients. This may be a direct consequence of BCR inhibition-mediated egress of the neoplastic cells from their niche. Interestingly, in patients who received ibrutinib intermittently, the CLL cells were able to re—populate the lymph nodes during the off-time. Furthermore, reports of ibrutinib resistance due to ons in the drug—binding cysteine residue in BTK have ly emerged. Other mechanisms of resistance may account for reduced ef?cacy of the BCR- targeting . For example, in vitro data suggest that lation ofa PI3K isoform might rescue lymphoma cells from idelalisib, a PI3K-speci?c inhibitor. Thus, there is seen to be an increase in resistance to BCR-targeting agents, tence ofresidual disease and the ability of CLL cells to re—populate their niche Accordingly, there is a need for a more ef?cacious and selective treatment ofCLL.
SUMMARY The present invention is predicated on the identi?cation of CLL effective combination treatments which involve a compound offormula (I) and a compound that drives CLL cells from the lymph node or bone marrow.
In an embodiment the effective ent for CLL involves the use of a combination of, in either order or simultaneously, a compound which induces CLL cell egress from lymph node or bone marrow, or a pharmaceutically acceptable salt, solvate, stereoisomer or prodrug thereof and a nd of Formula (I) or a pharmaceutically acceptable salt, e, or prodrug thereof. In an embodiment the ive treatment for CLL involves the use of a combination of, in either order or aneously, ibrutinib or a pharmaceutically acceptable salt, solvate, stereoisomer or prodrug thereof and a compound ofFormula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof. In another embodiment the effective treatment for CLL involves the use of a combination of, in either order or aneously, idelalisib or a pharmaceutically acceptable salt, solvate, stereoisomer or prodrug thereofand a compound ofFormula (I) or a ceutically acceptable salt, solvate or prodrug thereof. In an embodiment the combination is useful in the treatment of patients with relapsed or refractory CLL or a CLL which is or has the ial of becoming recalcitrant to treatment.
As used herein "ibrutini " refers to the compound of structure: also known as PCI—32765 (Pharmacyclics) and ed under the name Imbruvica. Its atic (or IUPAC) name is l- [(3R)-3 -[4—Amino(4-phenoxyphenyl)-lH—pyrazolo[3 ,4- d]pyrimidin— l -yl]piperidin— l -yl]prop—2-en— 1 -one and includes its pharmaceutically able salt, e, stereoisomer and prodrug forms.
As used herein "idelalisib" refers to the compound of the structure: also known as Zydelig, GS-l 101 or CAL-101. Its systematic (or IUACC) name is 5-?uorophenyl-2 [(l S)- l -7H-purinylamino)propy1]-4(3H)—quinazolinone; and includes its pharmaceutically acceptable salt, solvate, stereoisomer and prodrug forms.
The compound ofFormula (I) is represented below: H3CO O H3CO 0 ()II (I).
The compound ofFormula (I) [2-Methy1—7—hydroxy(3,4,5—t1imethoxybenzoyl)—6— methoxybenzofuran] can be prepared by the synthetic methodology bed in (WO 07/087684), the contents ofwhich are incorporated by reference, and reference to the a (1) compound includes its pharrnaceutically acceptable salt, solvate and prodrug forms.
Ibrutinib and idelalisib inhibit the pro-survival BCR ing ofCLL cells in the stromal niche resulting in their egress to the periphery. antly, if administration of ibrutinib or idelalisib is stopped, the CLL cells rapidly return to the lymph node. In some ts, the drug—induced increase in circulating CLL cells has been seen for more than a year ing the fact that the cells do not y die once they exit the lymph node. Resistance to ibrutinib has been observed as mutations in the drug—binding cysteine in its target, BTK.
Without wishing to be bound by theory, this resistance is likely to become far more prevalent as patients remain on ibrutinib for months or years. The present invention is predicated, in part, on the determination that certain CLL approved drugs which induce egress from lymph node orbone marrow Will have far greater ef?cacy when they are combined with compounds ofFormula (I) that kill the CLL cells in peripheral circulation, thereby preventing them from returning to the protective lymph node niche. Compounds of a (I) work through an ly different mechanism, i.e. tipping the balance ofpro—survival and optotic BCL2 family member proteins toward the latter, resulting in cell death. This pathway of apoptosis occurs at all stages of the cell cycle which is important considering that the majority of peripheral CLL cells are non—cycling (in Go). The cells which leave the stromal niche following ibrutinib therapy will be susceptible to compounds of a (1) due to lack of additional pro—survival signals which emanate from stromal support.
Other compounds or drugs which induce egress 0fCLL cells from lymph node or bone marrow include: BTK inhibitors such as Acalabrutinib, ONO-4059, and spebrutinib (AVL—292, CC—292), or phosphoinositide 3—kinase inhibitors such as Perifosine, BKM120, Duvelisib, (IPI-l45), , BAY 80—6946, BEZ235, RP6530, TGR 1202, SF1126, INK1117, GDC-0941, XL147 (also known as 408), XL765 (also known as 409), Palomid 529, GSK1059615, ZSTK474, PWT33597, IC87114, TGlOO—llS, CAL263, RP6503PI—103, GNE—477, CUDC—907, and AEZS-l 36, orBCL—2 inhibitors such as venetoclax (ABT-199), ABT—737, or ABT—263, or CDK—inhibitors such as dinaciclib (SH— 727965).
It is ed herein that, in either order or aneously the nd of Formula (I) induces selected and preferential apoptosis of CLL cells via the JNK apoptotic pathway in combination with activating NOXA. It is for this proposed reason that only some microtubule drugs are effective in the ent of certain leukemias. In accordance with the instant disclosure the compounds of Formula (I) are found effective t CLL cells facilitating their apoptosis.
Hence, enabled herein is a method of treating chronic lymphocytic leukemia (CLL) in a patient including the step of administering effective amounts of, at least two compounds, in either order or simultaneously, a compound which induces egress ofCLL cells from lymph node or bone marrow or a pharmaceutically acceptable salt, solvate, isomer or prodrug thereof, and a compound of Formula (I): H3CO O H3CO 0 OH (I). or a pharmaceutically acceptable salt, solvate or g thereof;.
In an embodiment the compound which induces egress ofCLL cells from lymph node or bone marrow is ibrutinib or idelalisib. In an embodiment, the subject or patient is a human. In another embodiment, CLL is relapsed or refractory CLL. This may also be referred to as chronic, persistent or drug resistant CLL or a CLL recalcitrant to treatment.
In one aspect the present invention is predicated on the ing strategy for effectively treating patients with CLL is adopted. Patients are administered a compound which induces egress of CLL cells from lymph node or bone marrow, driving the cells from the lymph node niche. Then a nd of Formula (I) is administered to kill the cells before they can return to the lymph nodes. In another aspect, the patient is given a compound of Formula (I) ?rst followed by a compound which induces egress ofCLL cells from lymph node or bone , such as ibrutinib. In yet another , both compounds are simultaneously administered.
Further taught herein is the use ofibrutinib or a pharmaceutically acceptable salt, solvate, stereoisomer or prodrug thereof and a compound of a (I): H3CO O H3CO 0 0H (1), or a pharmaceutically able salt, solvate or prodrug thereof in the manufacture of a medicament for treating a patient with chronic lyrnphocytic ia (CLL) ing relapsed or refractory CLL. The ment is intended to be used in a protocol to manage CLL therapy in a t, the protocol comprising the combination of ibrutinib and a compound of Formula (1), in either order or simultaneously. In a further embodiment the medicament is a pharmaceutical composition comprising ibrutinib or a pharmaceutically acceptable salt, solvate, stereoisomer or prodrug thereof and a compound ofFormula (I) or a salt, solvate or prodrug thereof.
The "pharmaceutical composition" may be a single composition or a combination composition ofseparate, distinct therapeutics maintained in a therapeutic kit or administered as part of a therapeutic protocol.
In a related embodiment, the present speci?cation is instructive on a compound which induces egress of CLL cells from lymph node or bone marrow or a pharmaceutically acceptable salt, solvate, stereoisomer or prodrug thereof in combination with a nd of Formula (I): H3CO O O \ CH3 H3CO 0 OH (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof; for use in treating CLL in a The present invention further provides a kit for the treatment ofCLL comprising: (a) a compound which induces egress of CLL cells from lymph node or bone marrow or a pharmaceutically acceptable salt, solvate, stereoisomer or prodrug thereof; (b) an amount of a compound of Formula (I): H3CO O 0 (I) H3CO 0 or a pharmaceutically acceptable salt, solvate or g thereof; and (c) instructions for use of (a) and (b) in combination.
The ctions include use ofthe compound which induces egress ofCLL cells from lymph node or bone marrow and Formula (1) in a therapeutic protocol to treat or manage CLL in a human subject. Either compound may be stered ?rst or both be simultaneously administered. In an embodiment a compound which induces egress ofCLL cells from lymph node or bone marrow is ?rst administered.
In relation to the above embodiments, in a further embodiment the nd which induces egress of CLL cells from lymph node or bone marrow is ibrutinib.
In an embodiment, the instructions are directed speci?cally for treating relapsed or ‘ refractory CLL in a human subject.
Without intending to be bound to any particular theory or mode of , incubation with nds of Formula (I) activates the JNK tic pathway and upregulates functional Noxa in CLL cells at concentrations that cause cleavage ofPARP and chromatin condensation. The Formula (1) compounds activity results in acute sis of CLL cells. The effect ofthe compound ofFormula (I) is achieved with an unexpectedly lower concentration than with other microtubule targeting drugs (i.e., increased potency). When incubated with CLL cells, vinblastine and combretastatin A4 show similar effects, but the compounds of Formula (I) are a more potent inducer of CLL activated pJNK and Noxa enabling greater levels of selective apoptosis ofCLL cells. Furthermore, only a 1h incubation is suf?cient to activate JNK, and apoptosis is still observed 5h after removal of a compound of Formula (I).
The instant speci?cation s that apoptosis is dependent on the tion of INK. t limiting the present invention to any one theory or mode ofaction it is proposed herein that both Noxa and JNK are ed for this acute apoptosis to occur in CLL cells.
JNK is also activated in normal lymphocytes but in the absence ofNoxa, were resistant to the compound of Formula (I).
Accordingly, in another embodiment the method es initially treating a subject in need thereof with an ive amount of compound of Formula (I) in order to induce INK—dependent apoptosis in CLL cells.
In an alternative embodiment, the method involves initially treating a subject in need thereof with an effective amount a compound which induces egress of CLL cells from lymph node or bone marrow, such as ibrutinib followed by a compound of Formula (I) in order to induce JNK-dependent apoptosis in CLL cells.
CLL cells are much more resistant to drugs when incubated with stroma cells that mimic the lymph node nment. Therefore, al drug combinations are tested in an effort to circumvent this resistance. However the extent to which such combinations result in synergistic ef?cacy is limited. The compound of the present invention is able to induce apoptosis as a single agent through a ism that primarily involves BCL—2 and MCL-l inhibition (Figure 10). CLL cells grown on stroma are ant to ABT-199 (a BCL—Z inhibitor), but are sensitized by compounds of the present invention. This sensitization is likely due to induction of Noxa. However, it is determined herein that the incubation with stroma cells also upregulates BCL-X which elicits resistance to Noxa induction. Since Noxa can also bind to BCL—X when present in excess over the binding capacity of MCL—l, this allows the combination to overcome the stroma-mediated chemoresistance ofCLL cells.
Higher potency is a desired characteristic ofa new drug because obviously a lower amount of drug is needed to assert an effect, but it can be detrimental if it is anied by higher toxicity or offtarget effects. The compounds ofFormula (I) ofthe present ion do not have any toxic effects as a single agent in peripheral normal lymphocytes even when used at high concentration, comparable to those achievable in plasma. Activation ofINK but no PARP cleavage or Noxa induction is observed. The ability to act in synergy with ibrutinib s much greater y in a treatment ofCLL in ts.
Reference to "CLL" includes its subtypes and its related forms including relapsed or tory forms of CLL and other forms recalcitrant to treatment.
Reference to relapsed or refractory CLL refers to CLL which does not respond to single agent y and is encompassed under chronic and drug resistant CLL. This is also sometimes referred to as "relapse" meaning the return of the disease after some time in patients who were categorised as being in complete or partial remission.
BRIEF DESCRIPTION OF THE FIGURES Figure 1: Compounds of Formula (I) induce apoptosis in peripheral CLL cells. A) Western blots of CLL cells from patient ted for 6h with 0-1 ,uM compounds ofFormula (I). B) Survival curve ofthe same CLL cells measured by chromatin condensation with Hoechst stain.
WO 2016138559 2016/050135 Figure 2: Compounds ula (I) are the morepotent apoptosis inducer in CLL cells.
A) Western blots ofCLL cells from patient 49 incubated for 6h with 0-1 uM compounds ofFormula (I), vinblastine or combretastatin A4. "C" = a control cell line incubated with 2 ,uM vinblastine as a positive l for protein expression. B) Survival curve of the same CLL cells measured by chromatin condensation with Hoechst stain. C) Comparison of the survival curves of CLL cells from 3-6 patients incubated for 6h with a compound of Formula (I), stine or combretastatin A4 (Mean +/- SEM).
Figure 3: Compounds ofFormula (1) -induced apoptosis in CLL cells is JNK dependent A) n blots ofCLL cells from patient 15 incubated for 6h with O — 1 uM compounds of Formula (I) (IO-IOOOnM). "C"= a control cell line incubated with 2 MM vinblastine as a positive l for protein expression. B) Parallel incubations were performed but in the presence ofINK inhibitor VIII. C) Survival curve of the same CLL cells measured by chromatin condensation.
Figure 4: Compounds ofFormula (I) induce apoptosis after 1hpulse incubation in CLL cells. A) Left: Western blots of CLL cells from patient 66 incubated for 6h with 0-1 uM compounds of Formula (I) (1-1000nM). Right: The same cells were incubated with compounds of Formula (I) for 1h, then in the absence of media for an additional 5h. B) Survival curve ofthe same CLL cells measured by chromatin condensation assay with t stain.
Figure 5: Kinetics of compounds of Formula (.0 -induced apoptosis in CLL cells.
Western blots ofCLL cells from t 1 14 ted with 20nM compounds of Formula (1).
Figure 6: Kinetics of compounds of Formula (1) induced apoptosis in Jeko-I cells.
Western blots ofJeko-l cells incubated with 20 nM compounds ofFormula (I).
"C"=a control cell line incubated with 2 ,uM vinblastine as a positive control _ 12 _ for protein expression.
Figure 7: Compounds of Formula (I) sensitize peripheral CLL cells to ABT-]99 incubated on stroma. A) Survival curves of the CLL cells from patient 66 incubated with Compounds ofFormula (I) alone, or in combination with ABT- 199. B) The CLL cells were incubated for 24h on L4.5 l cells, then incubated with nds ofFormula (I) alone or in combination with ABT- 199 for 6h. C) Similar to A except performed on cells from patient 125.
Figure 8: Compounds ofFormula (I) show no ty to normalperipheral cytes and does not sensitize the cells to ABT—199. A) Western blots of cells from a healthy volunteer were incubated for 6h with 0-1 MM compounds ofFormula (I) alone or with 1-10 nM ABT-l99, or 100 nM dinaciclib. " "=a control cell line incubated with 2 uM vinblastine as a positive control for protein expression. B) survival curve of the same cells was measured by chromatin condensation assay with Hoechst stain.
Figure 9: Possible mechanism of action of nds of Formula (1) leading to apoptosis. Compounds of Formula (I) bind to the colchicine site in microtubules, disrupting dynamic stability and resulting in tubulin depolymerization. As a result, INK is phosphorylated and Noxa is induced. plNK can activate a phosphorylation cascade causing inhibition of BCL—2.
Noxa binds to MCL-l targeting it for degradation. Transcription inhibition by the CDK inhibitor dinaciclib results in a rapid decrease in levels of MCL—l.
Pro-apoptotic tors (e.g., BIM, BID) and effectors (e.g., BAX, BAK) ct leading to apoptosis. Co-culture with stromal cells causes protection through upregulation of BCL—XL and MCL-l (and potentially BFLl, not . The BH3 mimetic ABT-l99 ts only BCL-Z and will kill peripheral CLL cells, but not those on stroma. An agent that induces Noxa (e.g. compounds of Formula (1)) or inhibits CL-X expression (dinaciclib) can sensitize cells to ABT-l99.
Figure 10: Compounds ofFormula (I) induce ?VK-dependant apoptosis in CLL cells ex vivo. Freshly isolated CLL cells were incubated for 6h ex vivo with EX2 alone or in the presence ofINK Inhibitor VIII. (A) ents the sensitivity of each individual patient sample to EX2 alone. (B) summarizes the s when the same samples were ted with EX2 with or without the JNK Inhibitor VIIII (n=15).
Figure 11: Compounds of Formula (I) enhance apoptosis induced by 9 or dinaciclib. Leukemia cell lines or CLL cells were incubated with EX2 :: dinaciclib (A) or ABT—199 (B) for 6h. Consistent with us vinblastine results, dinaciclib-mediated apoptosis requires JNK but 9-mediated apoptosis does not. (C) Freshly isolated CLL cells were incubated alone and treated immediately, or plated on top of a monolayer of CD40L expressing L4.5 stroma cells for 24h, then d for 6h with EX2 :: ABT-l99 (n=16).
ED DESCRIPTION Throughout this speci?cation and the claims which follow, unless the context es otherwise, the word ise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion ofa stated integer or step or group of integers or steps but not the exclusion ofany other integer or step or group ofintegers or steps.
As used in the subject speci?cation, the singular forms "a", "an" and "the" include the plural aspects unless the context clearly dictates otherwise. Thus, for example, reference to "a CLL cell" includes a single cell, as well as two or more cells; reference to "an agen " includes a single agent, as well as two or more agents; reference to "the disclosure" includes a single and multiple aspects taught by the disclosure; and so forth. Aspects taught and enabled herein are encompassed by the term "invention". All such aspects are enabled within the width ofthe present invention.
The t disclosure teaches that certain compounds such as ibrutinib act in synergy with a compound ofFormula (I) to effectively inhibit, control or otherwise clinically manage CLL in a patient. An important aspect of the compounds of Formula (I) is the combination of the speci?c C-6 and C-7 substituents together with the C-2 Q-group ially C-2 methyl) which appears to confer greater potency and selectivity when compared to other structurally related TPI compounds. The compounds ofFormula (I) show selectivity towards tumor endothelial cells (activated) over normal endothelial cells (quiescent).
It will be appreciated that ibrutinib may be administered as itself or in a form a pharmaceutically acceptable salt, e, stereoisomer or prodrug thereof. Similarly, a compound ula (I) can be administered to a subject as a pharmaceutically acceptable salt, solvate or prodrug thereof. Suitable pharmaceutically acceptable salts include, but are not limited to salts ofpharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric, ic, and hydrobromic acids, or salts of pharmaceutically acceptable c acids such as , propionic, butyric, tartaric, , hydroxymaleic, fumaric, , citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, acetic, methanesulphonic, toluenesulphonic, benezenesulphonic, salicyclic sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and c acids.
Base salts include, but are not limited to, those formed with pharmaceutically acceptable cations, such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylammonium. In an embodiment, the method bed herein includes within its scope cationic salts e.g. sodium or potassium salts, or alkyl esters (e.g. methyl, ethyl) of the phosphate group.
It Will also be appreciated that any compound that is a prodrug of, for instance, ibrutinib or a compound of Formula (I) is also within the scope and spirit of the therapeutic protocol herein described. The term rug" is used in its broadest sense and encompasses those derivatives that are converted in vivo to a compound of the invention (for instance, ibrutinib or a compound ofFormula (1)). Such tives would readily occur to those skilled in the art, and include, for example, in relation to a (1), compounds where the free hydroxy group (for instance at C-7 position or R1D) is converted into an ester, such as an acetate or phosphate ester, or where a free amino group (for instance at C-7 position or R11) is ted into an amide (e.g., noacid amide). Procedures for esterifying, e. g. acylating, the compounds are well known in the art and may include treatment ofthe compound with an riate carboxylic acid, anhydride or de in the presence of a suitable catalyst or base. One prodrug is a disodium phosphate ester. The disodium ate ester (e.g., a C-7 disodium phosphate ester of a compound of formula I) of the compound of the present invention may be useful in increasing the solubility of the compounds. This would, for instance, may allow for delivery of the compound in a benign vehicle like saline. The disodium phosphate ester may be prepared in accordance with the methodology described in Pettit, et al, (1995) Anticancer Drug Des., @2299. Other texts which generally describe prodrugs (and the preparation thereof) include: Bundgaard (1985) Design of Prodmgs, (Elsevier); Wermuth et al. (1996) The Practice of Medicinal try, 31 , Chapter (Academic Press); and Bundgaard La].( 1 99 l ) A Textbook ofDrugDesign andDevelopment, Chapter 5, (Harwood Academic Publishers).
Accordingly, in an embodiment the compound of Formula (I) is a compound represented as: MeO 0 or We ,P\0Na or a pharrnaceutically acceptable salt, solvate or prodrug thereof.
The compounds ofFormula (I) or a pharmaceutically acceptable salt, solvate or g thereof) may be in crystalline form either as the free nd or as a solvate (e.g. hydrate) and it is ed that both forms are within the scope of the t invention.
Methods of solvation are generally known within the art. Similar considerations apply to ibrutinib or its pharmaceutically acceptable salt, solvate, stereoisomer or prodrug.
An "effective amount" is ed to mean that the amount ofa compound which induces egress of CLL cells from lymph node or bone marrow such as ibrutinib or a pharmaceutically acceptable salt, solvate, stereoisomer or prodrug thereof, and a compound of Formula (I), or a salt, solvate or prodrug thereofand when administered to a subject in need of such treatment, is suf?cient to effect ent for CLL. This includes alleviating symptoms of CLL as well as inducing remission, delaying development of CLL and overall effective management ofCLL in a t. Thus, for example, a eutically effective amount is a quantity suf?cient to reduce or alleviate CLL growth and development. An "effective dose" might require split dosing or cyclic dosing over a particular time interval. Hence, for example, ifa particular amount is required to be administered over a 24 to 48 hour period within a cycle tment, this total amount might be delivered over 6 to 12 hourly intervals to reach the desired dosage per cycle. Any ion on split or cyclic dosing is encompassed herein. Split or ittent dosing may involve cycles, for instance, tinib or a compound ofFormula (I) use in a ?rst cycle followed by the combination ofthe other ofibrutinib or a compound of a (I) in a subsequent cycle. A cycle maybe for 7 to 30 days such as 21 days and from 3 to 20 cycles maybe required such as about 6 . However, the number of cycles required will depend on the severity of CLL, age ofthe patient, the overall health status ofthe patient and so on. A physician would be able to assess. Reference to a subject includes a human of any age. By being in need of such ent includes patients suspected of having a high genetic or familial risk ofdeveloping CLL in an imminent time frame or patients with relapsed or refractory CLL or other recalcitrant CLL.
Treatment includes at least partially ing the desired effect, or delaying the onset of, or inhibiting the progression of, or halting or reversing altogether the onset or progression of CLL.
In an embodiment, treatment is assessed by an amelioration ofsymptoms ofCLL.
Clinical studies such as open-label, dose escalation studies in patients with CLL proliferative diseases are contemplated herein to identify ism of ibrutinib and a compound ofFormula (I). The bene?cial and/or synergistic effects canbe determined directly through the results of these studies which are known as such to a person skilled in the art.
These studies are also able to compare the effects of a monotherapy using either ibrutinib or a compound ofFormula (I) alone. In an ment, the dose of combination partner (a) may be escalated until the Maximum Tolerated Dosage (MTD) is d, and agent (b) is administered as a ?xed dose. Alternatively, ation r (a) is stered in a ?xed dose and the dose ofagent (b) is escalated. Each patient may receive doses of agent (a) either daily, ittently or cyclically. The ef?cacy of the treatment can be determined in such studies, e.g., after 6, 12, 18 or 24 weeks by evaluation of symptom scores every 9 weeks. In this embodiment one of r (a) or agent (b) is considered one or both of ibrutinib or a compound ofFormula (I) and the other ofpartner (a) or agent (b) is the other ofibrutinib or a compound ofFormula (I).
The administration of the pharmaceutical combination of the present invention may result not only in abene?cial effect, e.g., an additive or synergistic therapeutic effect, for instance, with regard to alleviating, ng progression of or inhibiting or ameliorating the symptoms ofCLL, or refractory CLL, but also in further surprising bene?cial effects. Such other effects may e fewer adverse side effects, an improved quality oflife or a decreased morbidity, compared with a monotherapy applying only one of the pharmaceutically active ingredients used in the combination of the present invention.
A ?irther bene?t of the instant therapeutic ol is that lower doses of the active ingredients of, for instance, ibrutinib and/or the compound ofFormula (I) can be used.
The dosages of each component (ibrutinib or a compound of a (1)) need not only be smaller but may also be applied less frequently, which may diminish the incidence or severity of side effects.
The ent protocol herein described may further involve selecting a patient for treatment based on certain clinical parameters such as age, level ofprogression ofthe disease and/or other factors. In addition, patients are generally monitored for progression ofCLL after initiation of ent. Hence, after cessation of treatment, additional treatment may be required subsequently dependent on state or level of remission.
The term "administration" relates to the administration of ibrutinib or a pharmaceutically acceptable salt, solvate, stereoisomer or prodrug thereof, together with a compound ofFormula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof, to a single patient. Combination therapy includes treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
Accordingly, combination partners may be administered together, one after the other or separately in one ed unit dosage form or in two te unit dosage forms. The unit dosage form may also be a ?xed combination such as a pharmaceutical composition Which comprises both partners or in separate doses in an intermittent or cyclic manner.
In an embodiment, a therapeutically effective amount of, for instance, ibrutinib may be administered alone or simultaneously or tially with a compound ula (I) and in any order, and the components may be stered separately or as a ?xed combination. For example, the method of treating CLL or relapsed or refractory CLL according to the invention may comprise: (i) administration ofa ?rst combination partner in free or pharmaceutically acceptable salt, solvate, stereoisomer or prodrug form; and (ii) administration of a second combination r in free or pharmaceutically able salt, solvate, stereoisomer or prodrug form, simultaneously or sequentially in any order, in jointly therapeutically effective amounts, generally in synergistically effective s, e.g., in daily or intermittent dosages or in a cyclical regimen corresponding to the amounts described herein. Where a combination partner is ibrutinib, then a form of ibrutinib includes a stereoisomer thereof. The individual ation partners ofthe combination ofthe invention may be administered separately at different times during the course oftherapy or rently in divided or single forms. The term administering also encompasses the use of a pro-drug of a combination partner that converts in vivo to the combination partner as such. The present invention is, therefore, to be understood as embracing all such regimens of simultaneous or alternating treatment and the term "administering" is to be accordingly interpreted.
In an embodiment, the one combination partner for administration is ibrutinib and another combination partner is a compound of Formula (I). In r embodiment, one combination partner is a compound of Formula (I) and the other ation partner is ibrutinib.
As such it Will be iated that a combination ners maybe presented as a "kit ofparts" or a "pharmaceutical kit" for use in the treatment ofCLL. The kit may comprise a e Where the combination partners are supplied separately for co-administration With ctions for use in the particular therapeutic regimen.
The effective dosage may vary depending on the particular compound or pharmaceutical composition employed, the mode of stration and the severity of CLL condition being treated. Thus, the dosage regimen is selected in accordance with a variety of factors ing the route of administration and the renal and hepatic function ofthe patient.
A physician of ry skill can readily determine and prescribe the effective amounts of each component in the combination required to alleviate, counter or arrest the progress of Daily dosages Will, of course, vary depending on a variety of factors, e.g., the compound chosen, the particular type of CLL to be treated and the desired outcome. In general, however, satisfactory results are achieved on administration of a compound of Formula (I) at daily dosage rates of about 0.05 to 20 mg/kg per day, particularly 1 to 20 mg/kg/per day, e.g. 0.4 to 16 mg/kg per day, as a single dose or in divided doses. As indicated above the dosage regimen per particular interval (e. g. 24 to 48 hours) may be split to achieve total dose over that period rather than bolus. The compound may be administered by any conventional route, in particular enterally, e. g., orally, e. g., in the form of tablets, capsules, drink ons or parenterally, e. g., in the form ofinjectable solutions or sions. Suitable unit dosage forms for oral administration comprise from about 0.02 to 50 mg active ingredient, y 0.1 to 30 mg and 2 to 25 mg, 4 to 20 mg, er with one or more pharmaceutically acceptable diluents or carriers therefore. Put in alternative terms the compound ofFormular (I) may be provided in amounts of from 1 to 280mg/m2 per cycle. Ibrutinib may similarly be administered in amounts of about 200 to 800mg per cycle.
An administration regime may include adding a compound of Formula (I) at an assigned dose level by iv on days 1 and 8 (ofan at least 20 day cycle). In this embodiment the compound ofFormula (I) may be dosed at a level ofbetween 1 to 20 mg/m2.
Adminstration ofibrutinib may include oral administration, e.g., orally, e.g., in the form of tablets, es, drink solutions or erally, e.g., in the form of injectable solutions or suspensions. Suitable unit dosage forms for oral administration comprise from about 200 to 800mg daily, for ce, 480mg daily.
In an embodiment, the compound ofFormula (I) is given intravenously on days 1 and 8 at a ?rst dose (approximately 1 to 8mg/m2 (e.g. 8mg/m2) ; cycle 1) followed by cycle 2 on days 8 and 15 at the same dose with 200 to 800mg (e.g. 480mg) daily ibrutinib. Cycle length is imately 20 days, with 6 cycles required. Any number of cycles may be ed, depending on the response by the patient. Further, ibrutinib may be given as a ?rst cycle ed by the compound of Formula (I).
The present invention also relates to pharmaceutical compositions which comprise compositions of ibrutinib and a compound of Formula (I) or salts, solvates, stereoisomers or prodrugs thereof, which for instance, contain, e.g., from about 0.1% to about 99.9% w/w or w/V, including from about 1% to about 50% w/w or w/V, ofboth ibrutinib and a compound of Formula ( 1) .
The composition may contain any suitable carriers, diluents or excipients. These include all conventional solvents, dispersion media, ?llers, solid carriers, gs, antifungal and antibacterial , dermal penetration agents, surfactants, isotonic and tion agents and the like. It will be understood that the compositions of the invention may also include other supplementary physiologically active agents.
The carrier must be pharmaceutically "acceptable" in the sense of being compatible with the other ingredients of the composition and not injurious to the subject.
Compositions include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parental (including subcutaneous, intramuscular, intravenous and intradermal) administration. The compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art ofpharmacy. Such methods include the step of bringing into association the active ient with the carrier which constitutes one or more ory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or ?nely divided solid carriers or both, and then if necessary shaping the product. itions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets or tablets each containing a predetermined amount ofthe active ingredient; as a powder or granules; as a on or a suspension in an s or non-aqueous liquid; or as an oil-in—water liquid emulsion or a water-in—oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a le machine the active ingredient in a free-?owing form such as a powder or granules, optionally mixed with a binder (e.g. inert diluent, preservative disintegrant (e. g. sodium starch glycolate, cross-linked polyvinyl pyrrolidone, linked sodium carboxymethyl cellulose) surface- active or dispersing agent. Moulded tablets may be made by moulding in a suitable machine a mixture ofthe ed compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release ofthe active ient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release pro?le. Tablets may optionallybe provided with an c g, to provide release in parts of the gut other than the stomach.
Compositions suitable for topical administration in the mouth include es comprising the active ingredient in a ed base, usually sucrose and acacia or tragacanth gum; pastilles comprising the active ingredient in an inert basis such as gelatine and glycerin, or sucrose and acacia gum; and mouthwashes comprising the active ingredient in a suitable liquid carrier. itions suitable for topical administration to the skin may comprise the compounds dissolved or ded in any suitable carrier or base and may be in the form of lotions, gel, creams, pastes, ointments and the like. Suitable carriers include mineral oil, propylene glycol, yethylene, polyoxypropylene, emulsifying wax, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. Transdennal patches may also be used to administer the compounds ofthe invention. itions for rectal stration may be ted as a itory with a suitable base comprising, for example, cocoa butter, in, gelatine or polyethylene glycol.
Compositions suitable for vaginal administration may be ted as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
Compositions suitable for parenteral administration include aqueous and non— aqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bactericides and solutes which render the composition isotonic with the blood ofthe intended recipient; and aqueous and non-aqueous sterile suspensions which may e ding agents and thickening agents. The compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) ion requiring only the addition of the e liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets ofthe kind previously described.
In an embodiment unit dosage compositions are those ning a daily dose or unit, daily sub-dose, as herein above described, or an appropriate fraction thereof, ofthe active ingredient.
It should be understood that in addition to the active ingredients particularly mentioned above, the compositions ofthis invention may include other agents conventional in the art having regard to the type of composition in question, for e, those suitable for oral stration may include such further agents as binders, sweeteners, thickeners, ?avouring agents disintegrating agents, coating agents, vatives, lubricants and/or time delay agents. Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine.
Suitable egrating agents include cornstarch, methylcellulose, polyvinylpyrrolidone, xanthan gum, bentonite, alginic acid or agar. Suitable ?avouring agents include peppermint oil, oil ofWintergreen, cherry, orange or raspberry ?avouring. Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten. Suitable preservatives include sodium benzoate, Vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite.
Suitable lubricants include ium te, stearic acid, sodium oleate, sodium chloride or talc. Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
Those skilled in the art will appreciate that the subject invention bed herein is susceptible to variations and modi?cations other than those speci?cally described. It is to be tood that the invention includes all such variations and modi?cations which fall within the spirit and scope. The invention also includes all ofthe steps, features, compositions and compounds referred to or ted in this speci?cation, individually or collectively, and any and all combinations of any two or more of said steps or features.
Certain embodiments ofthe invention will now be bed with reference to the following examples which are ed for the purpose of illustration only and are not intended to limit the scope of the generality hereinbefore described.
Examples Synthetic Protocols Preparation of 2-Brom0—7-acetoxy—3-(3,4,5-trimethoxybenzoyl)methoxybenzofuran.
Step 1: 2-t—Butyldimethylsilyl(t—butyldimethylsilyloxymethylene)—6-methoxy isopropoxybenzofuran (Larock ng).
A suspension of 2—isopropoxy—3-methoxy—5-iodophenol (4.41 mmol), 1-(tert— butyldimethylsilyl)-3—(tert—butyldimethylsilyloxy)propyne (1.5 g, 5.28 mmol), lithium chloride (189 mg, 4.45 mmol) and sodium carbonate (2.34 g, 22.08 mmol) in dry dimethylformamide (5 mL) at 100 °C was deoxygenated 4 times by evacuation and back?lling with nitrogen.
Palladium acetate (135 mg, 0.60 mmol) was added and the reaction vessel was degassed twice with nitrogen. The reaction mixture was then stirred at this ature for 4 hours (tlc) and the solvent was removed by distillation under vacuum. The residue was dissolved in ethyl acetate (75 mL), stirred well, ?ltered and treated with triethylamine (5 mL). The solution was concentrated onto silica gel (10 g) and puri?ed by ?ash chromatography (silica gel, eluent = hexane/diethyl ether/triethylamine; 95:5: 1 %) to afforded the title compound as a yellow oil (1.45 g, 96 %); 1H NMR (300 MHz, CDC13) 5 , 1H, J= 8.45 Hz), 6.88(d, 1H, J= 8.47 Hz), , 2H, CH2), , 1H), 3.88(s, 3H, OMe), 1.36(d, 6H, J= 6.17 Hz), 0.94(s, 9H), 0.92(s, 9H), 0.35(s, 6H), 0.12(s, 6H).
Step 2: 2-t-Butyldimethylsilylformylmethoxy-7—isopropoxybenzofuran To a solution of2-t—butyldimethy1sily1—3—(t—butyldimethylsilyloxymethylene)-6—methoxy—7- isopropoxybenzofuran (2.69 mmol) in methanol (100 mL) was added concentrated hydrochloric acid (200 ,uL) and the reaction was stirred for 30 minutes (monitored by tlc), quenched with triethylamine (2 mL) and the solvent removed by distillation under vacuum.
The residue was ved in romethane (20 mL), washed with water (10 mL), dried over magnesium sulfate, concentrated under vacuum and co-distilled with toluene (20 mL).
The crude product was dissolved in dry dichloromethane (4 mL) and added to a stirred solution of Collin‘s reagent (chromium trioxide (1.01 g), pyridine (1.65 mL) in dry dichloromethane (3 0 mL)). The suspension was stirred for 10 minutes, ?ltered and the residue washed with diethyl ether (20 mL). The ?ltrate was concentrated onto silica (10 g) and puri?ed by ?ash chromatography (silica gel, eluent = hexane/diethyl-ether/triethylamine (90:9: 1) to afford the title compound as a light yellow oil (503 mg, 48%); 1H NMR (300 MHz, CDC13) 5 10.25(s, 1H, CHO), , 1H, J= 8.45 Hz), 6.98(d, 1H, J= 8.46 Hz), 4.65(m, 1H), , 3H, OMe), 1.3 5(d, 6H, J: 6.17 Hz), 0.97(s, 9H), 0.45(s, 6H).
Step 3: 2-t-Butyldimethylsilyl(3,4,5-trimethoxybenzoyl)methoxy-7— isopropoxybenzofuran To a stirred solution of 3,4,5-trimethoxyiodobenzene (3 77 mg, 1.27 mmol) in dry tetrahydrofuran (1 mL) at -78 °C under nitrogen was added n-butyllithium (795 ML, 1.59 mmol, 2M solution in cyclohexane) and the reaction mixture was stirred at this ature for 40 minutes. After this time a solution of 2—t—butyldimethylsilyl-3—formy1methoxy—7- xybenzofuran (1.07 mmol) in dry tetrahydrofuran (1 mL) was added to the reaction dropwise via e pipette. The reaction mixture was stirred at -60 °C for 20 minutes and then allowed to warm to 0°C, stirred for 10 minutes, quenched with saturated ammonium de solution (2 mL) and diluted with ethyl acetate (20 mL). The organic layer was washed with water (10 mL), dried over magnesium sulfate and the solvent was removed under vacuum to give a residue that was co-distilled with toluene. The crude product (908 mg) was dissolved in dry tetrahydrofuran (10 mL) and treated with 2,3-dichloro-5,6-dicyano-l,4- benzoquinone (900 mg, 1.59 mmol) was added. The reaction mixture was stirred at room temperature for 16 hours (monitored by tlc) and then loaded onto silica (10 g) and puri?ed by ?ash chromatography (silica gel, eluent = hexane/diethyl ether/triethylamine, 90:9 : 1) to afford the title compound as a light yellow oil (498 mg, 69%); 1H NMR (300 MHz, CDCl3) 6 7. l4(s, 2H, benzoyl Hs), 6.81(d, 1H, J= 8.64 Hz), 6.77(d, 1H,J= 8.64 Hz) 4.74(m, 1H), , 3H, OMe), 3.86(s, 3H, OMe), 3.78(s, 6H, 2 X OMe), l.39(d, 6H, J= 6.14 Hz), 1.01(s, 9H), 0.26(s, Step 4: 2-(tert-butyldimethylsilyloxy)acetoxy(3,4,5-trimethoxybenzoyl) methoxybenzofuran To a stirred solution of 2-(t—butyldimethylsilyloxy)-7—isopropoxy(3,4,5- trimethoxybenzoyl)—6-methoxy-benzofuran (160 mg, 0.31 mmol) in dryDCM (2 mL) at room temperature under nitrogen was added solid aluminium trichloride (83 mg, 0.62 mmol) and the reaction e was stirred for 15 minutes (monitored by tlc). The reaction was quenched with a saturated solution of ammonium chloride, extracted with romethane and dried over magnesium sulfate. The solvent was removed by distillation and residue was dried by azeotropic removal of water with toluene. The crude product was dissolved in pyridine (2 mL), acetic anhydride (1 mL) was added and reaction e was stirred for 2 hours at room temperature. The solvent was distilled under vacuum and the residue was loaded onto silica gel (1 g) and puri?ed by column chromatography (silica gel, eluent, hexaneIdiethyl-ether; 80:20) (134 mg, 84%); 1H NMR (300 MHz, CDCl3) 6 7. 14(s, 2H, benzoyl Hs), , 1H, J= 8.72 Hz), 6.85(d, 1H, J= 8.72 Hz), 3.93(s, 3H, OMe), 3.86(s, 3H, OMe), 3.80(s, 6H, 2 x OMe), 2.4l(s, 3H), , 9H), 0.25(s, 6H).
Step 5: 2-Bromoacetoxy(3,4,5-trimethoxybenzoyl)methoxybenzofuran To a stirred solution of 2-t-butyldimethylsily1—7-acetoxy—3-(3,4,5-trimethoxybenzoyl) methoxybenzofuran (120 mg, 0.44 mmol) in chloroethane (1 mL) at room temperature under nitrogen was added bromine (12 ul, 0.44 mmol) se and the reaction mixture was stirred at this temperature for 10 minutes. After this time the reaction was quenched with saturated sodium thiosulfate on, ted with ethyl acetate (20 mL), dried over magnesium sulfate and the solvent d by distillation under vacuum. The crude product was puri?ed by silica gel column chromatography (eluent = Hexanezdiethyl ether; 8 :2 - 7:3) to afford the title compound as a colourless crystalline solid (91 mg, 81%); 1H NMR (300 MHz, CDC13) 6 , 1H, J= 8.70 Hz), 7.14(s, 2H, benzoyl-Hs), 6.98(d, 1H, J= 8.75 Hz), 3.94(s, 3H, OMe), 3.89(s, 3H, OMe), 3.86(s, 6H, 2 x OMe), 2.43 (s, 3H); 13C NMR (75 MHz, CDC13) 6 187.95(CO), 167.71, 152.75, 149.54, 147.49, 142.59, 131.92, 131.80, 123.91 , 121.84, 119.89, 117.72, 109.89, 106.92, 60.69, 56.61, 56.00, 20.09.
Example 1 Preparation of 2-Methyl-7—hydroxy—3—(3,4,5-trimethoxybenzoyl)methoxybenzofuran M90 0 Preparation A To a stirred solution of 2-Bromo—7-acetoxy—3-(3,4,5-trimethoxybenzoyl)-6— methoxybenzofuran (20 mg, 0.042 mmol), methyl-boronic acid (40 mg, 0.67 mmol), in 1,4- dioxane (2 mL) at 90°C was added tetrakz’s—triphenylphosphine palladium (1 1 mg, 0.01 mmol) followed by the addition of a solution of sodium bicarbonate (40 mg, 0.48 mmol) in distilled water (0.5 mL). The reaction mixture turned red after 5 minutes. After 2 hours (tlc) the reaction mixture was brought to room temperature and was added saturated um chloride (2 mL) and diluted with dichloromethane (20 mL). The organic layer was separated and washed with water, dried over magnesium sulfate and the solvent was removed by distillation under vacuum. The residue was puri?ed by PTLC (eluent = romethane/Methanol, 1 :1) to give the title nd (acetate cleaved during reaction) as a ?uffy white solid; (3 mg, 19%). - 27 _ Preparation B (Negishi Coupling) To a stirred solution ofzinc-bromide (592 mg, 2.63 mmol) in dry THF(1.5 mL) at 0°C was added the solution ofmethyl lithium (1.6 M solution in l-ether, 2.6 mL, 4.15 mmol) and the reaction mixture was stirred for 2 hours. Solid 2-bromoacetoxy—3-(3,4,5- trimethoxybenzoyl)methoxy-benzofuran (300 mg, 0.63 mmol) was added and the ether was removed under vacuum and to the rest suspension was added dichlorobis(triphenylphosphine)palladium catalyst (21 mg) and tic amount ofcopper (I) iodide. The reaction mixture was stirred at room temperature for 36 hours ored by tlc), quenched with saturated ammonium chloride solution and extracted with dichloromethane (10 mL), dried over magnesium sulfate and solvent distilled under vacuum and the product was puri?ed by silica gel column (eluent = /ethyl acetate; 8:2). The product was llized in methanol (106 mg, 46%); 1H NMR (300 MHz, CDC13) 6 7.09(s, 2H, benzoyl Hs), 6.93(d, 1H,J= 8.54 Hz), 6.83(d, 1H, J: 8.56 Hz), 5.70(bs, 1H, OH), , 3H, OMe), 3.92(s, 3H, OMe), 3.83(s, 6H, 2 x OMe), 2.54(s, 3H, 2-Me) Example 2 Preparation ofDisodium 6-methoxy—Z-methyl(3,4,5-trimethoxybenzoyl)benzofuran yl phosphate MeO O 0‘ ,ONa /,P\0Na Step 1: Dibenzyl 6-methoxy-Z-methyl(3,4,5-trimethoxybenzoyl)benzofuran—7—yl phosphate: To a mixture of 0.081 g (0.22 mmol) of (7-hydroxy—6-methoxy—2-methylbenzofuran—3- yl)(3,4,5-trimethoxyphenyl)methanone, 0.086 g (0.261 mmol) of carbon tetrabromide and 0.063 ml (0.283 mmol) of dibenzylphosphite in 2.5 ml of ous acetonitrile 0.046 ml of anhydrous triethylamine was added dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temperature, then diluted to 20 ml with ethyl acetate, washed with water brine, dried over anhydrous magnesium sulfate, ?ltered offand evaporated to dryness under d pressure. The residue was puri?ed by ?ash column chromatography (dichloromethane/ ethyl acetate, 9:1) to give the title nd as a colorless foam (0.13 g, 94%); 1H NMR ) 6 2.42 (s, 3H, Me-2); 3.83 (s, 1H, OMe); 3.93 (s, 3H, OMe); 5.33 (m, 4H, CHzPh); 6.89 ((1, CH aromatic, J= 8.7 Hz); 7.21 (dd, 1H, CH aromatic, J= 8.72 HZ; J = 1.2 Hz); 7.08 (s, 2H, CH aromatic); 7.29 — 7.43 (m, 10 H, CH aromatic).
Step 2: um 6-methoxy-Z-methyl(3,4,5-trimethoxybenzoyl)benzofuran-7—yl phosphate: To a d solution of0. 122 g (0.193 mmol) ofthe product from Step 1 in 1 ml ofanhydrous acetonitrile 0.075 ml (0.58 mmol) ofbromotrimethylsilane was added at —5°Cunder nitrogen atmosphere. The resulting mixture was stirred for 1 h at 0°C, then evaporated to dryness in vacuo. The e was diluted to 5 ml with anhydrous methanol and pH ofthe on was brought up about 10 by the addition ofsodium methoxide. After evaporation ofthe resulting mixture under reduced pressure the solid residue was washed with anhydrous isopropanol (4 x 1.5 ml) and anhydrous ethanol (3 x 1.5 ml) and dried under vacuum to give 0.062 g (65 % yield) of title compound as an colorless solid; 1H NMR (D20) 6 2.37 (s, 3H, Me-2); 3.76 (s, 6H, OMe); 3.79 (s, 3H, OMe); 3.82 (s, 3H, OMe); 4.66 (s, H20); 6,93 (d, 1H, CH aromatic, J = 8.6 Hz); 7.04 (d, 1H, CH aromatic, J: 8.6 Hz); 7.10 (s, 2H, CH aromatic).
Biological data Materials and Methods Reagents Example 2 (EX2) used in these s was obtained from Bionomics Ltd. ABT— 199 was purchased from Active Biochem. Dinaciclib was obtained from the Cancer Therapy Evaluation Program, al Cancer Institute. c—Jun—NH2—terminal kinase (JNK) inhibitor VIII was purchased from Calbiochem. Hoechst 3 3342 was purchased from Molecular Probes.
Vinblastine, combretastatin A and other reagents were purchased from Sigma.
The ing antibodies were used: phospho-c-Jun (Ser-63; 9261), phospho- INK1/2 (9255), INK1/2 (9252), and poly ADP ribose polymerase (PARP; 9542; Cell Signaling); Noxa (OP 180) and actin (EMD Biosciences; JLA20). Secondary dies were purchased ?om BioRad.
Cell Culture CLL cells were obtained from consented patients at the Norris Cotton Cancer Center. Cells were puri?ed by centrifugation in Ficoll-Paque PLUS from 10 mL of blood.
Lymphocytes were plated in RPMI 1640 plus 10% serum at 1 X 106 cells/mL after three washes in PBS + 2 mmol/L EDTA. Cells were either incubated immediately with reagents or after 24h incubation with ent layers of CD154+ stromal cells (L 4.5) at a ratio of 5: 1.
Chromatin Staining Cells were incubated for 10 min with 2 ,ug/mL Hoechst 33342 at 37°C and Visualized with a ?uorescent cope. At least 200 cells were scored for each sample. The percentage of cells with condensed tin was ed.
Immunoblot analysis Cells were lysed in urea sample buffer [4 mol/L urea, 10% B—mercaptoethanol, 6% w/V SDS, 125 mmol/L Tris (pH 6.8), 0.01% w/V bromphenol blue, and protease/phosphatase inhibitor cocktail] and boiled for 5 min. Proteins were subsequently separated by SDS-PAGE (10 or 15% w/V) and transferred to polyvinylidene di?uoride membrane (Millipore). , Membranes were blocked with 5% w/v nonfat milk in TBS and 0.05% w/V Tween 20, and were probed with the appropriate primary antibody overnight. Subsequently, membranes were washed in TBS and 0.05% w/v Tween 20, and then incubated with secondary antibody conjugated to horseradish peroxidase. Proteins were Visualized by enhanced chemiluminescence (Amersham). Actin was used as a loading l in Western blots.
Results Single agent y 0fEX2 in CLL cells To determine whether EX2 induces apoptosis, freshly isolated CLL cells were incubated in media containing 0 — 1 [.LM EX2. Chromatin condensation was scored as a classic marker of sis. Apoptosis was observed following incubation of cells with 10 — 100 nM EX2 and this also correlated with the cleavage ofPARP (Figure 1). Protein lysates were also assessed for both pJNK and NOXA, both ofwhich were increased by the same concentrations ofEX2.
The efficacy of three microtubule ting agents, EX2, vinblastine and combretastatin A4 was the compared, in r detail. Figures 2A and B re?ect one individual patient, while Figure 2C re?ects an average of3-6 patients. EX2 is the more potent inducer of apoptosis in CLL cells, as assessed by both chromatin condensation and PARP cleavage. In each case, pJNK and Noxa expression ated with the appearance of apoptosis, which in the case ofEX2 began to appear at concentrations as low as 5 nM (Figure Apoptosis induced in CLL cells is JNK dependent CLL cells were ted With 0-1 ,uM EX2 in the presence or absence ofthe JNK inhibitor VIII. PARP cleavage is seen in the absence of the inhibitor but is completely prevented by the JNK inhibitor (Figures 3A, B, and 10A, B), and this correlates with the observed cell survival measured by condensed tin staining (Figure 3 C). Phosphorylated JNK is observed in all conditions with and without the tor. These s suggest that the mechanism leading to apoptosis induced by EX2 in CLL cells is dependent on pJNK activity.
The activation of JNK occurs rapidly (in less than one hour). It was then ined whether a 1h pulse treatment with EX2 would be as effective as a continuous incubation with EX2. Five hours after removing EX2, JNK activation and PARP cleavage were still observed albeit slightly less than when the EX2 was incubate with the cells continuously (Figure 4).
It was noted that in many ofthese experiments, PARP cleavage was incomplete at 6h. To determine whether greater apoptosis occurred at later time points, we ted cells for up to 24h with EX2 e 5). Apoptosis increased over this time frame with almost total cleavage of PARP observed by 24h, albeit the example shown appears to be particularly sensitive to EX2 even at 6h. However, in a el experiment using Jeko-l cells, it was found that the majority of apoptosis occurred between 6 and 12h and was complete by 24h (Figure 6). Hence it appears that apoptosis is not cted to any subpopulation but can occur in the entire population of cells.
Stroma mediates resistance to EX2 which can be circumvented by novel drug combinations The experiments above have shown that CLL cells are usually very sensitive to EX2. However, these cells were ed from peripheral blood, and the real m to curing CLL is to be able to kill cells that reside in the lymph node or bone marrow niche. To mimic this niche, we have used 1.4.5 cells that express CD154. Co-incubation ofCLL cells on this stroma for 24h elicits marked resistance to many drugs including the BCL-Z inhibitor ABT-l 99 (Figure 7). These co-cultured CLL cells are also markedly resistant to EX2 with no apoptosis observed at 1 ,uM (Figure 7). However, when ABT-l99 and EX2 were combined, marked apoptosis was again observed. For example, 100 nM ABT-l99 alone induced about % apoptosis, whereas when combined with l aM EX2, >60% apoptosis was observed within 6h. The ation of 1 ,uM EX2 and 1 ,uM ABT-l 99 induced about 80% apoptosis.
This patient’s cells appeared to be more resistant than those ized in Figure 2 which may therefore understate the impact of this combination. In cells from another patient that were more sensitive to EX2 alone a greater sensitization to ABT—l99 was observed.
Normalperipheral lymphocytes are resistant to EX2 To test the potential toxicity ofEX2, normal peripheral lymphocytes were isolated from a healthy eer and incubated with EX2 alone or in combination with ABT—l99.
There was no signi?cant PARP cleavage or chromatin condensation induced by EX2, although pJNK was ted; however, no Noxa was d (Figure 8). ABT—199 appeared to induce slight PARP cleavage but this was not increased by EX2, and no chromatin condensation was observed. This ?gure also shows the impact of combining EX2 with the CDK9 inhibitor dinacilib, which functions in this model primarily by preventing expression of MCL-l. Dinaciclib alone induced some apoptosis in normal leukocytes but this was not increased by EX2.
Figure 11 shows Leukemia cell lines or CLL cells were ted with EX2 i dinaciclib (A) or ABT—199 (B) for 6h. Consistent with previous vinblastine s, dinaciclib- mediated apoptosis requires JNK but ABT-l99-mediated apoptosis does not. (C) Freshly isolated CLL cells were incubated alone and treated immediately, or plated on top of a yer of CD40L expressing L4.5 stroma cells for 24h, then d for 6h with EX2 d: ABT-199 (n=16).
Example 3 Phase 1b of ibrutinib and Example 2 in patients with relapsed or refractory CLL.
A phase lb trial is conducted. Table 1 provides a summary ofthe trial conditions.
A list ofabbreviations used in this Example is provided at the end ofthe e. Example 2 is an example of a compound of Formula (I). The study is also capable of variation such a providing the patient ?rst with ibrutinib followed by exposure to Example 2. Such a variation is to be taken into account during the following discussion of this one, non-limiting, embodiment.
Table 1: Summary oftrial A Phase Ib Study of Example 2 and Ibrutinib in Patients with T‘tl1 e Relapsed/Refractory Chronic Lymphocytic Leukemia Short Title EXAMPLE 2 and ibrutinibin CLL Methodology entional study Study Duration 24 months T0 study the safety and e?icacy ofEXAMPLE 2 in combination with Objectives ibrutinib in ts with CLL Number of Subjects Up to 27patients Diagnosis and Main Patients with CLL Inclusion Criteria. _ Study product — e 2, route — intravenous; Study Product, dose level I .' 8 mg/m2 on days I and 8 (cycle I)followed by Example 2 Dose, Route, on days 8 and 15 in combination with ibrutinib 420 mg daily Regimen beginning with cycle 2; cycle length is 21 days Duration of 6 cycles admlnistration. . .
Statistical An open label, dose escalationprospective drug combination study Methodology Table 2 es the study schema.
Table 2: Study schema Example 2 START 8 mg/m IV on days 1, 8 (dose level 1) 12 mg/m2 on days 1, 8 (dose level 2) 16 mg/m2 on days 1, 8 (dose level 3) 4 mg/m2 on days 1, 8 (dose level -1) 2 mg/m2 on days 1, 8 (dose level -2) See the decision tree below for dose modi?cation in subsequent patients 2-16 mg/m IV on days 8,15 420 mg PO on days (Dose corresponds to cycle 1 dose level in absence of 1—21 DLT’s) Decision tree based on DLT’s with cycles 1-2 Expansion cohort STOP (up to N=15) DLT in 0/3 or DLT in 22/3—6 51/6 subjects subjects Dose level -2 DLT in 22/3-6 2 mg/m2 Dose level -1 subjects Example 2 2 . Expansion cohort at 4mg/m DLT "19/3 or DLT in 23-6 dose level -1 (up to 31/6 subjects subjects N=15) Dose level 1 Example 2 DLT in 2213-6 Expansion cohort at 8 mg/m2 dose level 1 DLT - (up to in 0/3 or subjects Dose leve12 N=15) _1 6 su jects< / b' Example 2 12 mg/m2 DLT in 0/3 or . a _1/6 subjects< $153213 16 mg/m2 DLT in 0/3 or DLT in 22/3-6 31/6 subjects subjects Expansion cohort Expansion cohort at (up to N=15) dose level 2 (up to N=15) _ 35 _ STUDY DESIGN AND OBJECTIVES Study design This is a non—randomized open label Phase 1b dose escalation/?nding study of EXAMPLE 2 in combination with ibrutinib in ts with relapsed/refractory CLL.
The study follows a standard 3+3 Phase I design. Once the maximum tolerated dose (MTD) is determined, an expansion cohort is enrolled. Dose limiting toxicities (DLT) are assessed during treatment cycles 1 and 2 to determine the MTD. ts with relapsed/refractory CLL who have not previously received ibrutinib or an alternative Bruton tyrosine kinase (BTK) inhibitor are accrued into this study. Patients who previously received drugs which inhibit kinases within the B—cell receptor (BCR) signaling e other than BTK (e.g. idelalisib, a PI3K inhibitor) are eligible. At dose level I receive Example 2 8 mg/m2 in combination with ibrutinib (420 mg beginning with , patients cycle 2). If safe, the dose ofExample 2 is escalated to 12 and 16 mg/m2 (dose levels 2 and 3).
By contrast, dose de—escalation ofExample 2 to 4 and 2 mg/m2 (dose levels -] and ~2) occurs ifDLT’s are encountered. Once an MTD is determined an expansion cohort is accrued at that dose level of the combination to allow assessment ofDLT’s during subsequent cycles.
Accrual occurs aneously (see Table 2) and takes place at an ambulatory clinic under medical supervision.
Study objectives Primary: — to ish an MTD ple 2 in combination with ibrutinib, a BTK inhibitor, in patients with CLL Secondary: — to determine ef?cacy of e 2 in combination with ibrutinib in patients with CLL ry/exploratory objectives: - to explore the pharmacodynamic effects of Example 2 in CLL B—cells - to assess whether established biomarkers (chromosomal abnormalities, immunoglobulin heavy chain [IGHV] mutational status, ZAP-70 and CD38 expression; p53 mutational status) predict response to EXAMPLE 2 in combination with ibrutinib in patients with relapsed/refractory CLL.
Study nts Primary The y study endpoint is based on toxicity.
Secondary 1) Efficacy. Patients who te at least three 21 -day cycles of study y (one cycle of EXAMPLE 2 alone and two cycles in combination with ibrutinib) are evaluable for response. a) Overall response rate is be determined based on the proportion of study ipants who achieve CR, CRi, PR or nPR assessed two months after completion of therapy, as per IWCLL 2008 criteria (Hallek et al. (2008) BLOOD: [11:5446-5456). b) Event-free survival (EFS), de?ned as the interval between the date of ?rst study treatment and the date of objective signs of disease recurrence, subsequent anti—leukemic therapy, or death, whichever is ?rst reported. 2) Observe the number ofpatients and number of cycles of treatment completed. 3) Biomarkers — to identify patient populations that are more or less likely to respond to the study regimen through the evaluation of biomarker analyses. _ 37 _ Rationale for dose selection The recommended Phase 2 dose (RP2D) of e 2 is evaluated in subjects with advanced solid tumors. Dosing is at a level of 2.1 mg/mz. The dose of 16 mg/m2 is deemed to be an MTD. In this study, adverse events are seen across all dose groups. The two most common categories of AE's are gastrointestinal disorders a, vomiting, constipation) and general ers (predominantly fatigue). Disorders related to bone marrow suppression are rare. Anemia is reported in one patient at a dose level of 8.4 mg/m2 and in two patients at a dose level of 18.9 mg/mz. Two grade 4 events (myocardial infarction and peripheral neuropathy) occurred in the same subject at a dose level 18.9 mg/mz. Similarly, in a phase II study of e 2 in patients with malignant pleural elioma grade 1-2 gastrointestinal and l disorders are the predominant AE's: in 87% and 50% ofpatients correspondingly. Grade 3 e is observed in 8% ofpatients. Finally, grade l-2 anemia is reported in 33% of study participants.
The following considerations are taken into account when the combination therapy is being investigated in CLL.
First, the median age ofpatients with CLL at diagnosis is 72 years. Patients with CLL present with a median of2 comorbidities at diagnosis and 46% carry at least one major comorbidity Thurmes et al. (2008) Leuk Lymphoma 49:49-56.
Second, bone marrow involvement and cytopenias are ubiquitous in CLL. Hence patients with CLL may experience an increased ncy e 3—4 hematologic ties with treatment as compared with patients with solid tumors who have intact bone marrow.
Third, since tumor cells accumulate in the peripheral blood where they may be particularly susceptible to cytotoxic agents, tumor lysis syndrome (TLS) is a concern in CLL.
Since Example 2 induces rapid apoptosis of CLL cell in vitro it is wise to test lower doses of the drug than the currently proposed MTD, particularly in a setting where ibrutinib may provoke lymphocytosis. If rapid response occurs at lower doses of Example 2 than the previously established MTD, RP2D is d down for patients with CLL ultimately reducing the risk of ABS.
Fourth, importantly for this study, e 2 is shown to brie?y disrupt microtubules in PBMCs at doses of 12.6 and 16 mg/m2 ting that suf?cient plasma concentrations can be achieved that induce the anticipated pro-apoptotic biomarkers (P—JNK and NOXA).
Thus, this study employs 8 mg/m2 iv on days 1 and 8 (cycle 1) or 8 and 15 (cycles 2-6) of a 21—day cycle as a starting dose of Example 2. This dose corresponds to a dose one tier below that suf?cient to induce disruption of microtubules, yet also is lower than the currently established MTD in a Phase I study of Example 2 in patients with solid tumors.
Patients e e 2 for one 21-day cycle and receive ibrutinib at an FDA-approved dose (420 mg po daily) beginning with cycle 2. The study follows a standard 3+3 Phase I design (between 3 and 6 patients are ed at each dose tier). The dose of the drug is escalated to 16 mg/m2, toxicities permitting. If toxicities emerge at dose level 1, the dose of Example 2 is de—escalated to 4 mg/mz. Ibrutinib starting dose remains the same at every dose but is adjusted depending on toxicities. Preliminary assessment ofresponse and . tier (420 mg), pharmacodynamic endpoints of treatment with Example 2 is assessed at several dose levels allowing for more careful selection ofRP2D in CLL.
SELECTION OF PARTICIPANTS ] ility criteria 1. Patients have histologically or ?ow cytometry con?rmed diagnosis of B—cell chronic lymphocytic leukemia/small lymphocytic lymphoma /SLL) according to NCI—WG 1996 guidelines Cheson et al. (1996) Blood, 87:4990-4997.. The malignant B cells must co— express CD5 with CD19 or CD20. Patients who lack CD23 expression on their leukemia cells are examined for (and found not to have) either t(l 1 ;14) or cyclin D1 overexpression, to rule out mantle cell lymphoma. 2. Active disease meeting at least 1 ofthe IWCLL 2008 criteria for requiring treatment (Hallek et al. (2008) Supra): (l) A m of any one of the ing constitutional symptoms: (a) Unintentional weight loss >10% within the us 6 months prior to screening. (b) Extreme fatigue (unable to work or perform usual activities). (c) Fevers of greater than 100.5 F for 2 weeks without evidence of infection. ((1) Night sweats without ce of infection. (2) Evidence ofprogressive marrow failure as manifested by the development of, or worsening of anemia or thrombocytopenia. (3) Massive (i.e., >6 cm below the left costal margin), progressive or symptomatic splenomegaly. (4) Massive nodes or clusters (i.e., > 10 cm in longest diameter) or progressive lymphadenopathy. (5) Progressive lymphocytosis with an increase of>50% over a 2-month period, or an anticipated doubling time of less than 6 months. (6) Autoimmune anemia or ocytopenia that is poorly responsive to corticosteroids. 3. Patients must have ed at least one prior y for CLL. 4. Patients must have ECOG performance status 3.
. Patients must have organ function as de?ned below: — direct bilirubin 3 X institutional ULN (unless due to known Gilbert’s syndrome or compensated hemolysis ly attributable to CLL) — AST or ALT less than 2.5 X institutional ULN - estimated CrCL using the Cockroft—Gault equation 50 mL/min. - platelets 50,000/mm3 independent oftransfusion support with no active bleeding. 6. Women of childbearing potential must have a negative serum B-human nic gonadotropin or urine pregnancy test at ing. 7. All patients ofreproductive potential (heterosexually active men and women) must agree to a use of a barrier method of contraception and a second method of contraception and men must agree not to donate sperm during the study and for 4 weeks after receiving the last dose of study treatment.
Exclusion criteria 1. Prior therapeutic intervention with any of the following: a) ibrutinib or another inhibitor of Bruton ne kinase at any time; b) nitrosoureas or mitomycin C within 6 weeks; c) therapeutic anticancer antibodies ding rituximab) within 4 weeks; (1) radio— or toxin-immunoconjugates within 10 weeks; e) all other chemotherapy, ion therapy within 3 weeks prior to initiation of therapy. 2. uate recovery from adverse events related to prior therapy to grade 3 ding Grade 2 alopecia and neuropathy). 3. Chronic use of corticosteroids in excess of sone 20 mg/day or its equivalent.
Stem cell transplant recipients must have no evidence of active gra?-versus-host e. 4. Use of full dose, therapeutic anti-coagulation with warfarin, unfractionated or low lar weight heparins or other anticoagulants (e.g., direct thrombin inhibitors — dabigatran or anti-Xa agents - rivaroxaban/apixiban). Low dose warfarin for catheter prophylaxis or aspirin $25 mg/day is acceptable . Concomitant use of strong CYP rs or inhibitors including nutraceutical preparations, e.g., St John’s Wort 6. History of prior malignancy except: a) Malignancy treated with curative intent and no known active disease present for 22 years prior to initiation of therapy on current study; b) adequately treated non—melanoma skin cancer or lentigo maligna without evidence of disease; c) adequately treated in situ carcinomas (e. g., cervical, esophageal, etc.) without evidence of e; d) asymptomatic prostate cancer managed with "watch and wait" strategy; e) myelodysplastic syndrome which is clinically well controlled and no evidence of the cytogenetic abnormalities teristic ofmyelodysplasia on the bone marrow at screening. 7. Uncontrolled immune hemolysis or thrombocytopenia (positive direct antiglobulin test in absence ofhemolysis is not an exclusion). 8. Thrombotic events nary embolism; deep venous thrombosis) within 6 month prior to start of therapy 9. Human Immunode?ciency Virus (HIV) antibody positivity or active hepatitis B or C.
Intravenous immunoglobulin (IVIG) can cause a false positive hepatitis B gy. Ifpatients receiving routine IVIG have core antibody or surface antigen positivity without evidence of active Viremia (negative hepatitis B DNA) they may still participate in the study, but should have hepatitis serologies and hepatitis B DNA monitored periodically by the treating physician.
. Class III or Class IV New York Heart Association Congestive Heart Failure or acute coronary me within 8 weeks prior to ClDl. 1 1. Major y (requiring general anesthesia) within 30 days prior to tion oftherapy. 12. ity to swallow and retain an oral medication. Patients with clinically signi?cant l ion of malabsorption, in?ammatory bowel disease, chronic conditions which manifest with ea, refractory nausea, vomiting or any other condition that interferes signi?cantly with the absorption of study drugs are excluded. 13. Any condition for which participation in the study is judged by the Investigator to be detrimental to the patient with inter—current illness including, but not limited to an uncontrolled active infection; unstable angina pectoris; uncontrolled cardiac arrhythmia or psychiatric/social ions that would jeopardize compliance with study requirements.
TREATMENT PLAN Treatment is administered on either inpatient or outpatient basis. Expected toxicities and potential risks as well as dose modi?cations for Example 2 and ibrutinib are described below (Expected Toxicities and Dosing Delays/Modi?cations). No investigational or commercial agents or therapies other than those described below are administered with the intent to treat the participant’s CLL/SLL.
Study ures The study consists of a Pre~treatment Period with ne tumor assessment before Example 2 administration, a Treatment Period with up to six 21 -day cycles and a Post- treatment Period (end-of—treatment visit and post-treatment follow—up visits). Patients receive a total of six cycles of therapy unless treatmentis discontinued for one of the pre—specified reasons.
The timing of study assessments and procedures is presented by study cycle and day and are abbreviated by the following nces: Cycle (C) and Day (D) number, as in C1D1 (Cycle 1 Day 1). C1 D1 is the date of ?rst dose of Example 2. Cycles and days within each week are numbered tially thereafter.
Pretreatment period During the Pretreatment Period, patients are screened and consented for the study.
Evaluations performed as part ofroutine care before informed t are utilized as screening evaluations if done within the de?ned time period.
Patients undergo screening evaluations to determine study eligibility, including medical history, physical examination, hematology and biochemical/metabolic laboratory pro?les, urinalysis, coagulation, pregnancy test, and all ying e assessments. All qualifying screening and eligibility assessments are performed within 30 days before the ?rst dose of study treatment. Tests used for baseline disease assessments are performed within speci?ed time frame of the initial dose of study treatment (CT scans — 30 days, genetic markers enetics and FISH], CD38 and bone marrow biopsy — 6 months, HIV and hepatitis testing — 12 months, ZAP-70 - at any time since diagnosis and IGHV mutational status — at any time since diagnosis, if available).
Treatment period A cycle is de?ned as every 21 days. Example 2 is administered at the doses detailed below for up to 6 , and in combination with ibrutinib in cycles 2 through 6.
Clinic visits are performed every cycle on Day 1. Under certain circumstances Day 1 may be delayed by not more than 3 days or occur r than scheduled by not more than 1 day during cycles 2-6.
Assessment ofadverse events occur on Days 1, 2, 8 and 15 ofcycle 1; Days 1, 8, 9 and 15 of cycle 2; and Days 1, 8 and 15 of subsequent cycles.
Clinical laboratory assessments are collected on D1 of each cycle visit, or 318 hours before those visits, and the test results are available and reviewed before the ?rst dose of Example 2 (ClDl) or ibrutinib (C2-6). Screening assessment tests are considered as ClDl tests ifperformed S72 hours before the ?rst dose of study treatment; otherwise, the required evaluations are repeated within this timeframe. Additional clinical laboratory assessments are collected on C1D1, C1D2; as well as C1D8, ClDlS, C2D8, C2D9 and C2D15.
On ClDl and C2D8 all qualifying patients provide samples for ker analysis.
Patients also undergo CT staging on C4Dl (or g2 hours before C4Dl visit) to assess for disease progression. Ifit is suspected that disease progression has ocCurred prior to beginning of C4, CT ng may be performed during C1-3.
The study treatment period ends on day 21 of the last cycle of study ent.
Patients return to the study site 2 months (i7 days) after the last 21-day cycle of study treatment for an end tment visit. Laboratory and physical examinations as well as an ECG are med. Radiographic assessment is performed 2 months (17 days) after the last 21-day cycle. If a complete response is suspected, a bone marrow biopsy is performed no later than 3 months after the last 21 -day cycle ofstudy treatment. Adverse events that are related to study ent and are ongoing at the time ofthis visit are followed until resolution or until considered irreversible by the Lead PI.
Post ent period Disease assessments are obtained in the post treatment period, following the original schedule or earlier, if clinically indicated. Speci?cally, evaluations are performed 3, 6, 9 and 12 months after the end—of treatment Visit, followed by every 4-6 months thereafter and e laboratory assessments and al examination at every Visit. CT scans are obtained at 6 and 12 months after the end-of—treatment visit (i7 days) and as clinically indicated thereafter. Such evaluations are performed regardless her ts choose to continue ibrutinib treatment or not. Patients who continue ibrutinib therapy participate in EFS analysis.
Formulation, storage and handling of Example 2 [01 1 1] Example 2 is obtained from Bionomics Ltd, Australia. The investigational product is le 2 Solution For Injection’, which is a sterile solution of Example 2 manufactured under t Good Manufacturing Practices (cGMP) and which ns 10 mg/mL of the phosphate prodrug equivalent (un-ionized) dissolved in saline. The investigational product is a clear, colorless to yellow liquid presented in a clear glass vial and is intended to be diluted with commercially available sterile 0.9% saline prior to iv administration. Dilution of the investigational product for use in clinical trials is performed using aseptic techniques. The 10 mg/mL Example 2 Solution For ion drug product is intended to be stored and shipped frozen in order to maximize the shelf-life and quality of the drug. The diluted study drug is stored at controlled room temperature or lower (refrigerated) and can be kept for up to 28 hours before use. Protection from light is not ary. Example 2 Solution For Injection has been shown to be ible with commercially available saline iv administration bags and a range ofin?Jsion set components. Stability trials of Example 2 drug product stored at -20°C have shown acceptable product recovery and purity up to 48 months. [0 1 12] The investigational t is diluted, when required, with commercially available sterile 0.9% w/V saline using aseptic techniques. Time is allocated to thaw the investigational product to ambient temperature prior to any dilution with saline. It is recommended that the dilution of the investigational product with 0.9% w/v saline be performed on the day prior to dosing. _ 45 _ Formulation, storage and handling of ibrutinib lbrutinib is obtained from commercial supply and used according to manufacturers instructions.
Treatment description ent is summarized in Table 3.
Table 3. Treatment description Pre—medications-, Agent Dose Route le Cycle Length Precautions According to dose level - Days 1 and 8 (C1); Example 2 None iv over 10 min see below Days 8 and 15 (C2-6) 21 days 420 mg as per packa eg Daily beginning with (3 weeks) Ibrutinib* None PO insert C2D1 * Ibrutinib is self-administered by the study ipants.
Overall Study Design This is an abel, Phase Ib trial with a dose escalation phase, followed by a MTD dose expansion phase. The primary objective ofthe dose escalation phase is to evaluate the MTD ofExample 2 in combination with ibrutinib in patients with relapsed/refractory CLL.
The MTD dose expansion phase further evaluates the safety and ef?cacy ofthe combination in up to 15 patients at the MTD level.
Dose Escalation Phase Up to three dose levels are evaluated in the ‘dose tion’ phase.
The dose levels ofExample 2 are 8 mg/m2 (Dose Level 1), 12 mg/m2 (Dose Level 2) and 16 mg/m2 (Dose Level 3) IV on days 1 and 8 ofcycle 1 and days 8 and 15 ofcycles 2—6 (Table 4). ' [0116] IfDLTs are observed at dose level 1 (as described below), the dose ofExample 2 is de—escalated to 4 mg/m2 (Dose Level -1). If DLTs are observed at dose level -1 (as bed below), the dose of Example 2 is de-escalated to 2 mg/m2 (Dose Level -2).
Starting dose of Example 2 is 8 mg/m2 IV on days 1 and 8 of cycle 1, when BCN105P is administered as a single agent prior to initiation of ibrutinib. Beginning with cycle 2, ibrutinib is administered concomitantly with Example 2 at a starting dose of420 mg PO daily. To allow ibrutinib-mediated egress of CLL cells from the lymph nodes niche, Example 2 is administered on days 8 and 15 during cycles 2—6. The starting dose ofibrutinib remains unchanged at each dose level.
Each cycle lasts for 21 days. Provided no toxicities occur, each t is treated for 6 cycles.
The ‘dose escalation’ phase ofthe study follows a standard 3+3 Phase I design. At a given dose level, 3 patients are enrolled. If all 3 ts complete the ?rst cycle oftherapy without any imiting toxicities (DLTs), the next cohort of 3 patients is enrolled at the next higher dose level. If 1/3 patients develops a DLT the cohort are expanded to 6 patients. r, if either 2/3 or 2/6 patients in any dose tier have DLTs, the previous dose tier is de?ned as the maximum tolerated dose (MTD) of the combination. Once the MTD is determined, an expansion cohort is accrued to a total of 15 patients at that dose level, i.e. 12 or 9 onal patients are accrued.
Table 4: Dose levelsplannedfor dose escalation phase ofthe study Dose Level Example 2 (Cycles 1-6) Ibrutinib (Cycles 2-6) intravenously on days 1 and 8 of cycle 1 and p0 daily on days 1—21 of 21- days 8 and 15 of cycles 2-6 day cycles Each cycle is 21 days -2 2mm —.1— 4...... —1— my... 2 12mg... —3* Wm Dose Level —1 is studied only ifmore than one patlent develops a DLT 1n Dose Level 1 For all the Dose Levels in the scalation phase, should more than one patient develop a DLT in the respective Dose Level; the dose of Example 2 is reduced according to the plan shown in Table 4.
If no signi?cant toxicities are observed, the dose escalation part of the study is anticipated to enroll between 9 and 18 patients. Treatment continues for either a) 6 cycles; or b) until disease ssion or unacceptable toxicities, if they occur prior to completion of 6 cycles of therapy.
Dose Extension Phase In the ‘dose extension’ phase patients are d at the MTD of Example 2 in combination with ibrutinib, determined in the dose escalation phase. Treatment continues for either a) 6 cycles; or b) until disease progression or ptable toxicities, ifthey occur prior to completion of 6 cycles of therapy. In this phase, patients are assessed for safety (CTCAE v.4.03) and ef?cacy parameters (overall se rate [ORR] and progression free survival [PFS]).
Patients Who either: a) fail to complete the ?rst ef?cacy evaluatiOn (scheduled at the beginning of C4) for any reason or b) receive <2 doses of Example 2 during each of the ?rst 3 cycles or <14 of 21 doses of ibrutinib during each of cycles 2 and 3 are not ered for ef?cacy evaluations, unless disease progression has occurred prior to the ?rst scheduled ef?cacy evaluation.
Pre—treatment criteria C1 D1 Hematologic ters: platelets must be >50,000/mm3 (in absence of usion support); hemoglobin > 8 g/dL (trans?lsion support permissible); Non—hematologic parameters: direct bilirubin 3 X institutional ULN (unless due to known Gilbert’ s syndrome or hemolysis directly attributable to CLL); AST orALT < 2.5 X institutional ULN.
Vital signs, all laboratory data (including pregnancy testing) are reviewed by the treating physician prior to administering the ?rst dose of a study agent.
Subsequent cycles Hematologic parameters: platelets must be >50,000/mm3 or >75% of baseline, whichever is lower (without trans?lsion support); hemoglobin > 8 g/dL (trans?lsion support permissible); ANC>1000/mm3 or >75% of baseline, whichever is lower (G—CSF support permissible at the tion of the investigator in case of ANC<1000) matologic parameters: direct bilirubin 3 X utional ULN s due to known Gilbert’s syndrome or hemolysis directly attributable to CLL); AST orALT < 2.5 X institutional ULN.
Administration of Example 2.
The starting dose of Example 2 for all patients is 8 mg/m2 d IV over 10 minutes on Days 1 and 8 of a 21—day cycle with cycle 1; on Days 8 and 15 ofa 21-day cycle beginning with cycle 2 for a maximum of6 cycles. The dose ofExample 2 is calculated based on the actual body weight using Mosteller or DuB ois formulas for BSA.
The choice of a formula is based on the institutional guidelines.
Mosteller formula: BSA = SQRT ([Height (cm) x Weight (kg)]/3600), DuBois formula: BSA (m2) = 0.20247 x Height(m)°'725 x (kg)0'425.
The dose is ulated with each cycle according to the same formula used with previous cycle.
Recommended duration ofthe infusion: 1 0 minutes; observation period following infusion: 15 minutes.
Administration of ibrutinib Ibrutinib is self-administered ing with C2Dl. Ibrutinib is taken orally, with 8 ounces (approximately 240 mL) ofwater. The capsules are swallowed intact, not less than minutes before or 2 hours after a meal. Doses are taken in the morning at about the same time each day. Ifthe patient misses a dose, it can be taken as soon as possible on the same day with a return to the normal schedule the following day. The patient keeps a diarywhere he/she records the date and time that ibrutinib was taken.
On days when ibrutinib is administered with e 2 (days 8 and 15 ofcycles 2 through 6), ibrutinib is given in the clinic at least 30 minutes prior to administration of Example 2.
Duration of therapy and follow-up for individual patients Study participants receive up to 6 cycles of Example 2 and ibrutinib ng no limiting toxicity .
EXPECTED TOXICITIES AND DOSING /DOSE MODIFICATIONS Anticipated toxicities: Example 2 Based on the data available for Example 2, the following is a list of effects that could be encountered in patients administered Example 2 via the iv route: 0 Gastro—intestinal effects (nausea, vomiting, diarrhea or constipation).
- Hematological changes (myelosuppression, platelet counts, reticulocyte numbers, slight delays in coagulation) ° Drowsiness, fatigue ° ss in arms and legs 0 General malaise ° Rib pain ° Increase in symptoms of infections / infestations (herpes simplex; oral candidiasis) - Increased skin sensitivity 0 Effects on sperm count ° Cardiovascular effects, myocardial infarction and transient blood pressure changes.
- Peripheral sensory athy 0 Elevation in the liver function tests ' non—ST segment elevation myocardial infarction ° Thromboembolic events (including pulmonary sm, deep vein thrombosis) ° Stroke Although Example 2 showed no effects on the cardiovascular system when tested in dogs, potential effects on the vascular system cannot be ruled out. Based on s from the 2-cycle rat study for Example 2, dose-dependent, reversible cardiomyopathy was observed in rats. Signs of cardiomyopathy were decreased in severity and incidence by 14 days after the 2-cycles of treatment, indicating recovery or ibility of the effects. The implications ofthese ?ndings for humans are not clear as these cardiovascular s were not observed in dog toxicity studies. Furthermore, in a vascular safety pharmacology study in telemetered dogs, there were no effects on cardiovascular or atory function following doses up to 0.8 mg/kg (the highest dose assessed). Also, based on in vitro testing, the IC50 values for hERG channel inhibition for both Example 1 and Example 2 are much greater than 2.5 and 486.0 ,ug/mL, respectively, with the ?rst dose representing what was maximally le in the test system, and the latter dose representing a level 30 times the highest free plasma concentration predicted to be present after stration of Example 2 at 100 times the theoretical starting dose in the clinic. Thus an effect on the hERG channel is expected to be slight, if any, at ef?cacious doses of Example 2.
Of the effects listed above, effects on sperm count, hematologic changes, and ial cardiovascular effects (myocardial infarctions; transient blood re changes) could be considered to be risks of potential severity and seriousness. It is worth noting that hematologic effects and effects on sperm count are not cted effects s that inhibit tubulin polymerization and subsequent cell proliferation.
Ef?cacy All subjects who completed two cycles of study treatment are evaluable for ef?cacy.
Disease evaluations Physical examination, which focuses on documenting a change in the number of site and size oflymphadenopathy, hepato— and splenomegaly, is done as part ofthe full disease evaluation during treatment and at months 3 9 and 12 after the end—of-study visit and every 4—6 months thereafter, until disease progression or death.
Complete blood count (CBC) with measurement ofparameters including ALC are obtained on Days 1, 2, 8 and 15 of Cl; Days 1, 8, 9 and 15 of C2; on day l for the remaining four , every 3 months for the ?rst 12 months, and every 4-6 months thereafter until disease progression or death.
Serum biochemistry/metabolic panel are obtained on D1 of each cycle; 3 and 6 hours after administration of Example 2 on C1D1 and C2D8; and on C1D2 and C2D9.
Computed tomography (CT) scan of neck, chest, abdomen and pelvis with intravenous contrast where possible are performed at screening, on C4D1 (or $72 hours prior), at the end of treatment visit (2 monthsi7 days after completion of the last 21-day cycle of treatment), at months 6 and 12 after the end of treatment visit (i7 days), and as ally indicated thereafter. Site measurement is performed according to IWCLL 2008 criteria.
A eral bone marrow aspirate and biopsy is obtained during screening or up to 6 months before the ?rst dose of study drug. Subjects who have bone marrow aspirate and biopsy results since completion oftheir last therapy for CLLmay use those results ifthey were obtained within 6 months prior to the ?rst dose of study drug. If the subject's physical examination ?ndings, tory and radiographic evaluations suggest that CR has been obtained, a bone marrow aspirate/biopsy is obtained to con?rm the CR within 30 days after the end-of-treatment visit.
Criteria for response Modi?ed IWCLL guidelines Hallek et a1. (2008) Blood, m:5446-5456 are used to measure response in CLL/SLL patients.
Objective response for CLL/SLL patients is de?ned as CR, Cri, nPR and PR.
Patients are ed for response at the end of treatment. If there is a clinical suspicion for ssion, disease assessment is performed at any time.
Complete remission (CR) requires all ofthefollowing: 1. Peripheral blood lymphocytes (evaluated by blood and differential count) below 4 X 109/L 2. Absence of signi?cant lymphadenopathy (lymph nodes >1.5 cm in er) by physical examination and imaging, ifbaseline scans were abnormal 3. No hepatomegaly or splenomegaly by physical examination and imaging, ifbaseline scans were al 4. Absence of tutional symptoms (B symptoms) . Blood counts: - Neutrophils > 1.5 X 109/L without need for exogenous growth factors - Platelets > 100 X 109/L t need for exogenous growth factors — Hemoglobin > 11.0 g/dL without red blood cell trans?lsion or need for exogenous opoietin 6. Bone marrow aspirate and biopsy must have the following ?ndings - normocellular for age - less than 30% of nucleated cells being lymphocytes - no B-lymphoid nodules (con?rmed by IHC) Complete response with incomplete marrow recovery (CRi): patients who ful?ll all the criteria for a CR but who have a hypocellular marrow and persistent anemia or thrombocytopenia or penia unrelated to CLL but secondary to drug toxicity. If the marrow is hypocellular, a repeat determination is performed after 4 weeks, or when eral blood counts have recovered.
Nodular partial response (nPR): patients who ?Jl?ll all the criteria for CR but who have bone marrow evidence of B—lymphoid nodules by IHC.
Partial remission (PR) requires: 1. Blood count should show one ofthe following results: - Neutrophils more than 1.5 X 109/L without need for exogenous growth factors - Platelet counts > 100 X 109/L or 50% improvement over baseline t need for exogenous growth factors - Hemoglobin > 1 1.0 g/dL or 50% improvement over baseline without ing red blood cell trans?lsions or exogenous erythropoietin And two of the following three ia: 2. Decrease in number of blood lymphocytes by 50% or more from the value before therapy 3. Reduction in denopathy by al examination or imaging as de?ned by: - A decrease in lymph node size by 50% or more either in the sum products of up to 6 lymph nodes, or in the largest diameter of the enlarged lymph node(s) detected prior to therapy - No increase in any lymph node, and no new enlarged lymph node - In small lymph nodes (<2 cm), an increase of less than 25% is not considered to be signi?cant 4. A reduction in splenomegaly and hepatomegaly by 50% or more, by physical examination or imaging.
PR with lymphocytosis: Since ibrutinib may induce persistent lymphocytosis, it should not interfere at the time of designation of a PR. PR with lymphocytosis should be based on other measurable s of disease other than ALC Cheson er a1. (2012) J Clin Oncol, 30:2820-2822.
CORRELATIVE STUDIES All study participants undergo peripheral blood collection before drug stration and 0.5, 3 and 6 hours after tion of the infusion of the lst dose of Example 2 with cycle 1 (Example 1 alone), as well as with cycle 2 (C2D8 - Example 2 in ation with ibrutinib). 15 mL ofblood is collected at all time points (participant number, and date and time of collection are recorded). Venous blood samples are transported to Dr.
Danilov/Eastman's laboratories within 1 hour after collection. CLL B-cells are isolated using Ficoll-Hypaque gradient. An aliquot ofcells are ?ash frozen immediately a?er puri?cation for subsequent protein analysis.
The sample ofCLL cells ed prior to therapy is incubated with Example 2 ex vivo and analyzed for expression ofP—INK and NOXA similar to that presented in Figure 14.
This provides baseline data re?ecting variability between patient samples, and thereby help to explain any potential variation observed in the samples analyzed ing treatment.
The following pharmacodynamic endpoints and additional biomarkers are Protein analysis from all blood collections are d byimmunobloting for INK activation (phospho-JNK) and NOXA expression.
CLL cells are also analyzed by centrifugation for Example ated dissociation of tubulin.
The remaining CLL B-cells are viably frozen at —70°C until further processing including RNA and DNA isolation.
A preliminary assessment is performed as to whether prognostic biomarkers (IGHV, ZAP-70 expression, CD38 expression and CLL FISH panel) are of value to predict response to e 2/ibrutinib combination in CLL. Such biomarkers (with the exception of IGHV) are routinely obtained during the diagnostic work-up ofCLL to delineate prognosis in an individual patient.
- IGHV mutational status (if not available after e testing) is assessed employing IgH Somatic Hypermutation Assay v.2.0 (InVivoSribe Technologies). - p53 mutational status (direct sequencing, at OHSU) STATISTICAL CONSIDERATIONS This Phase lb study is conducted using the ‘3+3 ’ strategy: in the ?rst stage, up to 6 ts are administered 8 mg/m2 Example 2 in combination with ibrutinib (dose level 1).
This is done in up to two steps. First, up to 3 patients receive the drug. Ifthere is two or more toxicities there is no dose escalation. Ifthere are no toxicities the dose is escalated in another cohort. If there is exactly one ty, up to 3 additional ts are stered the same dose. Ifthere is no toxicities in this second cohort of3 the dose is escalated in another cohort.
If there is at least one more toxicity the dose is not escalated. In this second stage, if 2 (or more) experience toxicity, dose level 1 is considered MTD, following by an expansion cohort at that dose level. If 1 (or less) experience a toxicity at the second stage, then ion cohorts are accrued at that dose level (up to 15 subjects).
If2 (or more) experience toxicity at stage 1, an alternative second cohort ofup to 6 subjects is administered 4 mg/m2 Example 2. If the number of toxicities in this alternative second stage is 2 or more, a third cohort ofup to 6 subjects is administered 2 mg/m2 Example 2. If the number of toxicities in this cohort is 2 or more, Example 2/ibrutinib combination is rejected.
Table 5 below shows the probability of dose tion as a function of the ying toxicity frequency. For instance, if the true ncy of toxicity at a ular dose level is 10% there is a 91% chance that the dose is escalated. Ifthe true frequency is 50% there is only a 17% chance it is escalated.
Table 5: Probability ofdose escalation Underlying Escalation Toxicity Probability (%) Frequency (%) The frequency oftoxicities in the expansion cohorts (N=1 5) is reported along with 95% exact binomial nce intervals. Table 6 below shows the expected limits of these intervals. For instance, ifthe true frequency oftoxicities is 10%, the 95% con?dence interval is expected to range from 1.6% to 34.8%.
Table 6: Interval Limits Actual probability of Expected Limits of 95% Exact Binomial, % toxicity, % .6 57.6 17.1 67.0 24.6 75.4 In a Phase II study ofibrutinib in relapsed/refractory CLL, an overall response rate (ORR) has been reported in 90% ofpatients (including PR with lymphocytosis), however CR was uncommon (Byrd er al. (2013) NEngl JMed, 369:32—42). The e 2 and ibrutinib combination is considered ious and deemed for further evaluation if a CR rate>30% is achieved on an expansion cohort. The frequency of CR is ed along with 95% exact binomial con?dence intervals (assuming that an expansion cohort s ten patients, ifa true CR is 30% the 95% con?dence interval is expected to range from 8.1% to 63.9%). The distribution of EFS are reported using a Kaplan-Meier estimate with con?dence als.
It is expected that the clinical Ib data will show synergy between ibrutinib and Example 2 in the treatment and management of CLL. _ 53 _ List ofAbbreviations AE — adverse events ALC — absolute lymphocyte count ALT — alanine minase ANC — absolute neutrophil count aPTT- activated partial thromboplastin time AST — aspartate aminotransferase BCR - B-cell receptor BTK - Bruton tyrosine kinase CBC — complete blood count CCRC — Clinical Cancer Review Committee CIRS - tive Illness Rating Scale CLL — chronic lymphocytic leukemia CPHS — Committee for the Protection ofHuman Subjects CR — te response CrCL — (estimated) creatinine clearance CRi — te se with lete marrow recovery CT — computed tomography CTO — Clinical Trials Of?ce DHMC — Dartmouth-Hitchcock Medical Center DLT — dose-limiting toxicity DSMAC — Data Safety Monitoring and Accrual Committee eCRF — electronic case report form EFS — event-free survival FISH — ?uorescent in situ hybridization IGHV — immunoglobulin heavy chain gene IHC — immunohistochemistry IRB — Institutional Review Board IV — intravenously IWCLL — International Workshop on Chronic Lymphocytic Leukemia KCI — Knight Cancer ute LDH — lactate dehydrogenase MTD — maximum tolerated dose NCCC — Norris Cotton Cancer Center nPR — nodular l response OHSU - Oregon Health and Science University ORR — overall response rate OS — overall survival PFS — progression-free survival PI — principal investigator PI3K - phosphoinositide-3 kinase PO — by mouth PR _ partial remission SAE — serious e events SLL — small lymphocytic lymphoma tl/z - half-life ULN - upper limit of normal WBC — white blood cells ZAP—70 — zeta chain—associated T—cell receptor protein kinase 70 kDa AE — adverse events ALC — absolute lymphocyte count ALT — alanine transaminase ANC — absolute neutrophil count aPTT— activated partial thromboplastin time AST — aspartate aminotransferase BCR - B-cell receptor BTK - Bruton ne kinase CBC — complete blood count CCRC — Clinical Cancer Review Committee CIRS — Cumulative Illness Rating Scale CLL — chronic lymphocytic leukemia CPHS — Committee for the Protection ofHuman Subjects CR — complete response CrCL — (estimated) creatinine nce CRi — complete response with incomplete marrow recovery CT — computed tomography CTO — Clinical Trials Of?ce DHMC — Dartmouth-Hitchcock Medical Center DLT — dose-limiting ty DSMAC — Data Safety ring and Accrual Committee eCRF — electronic case report form EFS — event—free survival FISH — ?uorescent in situ hybridization IGHV — immunoglobulin heavy chain gene IHC — histochemistry IRB — Institutional Review Board IV — intravenously IWCLL — International Workshop on Chronic Lymphocytic Leukemia KCI - Knight Cancer Institute LDH — lactate dehydrogenase MTD — maximum tolerated dose NCCC — Norris Cotton Cancer Center nPR — nodular partial response OHSU - Oregon Health and Science University ORR — overall response rate OS — l survival PFS — progression—freesurvival PI — principal investigator PI3K - phosphoinositide-3 kinase PO — by mouth PR — partial remission SAE — serious adverse events _ 61 _ SLL — small lymphocytic ma tl/z - half-life ULN - upper limit of normal WBC — White blood cells ZAP-70 — zeta chain—associated T-cell receptor protein kinase 70 kDa BIBLIOGRAPHY 1. Bundgaard (1985) Design ofProdrugs, (ElseVier); 2. Bundgaard et al.(1991) A Textbook ofDrugDesign andDevelopment, Chapter 5, (Harwood Academic Publishers). 3. Byrd et al. (2013) NEngl JMed, 369232-42. 4. Cheson et al. (1996) Blood, 87:4990-4997.
. Cheson et al. (2012), J Clin Oncol, 30:2820-2822. 6. Hallek et al. (2008) Blood, 111:5446-5456. 7. Thurmes et al. (2008) Leuk Lymphoma, 49:49-56. 8. Wermuth et al. (1996) The ce ofMedicz‘nal Chemistry, Chapter 31 (Academic Press); THE

Claims (8)

CLAIMS DEFINING THE INVENTION ARE AS S:
1. Use of an effective amount of a compound of Formula (I): H3CO O OH (I) or a pharmaceutically acceptable salt, or solvate thereof; in the manufacture of a medicament for ng CLL in a human patient, wherein the medicament is to be administered together with ibrutinib or a pharmaceutically acceptable salt, or solvate thereof; wherein ibrutinib is to be administered in an amount of from 200 to 800mg daily, and wherein the effective amount of the nd of Formula (I) is from 1 to 20mg/m2, and wherein one of ibrutinib or a compound of Formula (I) is to be administered in a first cycle; and the other of ibrutinib and a compound of Formula (I) in a second cycle and such respective cycles as necessary for amelioration of symptoms of CLL in the patient.
2. The use of Claim 1, n the human patient has relapsed or refractory CLL.
3. The use of Claim 1, n ive amounts are to be administered in cycles comprising one of ibrutinib or a compound of Formula (I) in a first cycle; and the other of ibrutinib and a compound of Formula (I) in a second cycle and such respective cycles as necessary for amelioration of symptoms of CLL in the patient.
4. The use according to claim 3, wherein the compound of a (I) is to be administered intravenously on days 1 and 8 at a first dose followed by cycle 2 on days 8 and 15 at the same dose with 200 to 800mg daily ibrutinib.
5. The use according to Claim 4, wherein the compound of a (I) is to be administered intravenously on days 1 and 8 at a first dose at 1 to 8mg/m2 ; cycle 1 followed by cycle 2 on days 8 and 15 at the same dose with 200 to 800mg daily nib.
6. The use according to Claim 4, wherein the cycle length is approximately 20 days, with 6 cycles required.
7. The use of Claim 1, wherein the human patient is selected for treatment based on clinical parameters including age, level of progression of the disease, and/or other complicating ailments.
8. The use of a compound which is: in the manufacture of a medicament for treating CLL in a human patient, wherein the medicament is to be administered together with nib or a pharmaceutically acceptable salt, or e thereof; wherein ibrutinib is to be administered in an amount of from 200 to 800mg daily, and wherein the effective amount of the compound is from 1 to 20mg/m2and wherein one of ibrutinib or the compound is to be administered in a first cycle; and the other of ibrutinib and the compound in a second cycle and such respective cycles as necessary for amelioration of symptoms of CLL in the patient. 111111 Coxicsnn‘ati?n (211M) 1F{SURE E -2/M — ’Wh?a?m QMMta?tsm?g «\\\\\' 15X?
NZ734946A 2016-03-02 Combination treatment protocol NZ734946B2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2015900764A AU2015900764A0 (en) 2015-03-05 Combination Treatment Protocol
PCT/AU2016/050135 WO2016138559A1 (en) 2015-03-05 2016-03-02 Combination treatment protocol

Publications (2)

Publication Number Publication Date
NZ734946A NZ734946A (en) 2023-08-25
NZ734946B2 true NZ734946B2 (en) 2023-11-28

Family

ID=

Similar Documents

Publication Publication Date Title
KR101915942B1 (en) Mutant selectivity and combinations of a phosphoinositide 3 kinase inhibitor compound and chemotherapeutic agents for the treatment of cancer
EA030664B1 (en) Treatment of cancer with tor kinase inhibitors
US9364459B2 (en) 3-(indolyl)- or 3-(azaindolyl)- 4-arylmaleimide derivatives for use in the treatment of colon and gastric adenocarcinoma
CA2993605C (en) 1,3-benzodioxole derivatives for the treatment or prevention of adult t cell leukemia/lymphoma
EA028462B1 (en) Methods for treating advanced non-small cell lung cancer using tor kinase inhibitor combination therapy
US20140171433A1 (en) Multiple myeloma treatment
KR20120104196A (en) Biomarkers for predicting the sensitivity and response of protein kinase ck2-mediated diseases to ck2 inhibitors
JP2021524835A (en) AXL Kinase Inhibitors and Their Use
KR20170122787A (en) A combination of a phosphoinositide 3-kinase inhibitor compound and a CDK4 / 6 inhibitor compound for the treatment of cancer
KR20140040728A (en) Methods of treating mesothelioma with a pi3k inhibitor compound
US9987293B2 (en) Method for treating chronic lymphocytic leukemia
JP2010525046A (en) Methods for the treatment of hematological tumors
US11872220B2 (en) Methods and compositions for treating B-cell malignancies
CA2978640C (en) Combination treatment protocol
US8598188B2 (en) Method for predicting therapeutic efficacy of chemotherapy on non-small-cell lung cancer
NZ734946B2 (en) Combination treatment protocol
WO2021100677A1 (en) Combination drug
CA3148858A1 (en) Therapeutic agent for cancer having resistance to anti-ccr4 antibody
JP2022520802A (en) Combination therapy for use in the treatment of cancer
US11478445B2 (en) Method for treating acute myeloid leukemia
WO2024048555A1 (en) Combination drug
US20230321102A1 (en) TREATMENT OF CANCER USING COMBINATION THERAPIES COMPRISING GDC-6036 and GDC-0077
WO2023104809A2 (en) 2-oxothiazole compositions for treatment of t-cell acute lymphoblastic leukaemia
WO2012075484A2 (en) Novel drug targets to overcome de novo drug-resistance in multiple myeloma